1
|
Ramsden CE, Cutler RG, Li X, Keyes GS. HYPOTHESIS: Lipid-protecting disulfide bridges are the missing molecular link between ApoE4 and sporadic Alzheimer's disease in humans. Prostaglandins Leukot Essent Fatty Acids 2025; 205:102681. [PMID: 40209641 DOI: 10.1016/j.plefa.2025.102681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 04/01/2025] [Accepted: 04/02/2025] [Indexed: 04/12/2025]
Abstract
As the principal lipid transporter in the human brain, apolipoprotein E (ApoE) is tasked with transport and protection of highly vulnerable lipids that are required to support and remodel neuronal membranes, in a process that is dependent on ApoE receptors. APOE allele variants that encode proteins differing only in the number of cysteine (Cys)-to-arginine (Arg) exchanges (ApoE2 [2 Cys], ApoE3 [1 Cys], ApoE4 [0 Cys]) comprise the strongest genetic risk factor for sporadic Alzheimer's disease (AD); however, the specific molecular feature(s) and resultant mechanisms that underlie these isoform-dependent effects are unknown. One signature feature of Cys is the capacity to form disulfide (Cys-Cys) bridges, which are required to form disulfide-linked dimers and multimers. Here we propose the overarching hypothesis that super-ability (for ApoE2), intermediate ability (for ApoE3) or inability (for ApoE4) to form lipid-protecting intermolecular disulfide bridges, is the central molecular determinant accounting for the disparate effects of APOE alleles on AD risk and amyloid-β and Tau pathologies in humans. We posit that presence and abundance of Cys in human ApoE3 and ApoE2 respectively, conceal and protect vulnerable lipids transported by ApoE from peroxidation by enabling formation of disulfide-linked homo- and heteromeric ApoE complexes. We thus propose that inability to form intermolecular disulfide bridges makes ApoE4-containing lipoproteins uniquely vulnerable to peroxidation and its downstream consequences. Consistent with our model, we found that brain-enriched polyunsaturated fatty acid-containing phospholipids induce disulfide-dependent dimerization and multimerization of ApoE3 and ApoE2 (but not ApoE4). By contrast, incubation with the peroxidation-resistant lipid DMPC or cholesterol alone had minimal effects on dimerization. These novel concepts and findings are integrated into our unifying model implicating peroxidation of ApoE-containing lipoproteins, with consequent ApoE receptor-ligand disruption, as initiating molecular events that ultimately lead to AD in humans.
Collapse
Affiliation(s)
- Christopher E Ramsden
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH 251 Bayview Blvd., Baltimore, MD 21224, USA; NIH, Intramural Program of the National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD 20892, USA.
| | - Roy G Cutler
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH 251 Bayview Blvd., Baltimore, MD 21224, USA
| | - Xiufeng Li
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH 251 Bayview Blvd., Baltimore, MD 21224, USA
| | - Gregory S Keyes
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH 251 Bayview Blvd., Baltimore, MD 21224, USA
| |
Collapse
|
2
|
Xavier C, Pinto N. Navigating the blurred boundary: Neuropathologic changes versus clinical symptoms in Alzheimer's disease, and its consequences for research in genetics. J Alzheimers Dis 2025; 104:611-626. [PMID: 39956949 DOI: 10.1177/13872877251317543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2025]
Abstract
During decades scientists tried to unveil the genetic architecture of Alzheimer's disease (AD), recurring to increasingly larger sample numbers for genome-wide association studies (GWAS) in hope for higher statistical gains. Here, a retrospective look on the most prominent GWAS was performed, focusing on the quality of the diagnosis associated with the used data and databases. Different methods for AD diagnosis (or absence) carry different levels of accuracy and certainty applied to both subsets of cases and controls. Furthermore, the different phenotypes included in these databases were explored, as several incorporate other ageing comorbidities and might be encompassing many confounding agents as well. Age of the samples' donors and origin populations were also investigated as these could be biasing factors in posterior analyses. A tendency for looser diagnostic methods in more recent GWAS was observed, where greater datasets of individuals are analyzed, which may have been hampering the discovery of associated genetic variants. Specifically for AD, a diagnostic method conveying a clinical outcome may be distinct from the disease neuropathological assessment, since the first has a practical perspective that not necessarily needs a confirmation. Due to its properties and complex diagnosis, this work highlights the importance of the neuropathological confirmation of AD (or its absence) in the subjects considered for research purposes to avoid reaching statistically weak and/or misleading conclusions that may trigger further studies with powerless groundwork.
Collapse
Affiliation(s)
- Catarina Xavier
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Nádia Pinto
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IPATIMUP - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
- CMUP - Centro de Matemática da Universidade do Porto, Porto, Portugal
| |
Collapse
|
3
|
García-González P, Puerta R, Cano A, Olivè C, Marquié M, Valero S, Rosende-Roca M, Alegret M, Sanz P, Brosseron F, Martino-Adami P, de Rojas I, Heneka M, Ramírez A, Navarro A, Sáez ME, Tárraga L, Cavazos JE, Boada M, Fernandez MV, Cabrera-Socorro A, Ruiz A. APOE Haplotype Phasing Using ONT Long-Read Sequencing Reveals Two Common ε3 and ε4 intragenic haplotypes in the Spanish Population. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.25.25324541. [PMID: 40196265 PMCID: PMC11974914 DOI: 10.1101/2025.03.25.25324541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Background The apolipoprotein E (APOE) gene is a key genetic determinant of Alzheimer's disease (AD) risk, with the ε4 allele significantly increasing susceptibility. While the pathogenic effects of the ε4 allele are well established, the functional impact of distinct haplotype configurations within the broader ε3 and ε4 backgrounds remains poorly understood. This study investigates the role of intragenic sub haplotypes in modulating APOE expression and their potential influence on AD progression. Methods We utilized Oxford Nanopore Technology (ONT) long-read sequencing to phase variants within a 4-kilobase comprising the APOE locus in a cohort of 1,265 individuals with known APOE genotypes. We evaluated the impact of the identified intragenic haplotypes on APOE protein levels in cerebrospinal fluid (CSF) using the Olink platform, adjusting for demographic and molecular covariates. Statistical modeling was employed to assess the independent effects of these haplotypes alongside traditional APOE genotypes. Additionally, their influence on dementia progression in mild cognitive impairment (MCI) subjects was analyzed using adjusted Cox proportional hazards models. Results Our analysis identified 48 Single Nucleotide Variants (SNVs) within a 4-kilobase region containing the APOE gene, including nine novel variants. Phasing of variants within the APOE locus revealed 59 unique haplotypes in the Spanish population, which were grouped into five major haplogroups-ε2, ε3A, ε3B, ε4A, and ε4B-including two common haplogroups for each of the ε3 and ε4 isoforms. The ε4A haplogroup was associated with a significant decrease in APOE ε4 protein levels in CSF (p = 0.004), suggesting a regulatory mechanism that may mitigate the toxic gain-of-function effect typically attributed to the ε4 allele. Conversely, the ε3B haplogroup was linked to increased APOE ε3 protein levels in ε3/ε4 carriers (p = 0.025), potentially serving a compensatory role.These effects were independent of overall APOE genotype and remained significant after adjusting for covariates. Both haplogroups (ε4A and ε3B) demonstrated protective effects in the progression from MCI to dementia, underscoring their potential relevance in Alzheimer's disease. Conclusions This study provides new insights into the intragenic allelic variability of the APOE gene, demonstrating that intragenic APOE haplogroups within the ε3 and ε4 backgrounds can modulate APOE isoform expression in ways that might modulate AD. Our findings highlight the importance of considering haplotype-specific effects when interpreting the functional impact of APOE and in designing targeted therapeutic strategies. Further research is needed to explore the broader regulatory network of the APOE locus and its interaction with neighboring loci in the 19q13 region.
Collapse
Affiliation(s)
- Pablo García-González
- ACE Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- PhD program in Biotecnology, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028- Barcelona, Spain
| | - Raquel Puerta
- ACE Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Barcelona, Spain
- PhD program in Biotecnology, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028- Barcelona, Spain
| | - Amanda Cano
- ACE Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Claudia Olivè
- ACE Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Barcelona, Spain
| | - Marta Marquié
- ACE Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Sergi Valero
- ACE Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Maitee Rosende-Roca
- ACE Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Montserrat Alegret
- ACE Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Pilar Sanz
- ACE Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Barcelona, Spain
| | - Frederik Brosseron
- Universitätsklinikum Bonn & Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Bonn, Germany
| | - Pamela Martino-Adami
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Itziar de Rojas
- ACE Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Eschsur-Alzette/Belvaux, Luxembourg
| | - Michael Heneka
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Eschsur-Alzette/Belvaux, Luxembourg
| | - Alfredo Ramírez
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Medical Faculty, 53127 Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
- Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
- Department of Psychiatry and Glenn, Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San An TX USA
| | - Arcadi Navarro
- Institució Catalana de Recerca i Estudis Avançats (ICREA) and Universitat Pompeu Fabra. Pg. Lluís Companys 23, 08010, Barcelona, Spain
- IBE, Institute of Evolutionary Biology (UPF-CSIC), Department of Medicine and Life Sciences, Universitat Pompeu Fabra. PRBB, C. Doctor Aiguader N88, 08003 Barcelona, Spain
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Av. Doctor Aiguader, N88, 08003 Barcelona, Spain
- BarcelonaBeta Brain Research Center, Pasqual Maragall Foundation, C. Wellington 30, 08005, Barcelona, Spain
| | - María Eugenia Sáez
- ACE Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Barcelona, Spain
- CAEBI, Centro Andaluz de Estudios Bioinformáticos, Sevilla, Spain
| | - Lluís Tárraga
- ACE Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - José E. Cavazos
- South Texas Medical Science Training Program, University of Texas Health San Antonio, San Antonio, TX, USA
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, TX, USA
| | - Mercè Boada
- ACE Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | | | | | - Agustín Ruiz
- ACE Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, TX, USA
- Department of Microbiology, Immunology and Molecular Genetics. Long School of Medicine. University of Texas Health Science Center, San Antonio, TX, USA
| |
Collapse
|
4
|
Ramsden CE, Cutler RG, Li X, Keyes GS. Lipid-protecting disulfide bridges are the missing molecular link between ApoE4 and sporadic Alzheimer's disease in humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.17.633633. [PMID: 39868210 PMCID: PMC11761642 DOI: 10.1101/2025.01.17.633633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
As the principal lipid transporter in the human brain, apolipoprotein E (ApoE) is tasked with the transport and protection of highly vulnerable lipids required to support and remodel neuronal membranes, in a process that is dependent on ApoE receptors. Human APOE allele variants that encode proteins differing only in the number of cysteine (Cys)-to-arginine (Arg) exchanges (ApoE2 [2 Cys], ApoE3 [1 Cys], ApoE4 [0 Cys]) comprise the strongest genetic risk factor for sporadic Alzheimer's disease (AD); however, the specific molecular feature(s) and resultant mechanisms that underlie these isoform-dependent effects are unknown. One signature feature of Cys is the capacity to form disulfide (Cys-Cys) bridges, which are required to form disulfide bridge-linked dimers and multimers. Here we propose the overarching hypothesis that the super-ability (for ApoE2), intermediate ability (for ApoE3) or inability (for ApoE4) to form lipid-protecting intermolecular disulfide bridges, is the central molecular determinant accounting for the disparate effects of APOE alleles on AD risk and amyloid-β and Tau pathologies in humans. We posit that presence and abundance of Cys in human ApoE3 and ApoE2 respectively, conceal and protect vulnerable lipids transported by ApoE from peroxidation by enabling formation of ApoE homo-dimers/multimers and heteromeric ApoE complexes such as ApoE-ApoJ and ApoE-ApoD. We thus propose that the inability to form intermolecular disulfide bridges makes ApoE4-containing lipoproteins uniquely vulnerable to peroxidation and its downstream consequences. Consistent with our model, we found that brain-enriched polyunsaturated fatty acid-containing phospholipids induce disulfide-dependent dimerization and multimerization of ApoE3 and ApoE2 (but not ApoE4). By contrast, incubation with the peroxidation-resistant lipid DMPC or cholesterol alone had minimal effects on dimerization. These novel concepts and findings are integrated into our unifying model implicating peroxidation of ApoE-containing lipoproteins, with consequent ApoE receptor-ligand disruption, as the initiating molecular events that ultimately lead to AD in humans.
Collapse
Affiliation(s)
- Christopher E. Ramsden
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH 251 Bayview Blvd., Baltimore, MD, 21224, USA
- Intramural Program of the National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, 20892, USA
| | - Roy G. Cutler
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Xiufeng Li
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Gregory S. Keyes
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH 251 Bayview Blvd., Baltimore, MD, 21224, USA
| |
Collapse
|
5
|
Min JH, Sarlus H, Harris RA. MAD-microbial (origin of) Alzheimer's disease hypothesis: from infection and the antimicrobial response to disruption of key copper-based systems. Front Neurosci 2024; 18:1467333. [PMID: 39416952 PMCID: PMC11480022 DOI: 10.3389/fnins.2024.1467333] [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: 07/19/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
Microbes have been suspected to cause Alzheimer's disease since at least 1908, but this has generally remained unpopular in comparison to the amyloid hypothesis and the dominance of Aβ and Tau. However, evidence has been accumulating to suggest that these earlier theories are but a manifestation of a common cause that can trigger and interact with all the major molecular players recognized in AD. Aβ, Tau and ApoE, in particular appear to be molecules with normal homeostatic functions but also with alternative antimicrobial functions. Their alternative functions confer the non-immune specialized neuron with some innate intracellular defenses that appear to be re-appropriated from their normal functions in times of need. Indeed, signs of infection of the neurons by biofilm-forming microbial colonies, in synergy with herpes viruses, are evident from the clinical and preclinical studies we discuss. Furthermore, we attempt to provide a mechanistic understanding of the AD landscape by discussing the antimicrobial effect of Aβ, Tau and ApoE and Lactoferrin in AD, and a possible mechanistic link with deficiency of vital copper-based systems. In particular, we focus on mitochondrial oxidative respiration via complex 4 and ceruloplasmin for iron homeostasis, and how this is similar and possibly central to neurodegenerative diseases in general. In the case of AD, we provide evidence for the microbial Alzheimer's disease (MAD) theory, namely that AD could in fact be caused by a long-term microbial exposure or even long-term infection of the neurons themselves that results in a costly prolonged antimicrobial response that disrupts copper-based systems that govern neurotransmission, iron homeostasis and respiration. Finally, we discuss potential treatment modalities based on this holistic understanding of AD that incorporates the many separate and seemingly conflicting theories. If the MAD theory is correct, then the reduction of microbial exposure through use of broad antimicrobial and anti-inflammatory treatments could potentially alleviate AD although this requires further clinical investigation.
Collapse
Affiliation(s)
- Jin-Hong Min
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital at Solna, Stockholm, Sweden
| | | | | |
Collapse
|
6
|
Yesiltepe M, Yin T, Tambini M, Bao H, Pan M, d'Abramo C, Giliberto L, Han X, D'Adamio L. Analysis of early effects of human APOE isoforms on Alzheimer's disease and type III hyperlipoproteinemia pathways using knock-in rat models with humanized APP and APOE. Cell Commun Signal 2024; 22:458. [PMID: 39334477 PMCID: PMC11438110 DOI: 10.1186/s12964-024-01832-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024] Open
Abstract
APOE is a major genetic factor in late-onset Alzheimer's disease (LOAD), with APOE4 increasing risk, APOE3 acting as neutral, and APOE2 offering protection. APOE also plays key role in lipid metabolism, affecting both peripheral and central systems, particularly in lipoprotein metabolism in triglyceride and cholesterol regulation. APOE2 is linked to Hyperlipoproteinemia type III (HLP), characterized by mixed hypercholesterolemia and hypertriglyceridemia due to impaired binding to Low-Density Lipoproteins receptors. To explore the impact of human APOE isoforms on LOAD and lipid metabolism, we developed Long-Evans rats with human APOE2, APOE3, or APOE4 in place of rat Apoe. These rats were crossed with those carrying a humanized App allele to express human Aβ, which is more aggregation-prone than rodent Aβ, enabling the study of human APOE-human Aβ interactions. In this study, we focused on 80-day-old adolescent rats to analyze early changes that may be associated with the later development of LOAD. We found that APOE2hAβ rats had the highest levels of APOE in serum and brain, with no significant transcriptional differences among isoforms, suggesting variations in protein translation or stability. Aβ43 levels were significantly higher in male APOE4hAβ rats compared to APOE2hAβ rats. However, no differences in Tau or phosphorylated Tau levels were observed across the APOE isoforms. Neuroinflammation analysis revealed lower levels of IL13, IL4 and IL5 in APOE2hAβ males compared to APOE4hAβ males. Neuronal transmission and plasticity tests using field Input-Output (I/O) and long-term potentiation (LTP) recordings showed increased excitability in all APOE-carrying rats, with LTP deficits in APOE2hAβand APOE4hAβ rats compared to ApoehAβ and APOE3hAβ rats. Additionally, a lipidomic analysis of 222 lipid molecular species in serum samples showed that APOE2hAβ rats displayed elevated triglycerides and cholesterol, making them a valuable model for studying HLP. These rats also exhibited elevated levels of phosphatidylglycerol, phosphatidylserine, phosphatidylethanolamine, sphingomyelin, and lysophosphatidylcholine. Minimal differences in lipid profiles between APOE3hAβ and APOE4hAβ rats reflect findings from mouse models. Future studies will include comprehensive lipidomic analyses in various CNS regions and at older ages to further validate these models and explore the effects of APOE isoforms on lipid metabolism in relation to AD pathology.
Collapse
Affiliation(s)
- Metin Yesiltepe
- Department of Pharmacology, Physiology & Neuroscience New Jersey Medical School, The State University of New Jersey, Rutgers, Newark, NJ, USA
| | - Tao Yin
- Department of Pharmacology, Physiology & Neuroscience New Jersey Medical School, The State University of New Jersey, Rutgers, Newark, NJ, USA
| | - Marc Tambini
- Department of Pharmacology, Physiology & Neuroscience New Jersey Medical School, The State University of New Jersey, Rutgers, Newark, NJ, USA
| | - Hanmei Bao
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Meixia Pan
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Cristina d'Abramo
- Litwin-Zucker Center for the Study of Alzheimer's Disease and Memory Disorders, Feinstein Institutes for Medical Research, Institute of Molecular Medicine, Manhasset, NY, USA
| | - Luca Giliberto
- Litwin-Zucker Center for the Study of Alzheimer's Disease and Memory Disorders, Feinstein Institutes for Medical Research, Institute of Molecular Medicine, Manhasset, NY, USA
- Institute of Neurology and Neurosurgery, Northwell Health System, Manhasset, NY, USA
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
- Department of Medicine - Diabetes, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Luciano D'Adamio
- Department of Pharmacology, Physiology & Neuroscience New Jersey Medical School, The State University of New Jersey, Rutgers, Newark, NJ, USA.
| |
Collapse
|
7
|
Jackson RJ, Hyman BT, Serrano-Pozo A. Multifaceted roles of APOE in Alzheimer disease. Nat Rev Neurol 2024; 20:457-474. [PMID: 38906999 DOI: 10.1038/s41582-024-00988-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2024] [Indexed: 06/23/2024]
Abstract
For the past three decades, apolipoprotein E (APOE) has been known as the single greatest genetic modulator of sporadic Alzheimer disease (AD) risk, influencing both the average age of onset and the lifetime risk of developing AD. The APOEε4 allele significantly increases AD risk, whereas the ε2 allele is protective relative to the most common ε3 allele. However, large differences in effect size exist across ethnoracial groups that are likely to depend on both global genetic ancestry and local genetic ancestry, as well as gene-environment interactions. Although early studies linked APOE to amyloid-β - one of the two culprit aggregation-prone proteins that define AD - in the past decade, mounting work has associated APOE with other neurodegenerative proteinopathies and broader ageing-related brain changes, such as neuroinflammation, energy metabolism failure, loss of myelin integrity and increased blood-brain barrier permeability, with potential implications for longevity and resilience to pathological protein aggregates. Novel mouse models and other technological advances have also enabled a number of therapeutic approaches aimed at either attenuating the APOEε4-linked increased AD risk or enhancing the APOEε2-linked AD protection. This Review summarizes this progress and highlights areas for future research towards the development of APOE-directed therapeutics.
Collapse
Affiliation(s)
- Rosemary J Jackson
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, USA.
| | - Alberto Serrano-Pozo
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, USA.
| |
Collapse
|
8
|
Medegan Fagla B, York J, Christensen A, Dela Rosa C, Balu D, Pike CJ, Tai LM, Buhimschi IA. Apolipoprotein E polymorphisms and female fertility in a transgenic mouse model of Alzheimer's disease. Sci Rep 2024; 14:15873. [PMID: 38982272 PMCID: PMC11233746 DOI: 10.1038/s41598-024-66489-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 07/02/2024] [Indexed: 07/11/2024] Open
Abstract
Apolipoprotein E (APOE) is a major cholesterol carrier responsible for lipid transport and injury repair in the brain. The human APOE gene (h-APOE) has 3 naturally occurring alleles: ε3, the common allele; ε4, which increases Alzheimer's disease (AD) risk up to 15-fold; and ε2, the rare allele which protects against AD. Although APOE4 has negative effects on neurocognition in old age, its persistence in the population suggests a survival advantage. We investigated the relationship between APOE genotypes and fertility in EFAD mice, a transgenic mouse model expressing h-APOE. We show that APOE4 transgenic mice had the highest level of reproductive performance, followed by APOE3 and APOE2. Intriguingly, APOE3 pregnancies had more fetal resorptions and reduced fetal weights relative to APOE4 pregnancies. In conclusion, APOE genotypes impact fertility and pregnancy outcomes in female mice, in concordance with findings in human populations. These mouse models may help elucidate how h-APOE4 promotes reproductive fitness at the cost of AD in later life.
Collapse
Affiliation(s)
- Bani Medegan Fagla
- Department of Obstetrics Gynecology, University of Illinois at Chicago College of Medicine, 820 S. Wood Street, Chicago, IL, 60612, USA
| | - Jason York
- Department of Anatomy and Cell Biology, University of Illinois at Chicago College of Medicine, Chicago, IL, 60612, USA
| | - Amy Christensen
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Cielo Dela Rosa
- Department of Obstetrics Gynecology, University of Illinois at Chicago College of Medicine, 820 S. Wood Street, Chicago, IL, 60612, USA
| | - Deebika Balu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago College of Medicine, Chicago, IL, 60612, USA
| | - Christian J Pike
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Leon M Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago College of Medicine, Chicago, IL, 60612, USA
| | - Irina A Buhimschi
- Department of Obstetrics Gynecology, University of Illinois at Chicago College of Medicine, 820 S. Wood Street, Chicago, IL, 60612, USA.
| |
Collapse
|
9
|
Lusiki Z, Blom D, Soko ND, Malema S, Jones E, Rayner B, Blackburn J, Sinxadi P, Dandara MT, Dandara C. Major Genetic Drivers of Statin Treatment Response in African Populations and Pharmacogenetics of Dyslipidemia Through a One Health Lens. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2024; 28:261-279. [PMID: 37956269 DOI: 10.1089/omi.2023.0122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
A One Health lens is increasingly significant to address the intertwined challenges in planetary health concerned with the health of humans, nonhuman animals, plants, and ecosystems. A One Health approach can benefit the public health systems in Africa that are overburdened by noncommunicable, infectious, and environmental diseases. Notably, the COVID-19 pandemic revealed the previously overlooked two-fold importance of pharmacogenetics (PGx), for individually tailored treatment of noncommunicable diseases and environmental pathogens. For example, dyslipidemia, a common cardiometabolic risk factor, has been identified as an independent COVID-19 severity risk factor. Observational data suggest that patients with COVID-19 infection receiving lipid-lowering therapy may have better outcomes. However, among African patients, the response to these drugs varies from patient to patient, pointing to the possible contribution of genetic variation in important pharmacogenes. The PGx of lipid-lowering therapies may underlie differences in treatment responses observed among dyslipidemia patients as well as patients comorbid with COVID-19 and dyslipidemia. Genetic variations in APOE, ABCB1, CETP, CYP2C9, CYP3A4, CYP3A5, HMGCR, LDLR, NPC1L1, and SLCO1B1 genes affect the pharmacogenomics of statins, and they have individually been linked to differential responses to dyslipidemia and COVID-19 treatment. African populations are underrepresented in PGx research. This leads to poor accounting of additional diverse genetic variants that could be important in understanding interindividual and between-population variations in therapeutic responses to dyslipidemia and COVID-19. This expert review examines and synthesizes the salient and priority PGx variations, as seen through a One Health lens in Africa, to improve and inform personalized medicine in both dyslipidemia and COVID-19.
Collapse
Affiliation(s)
- Zizo Lusiki
- Division of Human Genetics, Department of Pathology, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
- Platform for Pharmacogenomics Research and Translation (PREMED) Unit, South African Medical Research Council (SAMRC), Cape Town, South Africa
| | - Dirk Blom
- Platform for Pharmacogenomics Research and Translation (PREMED) Unit, South African Medical Research Council (SAMRC), Cape Town, South Africa
- Division of Lipidology and Cape Heart Institute, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Nyarai D Soko
- Division of Human Genetics, Department of Pathology, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
- Platform for Pharmacogenomics Research and Translation (PREMED) Unit, South African Medical Research Council (SAMRC), Cape Town, South Africa
| | - Smangele Malema
- Platform for Pharmacogenomics Research and Translation (PREMED) Unit, South African Medical Research Council (SAMRC), Cape Town, South Africa
| | - Erika Jones
- Platform for Pharmacogenomics Research and Translation (PREMED) Unit, South African Medical Research Council (SAMRC), Cape Town, South Africa
- Division of Nephrology and Hypertension, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Brian Rayner
- Platform for Pharmacogenomics Research and Translation (PREMED) Unit, South African Medical Research Council (SAMRC), Cape Town, South Africa
- Division of Nephrology and Hypertension, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Jonathan Blackburn
- Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa
| | - Phumla Sinxadi
- Platform for Pharmacogenomics Research and Translation (PREMED) Unit, South African Medical Research Council (SAMRC), Cape Town, South Africa
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Michelle T Dandara
- Platform for Pharmacogenomics Research and Translation (PREMED) Unit, South African Medical Research Council (SAMRC), Cape Town, South Africa
| | - Collet Dandara
- Division of Human Genetics, Department of Pathology, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
- Platform for Pharmacogenomics Research and Translation (PREMED) Unit, South African Medical Research Council (SAMRC), Cape Town, South Africa
| |
Collapse
|
10
|
Kondkar AA, Azad TA, Sultan T, Khatlani T, Alshehri AA, Radhakrishnan R, Lobo GP, Alsirhy E, Almobarak FA, Osman EA, Al-Obeidan SA. APOE ε2-Carriers Are Associated with an Increased Risk of Primary Angle-Closure Glaucoma in Patients of Saudi Origin. Int J Mol Sci 2024; 25:4571. [PMID: 38674156 PMCID: PMC11050284 DOI: 10.3390/ijms25084571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
This study investigated the association between apolipoprotein E (APOE) gene polymorphisms (rs429358 and rs7412) and primary angle-closure glaucoma (PACG) and pseudoexfoliation glaucoma (PXG) in a Saudi cohort. Genotyping of 437 DNA samples (251 controls, 92 PACG, 94 PXG) was conducted using PCR-based Sanger sequencing. The results showed no significant differences in the allele and genotype frequencies of rs429358 and rs7412 between the PACG/PXG cases and controls. Haplotype analysis revealed ε3 as predominant, followed by ε4 and ε2 alleles, with no significant variance in PACG/PXG. However, APOE genotype analysis indicated a significant association between ε2-carriers and PACG (odds ratio = 4.82, 95% CI 1.52-15.26, p = 0.007), whereas no notable association was observed with PXG. Logistic regression confirmed ε2-carriers as a significant predictor for PACG (p = 0.008), while age emerged as significant for PXG (p < 0.001). These findings suggest a potential role of ε2-carriers in PACG risk within the Saudi cohort. Further validation and larger-scale investigations are essential to elucidate the precise role of APOE in PACG pathogenesis and progression.
Collapse
Affiliation(s)
- Altaf A. Kondkar
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh 11411, Saudi Arabia (E.A.); (S.A.A.-O.)
- Glaucoma Research Chair in Ophthalmology, College of Medicine, King Saud University, Riyadh 11411, Saudi Arabia
- King Saud University Medical City, King Saud University, Riyadh 11411, Saudi Arabia
| | - Taif A. Azad
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh 11411, Saudi Arabia (E.A.); (S.A.A.-O.)
| | - Tahira Sultan
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh 11411, Saudi Arabia (E.A.); (S.A.A.-O.)
| | - Tanvir Khatlani
- Department of Blood and Cancer Research, King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University of Health Sciences, Ministry of National Guard Health Affairs, Riyadh 11426, Saudi Arabia
| | - Abdulaziz A. Alshehri
- Department of Ophthalmology, Imam Abdulrahman Alfaisal Hospital, Riyadh 14723, Saudi Arabia
| | - Rakesh Radhakrishnan
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55347, USA; (R.R.)
| | - Glenn P. Lobo
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55347, USA; (R.R.)
| | - Ehab Alsirhy
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh 11411, Saudi Arabia (E.A.); (S.A.A.-O.)
| | - Faisal A. Almobarak
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh 11411, Saudi Arabia (E.A.); (S.A.A.-O.)
| | - Essam A. Osman
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh 11411, Saudi Arabia (E.A.); (S.A.A.-O.)
| | - Saleh A. Al-Obeidan
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh 11411, Saudi Arabia (E.A.); (S.A.A.-O.)
- Glaucoma Research Chair in Ophthalmology, College of Medicine, King Saud University, Riyadh 11411, Saudi Arabia
| |
Collapse
|
11
|
Ciesielski TH, Sirugo G, Iyengar SK, Williams SM. Characterizing the pathogenicity of genetic variants: the consequences of context. NPJ Genom Med 2024; 9:3. [PMID: 38195641 PMCID: PMC10776585 DOI: 10.1038/s41525-023-00386-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 12/15/2023] [Indexed: 01/11/2024] Open
Affiliation(s)
- Timothy H Ciesielski
- The Department of Population and Quantitative Health Sciences at Case Western Reserve University School of Medicine, Cleveland, OH, USA.
- Mary Ann Swetland Center for Environmental Health at Case Western Reserve University School of Medicine, Cleveland, OH, USA.
- Ronin Institute, Montclair, NJ, USA.
| | - Giorgio Sirugo
- The Department of Population and Quantitative Health Sciences at Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Institute of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sudha K Iyengar
- The Department of Population and Quantitative Health Sciences at Case Western Reserve University School of Medicine, Cleveland, OH, USA
- The Department of Genetics and Genome Sciences at Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Cleveland Institute for Computational Biology, Cleveland, OH, USA
| | - Scott M Williams
- The Department of Population and Quantitative Health Sciences at Case Western Reserve University School of Medicine, Cleveland, OH, USA
- The Department of Genetics and Genome Sciences at Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Cleveland Institute for Computational Biology, Cleveland, OH, USA
| |
Collapse
|
12
|
Oriá RB, Smith CJ, Ashford JW, Vitek MP, Guerrant RL. Pros and Cons of APOE4 Homozygosity and Effects on Neuroplasticity, Malnutrition, and Infections in Early Life Adversity, Alzheimer's Disease, and Alzheimer's Prevention. J Alzheimers Dis 2024; 100:S179-S185. [PMID: 39093076 DOI: 10.3233/jad-240888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Fortea et al.'s. (2024) recent data analysis elegantly calls attention to familial late-onset Alzheimer's disease (AD) with APOE4 homozygosity. The article by Grant (2024) reviews the factors associated with AD, particularly the APOE genotype and lifestyle, and the broad implications for prevention, both for individuals with the lifestyles associated with living in resource-rich countries and for those enduring environmental adversity in poverty settings, including high exposure to enteric pathogens and precarious access to healthcare. Grant discusses the issue of APOE genotype and its implications for the benefits of lifestyle modifications. This review highlights that bearing APOE4 could constitute an evolutionary benefit in coping with heavy enteric infections and malnutrition early in life in the critical formative first two years of brain development. However, the critical issue may be that this genotype could be a health concern under shifts in lifestyle and unhealthy diets during aging, leading to severe cognitive impairments and increased risk of AD. This commentary supports the discussions of Grant and the benefits of improving lifestyle for decreasing the risks for AD while providing further understanding and modelling of the early life benefits of APOE4 amidst adversity. This attention to the pathophysiology of AD should help further elucidate these critical, newly appreciated pathogenic pathways for developing approaches to the prevention and management in the context of the APOE genetic variations associated with AD.
Collapse
Affiliation(s)
- Reinaldo B Oriá
- Department of Morphology, Laboratory of Tissue Healing, Ontogeny, and Nutrition, School of Medicine, Institute of Biomedicine, Federal University of Ceara, Fortaleza, Brazil
| | - Carr J Smith
- Society for Brain Mapping and Therapeutics, Pacific Palisades, CA, USA
| | - J Wesson Ashford
- War Related Illness and Injury Study Center, VA Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Psychiatry and Behavioral Sciences, Stanford University, Palo Alto, CA, USA
| | - Michael P Vitek
- Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - Richard L Guerrant
- Department of Medicine, Division of Infectious Diseases and International Health, School of Medicine, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
13
|
Grant WB. A Brief History of the Progress in Our Understanding of Genetics and Lifestyle, Especially Diet, in the Risk of Alzheimer's Disease. J Alzheimers Dis 2024; 100:S165-S178. [PMID: 39121130 PMCID: PMC11380269 DOI: 10.3233/jad-240658] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/11/2024]
Abstract
The two major determining factors for Alzheimer's disease (AD) are genetics and lifestyle. Alleles of the apolipoprotein E (APOE) gene play important roles in the development of late-onset AD, with APOEɛ4 increasing risk, APOEɛ3 being neutral, and APOEɛ2 reducing risk. Several modifiable lifestyle factors have been studied in terms of how they can modify the risk of AD. Among these factors are dietary pattern, nutritional supplements such as omega-3 fatty acids, and B vitamins, physical exercise, and obesity, and vitamin D. The Western diet increases risk of AD, while dietary patterns such as the Mediterranean and vegetarian/vegan diets reduce risk. Foods associated with reduced risk include coffee, fruits and vegetables, whole grains and legumes, and fish, while meat and ultraprocessed foods are associated with increased risk, especially when they lead to obesity. In multi-country ecological studies, the amount of meat in the national diet has the highest correlation with risk of AD. The history of research regarding dietary patterns on risk of AD is emphasized in this review. The risk of AD can be modified starting at least by mid-life. People with greater genetic risk for AD would benefit more by choosing lifestyle factors to reduce and/or delay incidence of AD.
Collapse
Affiliation(s)
- William B Grant
- Sunlight, Nutrition, and Health Research Center, San Francisco, CA, USA
| |
Collapse
|
14
|
Brugger SW, Davis MF. Influence of Admixture on Phenotypes. Curr Protoc 2023; 3:e953. [PMID: 38146906 DOI: 10.1002/cpz1.953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2023]
Abstract
Individuals of European descent have historically been the focus of genetic studies and possess relatively homogenous genomes. As a result, analytical methods have been developed and optimized with such genomes in mind. African-descent and Latino individuals generally possess genomes of greater architectural complexity due to mosaic genomic ancestry, which can extensively and intricately impact phenotypic expression. As such, genetic analyses of admixed individuals require that genetic admixture be quantified to accurately model the impact of genetic variation on phenotypic expression. In this overview, we explore how fundamental genetic concepts such as linkage disequilibrium and differential allele frequency interact with genetic admixture to uniquely influence phenotypes in admixed individuals. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Steven W Brugger
- Department of Molecular Biology and Microbiology, Brigham Young University, Provo, Utah
| | - Mary F Davis
- Department of Molecular Biology and Microbiology, Brigham Young University, Provo, Utah
| |
Collapse
|
15
|
Affiliation(s)
- Emile Wogram
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany.
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
16
|
Trumble BC, Charifson M, Kraft T, Garcia AR, Cummings DK, Hooper P, Lea AJ, Eid Rodriguez D, Koebele SV, Buetow K, Beheim B, Minocher R, Gutierrez M, Thomas GS, Gatz M, Stieglitz J, Finch CE, Kaplan H, Gurven M. Apolipoprotein-ε 4 is associated with higher fecundity in a natural fertility population. SCIENCE ADVANCES 2023; 9:eade9797. [PMID: 37556539 PMCID: PMC10411886 DOI: 10.1126/sciadv.ade9797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 07/11/2023] [Indexed: 08/11/2023]
Abstract
In many populations, the apolipoprotein-ε4 (APOE-ε4) allele increases the risk for several chronic diseases of aging, including dementia and cardiovascular disease; despite these harmful effects at later ages, the APOE-ε4 allele remains prevalent. We assess the impact of APOE-ε4 on fertility and its proximate determinants (age at first reproduction, interbirth interval) among the Tsimane, a natural fertility population of forager-horticulturalists. Among 795 women aged 13 to 90 (20% APOE-ε4 carriers), those with at least one APOE-ε4 allele had 0.3 to 0.5 more children than (ε3/ε3) homozygotes, while those with two APOE-ε4 alleles gave birth to 1.4 to 2.1 more children. APOE-ε4 carriers achieve higher fertility by beginning reproduction 0.8 years earlier and having a 0.23-year shorter interbirth interval. Our findings add to a growing body of literature suggesting a need for studies of populations living in ancestrally relevant environments to assess how alleles that are deleterious in sedentary urban environments may have been maintained by selection throughout human evolutionary history.
Collapse
Affiliation(s)
- Benjamin C. Trumble
- School of Human Evolution and Social Change, Arizona State University, Tempe, AZ, USA
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA
| | - Mia Charifson
- Department of Population Health, New York University Grossman School of Medicine, New York City, NY, USA
| | - Tom Kraft
- Anthropology Department, University of Utah, Salt Lake City, UT, USA
| | - Angela R. Garcia
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA
- Scientific Research Core, Phoenix Children's Hospital, Phoenix, AZ, USA
- Department of Child Health, University of Arizona, Tucson, AZ, USA
| | - Daniel K. Cummings
- Department of Health Economics and Anthropology, Economic Science Institute, Argyros School of Business and Economics, Chapman University, Orange, CA, USA
| | - Paul Hooper
- Department of Health Economics and Anthropology, Economic Science Institute, Argyros School of Business and Economics, Chapman University, Orange, CA, USA
| | - Amanda J. Lea
- Child and Brain Development Program, Canadian Institute for Advanced Research, Toronto, Ontario, Canada
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | | | | | - Kenneth Buetow
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Bret Beheim
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Riana Minocher
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | | | - Gregory S. Thomas
- MemorialCare Health System, Fountain Valley, CA, USA
- University of California, Irvine, CA, USA
| | - Margaret Gatz
- Center for Economic and Social Research, University of Southern California, Los Angeles, CA, USA
| | - Jonathan Stieglitz
- Institute for Advanced Study in Toulouse, Université Toulouse 1 Capitole, Toulouse, France
| | - Caleb E. Finch
- Leonard Davis School of Gerontology and Dornsife College, University of Southern California, Los Angeles, CA, USA
| | - Hillard Kaplan
- Department of Health Economics and Anthropology, Economic Science Institute, Argyros School of Business and Economics, Chapman University, Orange, CA, USA
| | - Michael Gurven
- Department of Anthropology, University of California Santa Barbara, Santa Barbara, CA, USA
| |
Collapse
|
17
|
Yang LG, March ZM, Stephenson RA, Narayan PS. Apolipoprotein E in lipid metabolism and neurodegenerative disease. Trends Endocrinol Metab 2023; 34:430-445. [PMID: 37357100 PMCID: PMC10365028 DOI: 10.1016/j.tem.2023.05.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 06/27/2023]
Abstract
Dysregulation of lipid metabolism has emerged as a central component of many neurodegenerative diseases. Variants of the lipid transport protein, apolipoprotein E (APOE), modulate risk and resilience in several neurodegenerative diseases including late-onset Alzheimer's disease (LOAD). Allelic variants of the gene, APOE, alter the lipid metabolism of cells and tissues and have been broadly associated with several other cellular and systemic phenotypes. Targeting APOE-associated metabolic pathways may offer opportunities to alter disease-related phenotypes and consequently, attenuate disease risk and impart resilience to multiple neurodegenerative diseases. We review the molecular, cellular, and tissue-level alterations to lipid metabolism that arise from different APOE isoforms. These changes in lipid metabolism could help to elucidate disease mechanisms and tune neurodegenerative disease risk and resilience.
Collapse
Affiliation(s)
- Linda G Yang
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA
| | - Zachary M March
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA
| | - Roxan A Stephenson
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA
| | - Priyanka S Narayan
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA.; National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health, Bethesda, MD, USA; Center for Alzheimer's and Related Dementias (CARD), National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
18
|
Smith CJ, Ashford JW. Apolipoprotein ɛ4-Associated Protection Against Pediatric Enteric Infections is a Survival Advantage in Pre-Industrial Populations. J Alzheimers Dis 2023:JAD221218. [PMID: 37125551 DOI: 10.3233/jad-221218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Until 300,000 years ago, ancestors of modern humans ubiquitously carried the apolipoprotein E (APOE) ɛ4/ɛ4 genotype, when the ɛ3 allele mutated from the ancestral ɛ4, which elevates the risk of Alzheimer's disease. Modern humans living today predominantly carry the ɛ3 allele, which provides protection against heart disease and dementia in long-lived populations. The ancestral ɛ4 allele has been highly preserved in isolated populations in tropical and Arctic regions with high pathogen burdens, e.g., helminths. Early humans experienced serious enteric infections that exerted evolutionary selection pressure, and factors that mitigate infant and childhood mortality from enteric infections also exert selection pressure. Some bacteria can exploit the host's defensive inflammatory response to colonize and invade the host. Pathogen-induced inflammation associated with infant and childhood diarrhea can damage the gut wall long after the invading organisms are no longer present. Inflammation not only resides in the mucosal wall, but also induces systemic inflammation. Baseline systemic inflammation is lower in ɛ4 carriers, yet ɛ4 carriers display a stronger host inflammatory response that reduces pathogen burdens, increasing infant and early childhood survival. Evolutionary selection of the ɛ3 allele likely occurred after humans moved into temperate zones with lower pathogen burdens, unrelated to protection from Alzheimer's disease.
Collapse
Affiliation(s)
| | - J Wesson Ashford
- Stanford University and VA Palo Alto Health Care System, Palo Alto, CA, USA
| |
Collapse
|
19
|
Li Y, Chang J, Chen X, Liu J, Zhao L. Advances in the Study of APOE and Innate Immunity in Alzheimer's Disease. J Alzheimers Dis 2023; 93:1195-1210. [PMID: 37182889 DOI: 10.3233/jad-230179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Alzheimer's disease (AD) is a progressive degenerative disease of the nervous system (CNS) with an insidious onset. Clinically, it is characterized by a full range of dementia manifestations including memory impairment, aphasia, loss of speech, loss of use, loss of recognition, impairment of visuospatial skills, and impairment of executive function, as well as changes in personality and behavior. The exact cause of AD has not yet been identified. Nevertheless, modern research indicates that genetic factors contribute to 70% of human's risk of AD. Apolipoprotein (APOE) accounts for up to 90% of the genetic predisposition. APOE is a crucial gene that cannot be overstated. In addition, innate immunity plays a significant role in the etiology and treatment of AD. Understanding the different subtypes of APOE and their interconnections is of paramount importance. APOE and innate immunity, along with their relationship to AD, are primary research motivators for in-depth research and clinical trials. The exploration of novel technologies has led to an increasing trend in the study of AD at the cellular and molecular levels and continues to make more breakthroughs and progress. As of today, there is no effective treatment available for AD around the world. This paper aims to summarize and analyze the role of APOE and innate immunity, as well as development trends in recent years. It is anticipated that APOE and innate immunity will provide a breakthrough for humans to hinder AD progression in the near future.
Collapse
Affiliation(s)
- Yujiao Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Jun Chang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Xi Chen
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Jianwei Liu
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lan Zhao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
20
|
Kulminski AM, Jain‐Washburn E, Philipp I, He L, Loika Y, Loiko E, Bagley O, Ukraintseva S, Yashin A, Arbeev K, Stallard E, Feitosa MF, Schupf N, Christensen K, Culminskaya I. APOE ɛ4 allele and TOMM40-APOC1 variants jointly contribute to survival to older ages. Aging Cell 2022; 21:e13730. [PMID: 36330582 PMCID: PMC9741507 DOI: 10.1111/acel.13730] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 09/23/2022] [Accepted: 03/10/2022] [Indexed: 11/06/2022] Open
Abstract
Age-related diseases characteristic of post-reproductive life, aging, and life span are the examples of polygenic non-Mendelian traits with intricate genetic architectures. Polygenicity of these traits implies that multiple variants can impact their risks independently or jointly as combinations of specific variants. Here, we examined chances to live to older ages, 85 years and older, for carriers of compound genotypes comprised of combinations of genotypes of rs429358 (APOE ɛ4 encoding polymorphism), rs2075650 (TOMM40), and rs12721046 (APOC1) polymorphisms using data from four human studies. The choice of these polymorphisms was motivated by our prior results showing that the ɛ4 carriers having minor alleles of the other two polymorphisms were at exceptionally high risk of Alzheimer's disease (AD), compared with non-carriers of the minor alleles. Consistent with our prior findings for AD, we show here that the adverse effect of the ɛ4 allele on survival to older ages is significantly higher in carriers of minor alleles of rs2075650 and/or rs12721046 polymorphisms compared with their non-carriers. The exclusion of AD cases made this effect stronger. Our results provide compelling evidence that AD does not mediate the associations of the same compound genotypes with chances to survive until older ages, indicating the existence of genetically heterogeneous mechanisms. The survival chances can be mainly associated with lipid- and immunity-related mechanisms, whereas the AD risk, can be driven by the AD-biomarker-related mechanism, among others. Targeting heterogeneous polygenic profiles of individuals at high risks of complex traits is promising for the translation of genetic discoveries to health care.
Collapse
Affiliation(s)
- Alexander M. Kulminski
- Biodemography of Aging Research Unit, Social Science Research InstituteDuke UniversityDurhamNorth CarolinaUSA
| | - Ethan Jain‐Washburn
- Biodemography of Aging Research Unit, Social Science Research InstituteDuke UniversityDurhamNorth CarolinaUSA
| | - Ian Philipp
- Biodemography of Aging Research Unit, Social Science Research InstituteDuke UniversityDurhamNorth CarolinaUSA
| | - Liang He
- Biodemography of Aging Research Unit, Social Science Research InstituteDuke UniversityDurhamNorth CarolinaUSA
| | - Yury Loika
- Biodemography of Aging Research Unit, Social Science Research InstituteDuke UniversityDurhamNorth CarolinaUSA
| | - Elena Loiko
- Biodemography of Aging Research Unit, Social Science Research InstituteDuke UniversityDurhamNorth CarolinaUSA
| | - Olivia Bagley
- Biodemography of Aging Research Unit, Social Science Research InstituteDuke UniversityDurhamNorth CarolinaUSA
| | - Svetlana Ukraintseva
- Biodemography of Aging Research Unit, Social Science Research InstituteDuke UniversityDurhamNorth CarolinaUSA
| | - Anatoliy Yashin
- Biodemography of Aging Research Unit, Social Science Research InstituteDuke UniversityDurhamNorth CarolinaUSA
| | - Konstantin Arbeev
- Biodemography of Aging Research Unit, Social Science Research InstituteDuke UniversityDurhamNorth CarolinaUSA
| | - Eric Stallard
- Biodemography of Aging Research Unit, Social Science Research InstituteDuke UniversityDurhamNorth CarolinaUSA
| | - Mary F. Feitosa
- Division of Statistical Genomics, Department of GeneticsWashington University School of MedicineSt LouisMissouriUSA
| | - Nicole Schupf
- Gertrude H. Sergievsky CenterColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Kaare Christensen
- Unit of Epidemiology, Biostatistics and Biodemography, Department of Public HealthSouthern Denmark UniversityOdenseDenmark
| | - Irina Culminskaya
- Biodemography of Aging Research Unit, Social Science Research InstituteDuke UniversityDurhamNorth CarolinaUSA
| |
Collapse
|
21
|
Cachide M, Carvalho L, Rosa IM, Wiltfang J, Henriques AG, da Cruz e Silva OAB. BIN1 rs744373 SNP and APOE alleles specifically associate to common diseases. FRONTIERS IN DEMENTIA 2022; 1:1001113. [PMID: 39081475 PMCID: PMC11285651 DOI: 10.3389/frdem.2022.1001113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 10/12/2022] [Indexed: 08/02/2024]
Abstract
APOE ε4 and BIN1 are the two main genetic risk factors for sporadic Alzheimer's Disease (AD). Among several BIN1 variants, the rs744373 is frequently associated with AD risk by contributing to tau pathology and poor cognitive performance. This study addressed the association of APOE and BIN1 rs744373 to specific characteristics in a Portuguese primary care-based study group, denoted pcb-Cohort. The study included 590 participants from five primary care health centers in the Aveiro district of Portugal. Individuals were evaluated and scored for cognitive and clinical characteristics, and blood samples were collected from the volunteers meeting the inclusion and exclusion criteria (N = 505). APOE and BIN1 genotypes were determined, and their association with cognitive characteristics and other diseases that might contribute to cognitive deficits, namely depression, hypertension, type 2 diabetes, dyslipidemia, osteoarticular diseases, gastrointestinal diseases, cardiovascular and respiratory diseases, was assessed. The diseases attributed to the study group were those previously diagnosed and confirmed by specialists. The results generated through multivariate analysis show that APOE ε4 carriers significantly associated with poorer cognitive performance (OR = 2.527; p = 0.031). Additionally, there was a significant risk of dyslipidemia for APOE ε4 carriers (OR = 1.804; p = 0.036), whereas BIN1 rs744373 risk-allele carriers were at a significantly lower risk of having dyslipidemia (OR = 0.558; p = 0.006). Correlations were evident for respiratory diseases in which APOE ε4 showed a protective tendency (OR = 0.515; p = 0.088), and BIN1 had a significative protective profile (OR = 0.556; p = 0.026). Not of statistical significance, APOE ε2 showed a trend to protect against type 2 diabetes (OR = 0.342; p = 0.093), in contrast BIN1 rs744373 risk-allele carriers were more likely to exhibit the disease (OR = 1.491; p = 0.099). The data here presented clearly show, for the first time, that the two top genetic risk factors for sporadic AD impact a similar group of common diseases, namely dyslipidemia, respiratory diseases, and type 2 diabetes.
Collapse
Affiliation(s)
- Maria Cachide
- Neurosciences and Signalling Group, Medical Sciences Department, Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro, Portugal
| | - Liliana Carvalho
- Neurosciences and Signalling Group, Medical Sciences Department, Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro, Portugal
| | - Ilka Martins Rosa
- Neurosciences and Signalling Group, Medical Sciences Department, Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro, Portugal
| | - Jens Wiltfang
- Neurosciences and Signalling Group, Medical Sciences Department, Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro, Portugal
- Department of Psychiatry and Psychotherapy, University Medical Centre Goettingen (UMG), Georg-August University, Goettingen, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Goettingen, Germany
| | - Ana Gabriela Henriques
- Neurosciences and Signalling Group, Medical Sciences Department, Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro, Portugal
| | - Odete A. B. da Cruz e Silva
- Neurosciences and Signalling Group, Medical Sciences Department, Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
22
|
Fernández-Calle R, Konings SC, Frontiñán-Rubio J, García-Revilla J, Camprubí-Ferrer L, Svensson M, Martinson I, Boza-Serrano A, Venero JL, Nielsen HM, Gouras GK, Deierborg T. APOE in the bullseye of neurodegenerative diseases: impact of the APOE genotype in Alzheimer's disease pathology and brain diseases. Mol Neurodegener 2022; 17:62. [PMID: 36153580 PMCID: PMC9509584 DOI: 10.1186/s13024-022-00566-4] [Citation(s) in RCA: 111] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 08/29/2022] [Indexed: 02/06/2023] Open
Abstract
ApoE is the major lipid and cholesterol carrier in the CNS. There are three major human polymorphisms, apoE2, apoE3, and apoE4, and the genetic expression of APOE4 is one of the most influential risk factors for the development of late-onset Alzheimer's disease (AD). Neuroinflammation has become the third hallmark of AD, together with Amyloid-β plaques and neurofibrillary tangles of hyperphosphorylated aggregated tau protein. This review aims to broadly and extensively describe the differential aspects concerning apoE. Starting from the evolution of apoE to how APOE's single-nucleotide polymorphisms affect its structure, function, and involvement during health and disease. This review reflects on how APOE's polymorphisms impact critical aspects of AD pathology, such as the neuroinflammatory response, particularly the effect of APOE on astrocytic and microglial function and microglial dynamics, synaptic function, amyloid-β load, tau pathology, autophagy, and cell-cell communication. We discuss influential factors affecting AD pathology combined with the APOE genotype, such as sex, age, diet, physical exercise, current therapies and clinical trials in the AD field. The impact of the APOE genotype in other neurodegenerative diseases characterized by overt inflammation, e.g., alpha- synucleinopathies and Parkinson's disease, traumatic brain injury, stroke, amyotrophic lateral sclerosis, and multiple sclerosis, is also addressed. Therefore, this review gathers the most relevant findings related to the APOE genotype up to date and its implications on AD and CNS pathologies to provide a deeper understanding of the knowledge in the APOE field.
Collapse
Affiliation(s)
- Rosalía Fernández-Calle
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Sabine C. Konings
- Department of Experimental Medical Science, Experimental Dementia Research Unit, Lund University, Lund, Sweden
| | - Javier Frontiñán-Rubio
- Oxidative Stress and Neurodegeneration Group, Faculty of Medicine, Universidad de Castilla-La Mancha, Ciudad Real, Spain
| | - Juan García-Revilla
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
- Departamento de Bioquímica Y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Lluís Camprubí-Ferrer
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Martina Svensson
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Isak Martinson
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Antonio Boza-Serrano
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
- Departamento de Bioquímica Y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - José Luís Venero
- Departamento de Bioquímica Y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Henrietta M. Nielsen
- Department of Biochemistry and Biophysics at, Stockholm University, Stockholm, Sweden
| | - Gunnar K. Gouras
- Department of Experimental Medical Science, Experimental Dementia Research Unit, Lund University, Lund, Sweden
| | - Tomas Deierborg
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| |
Collapse
|
23
|
Levy G, Levin B. An Evolution-Based Model of Causation for Aging-Related Diseases and Intrinsic Mortality: Explanatory Properties and Implications for Healthy Aging. Front Public Health 2022; 10:774668. [PMID: 35252084 PMCID: PMC8894190 DOI: 10.3389/fpubh.2022.774668] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 01/10/2022] [Indexed: 01/07/2023] Open
Abstract
Aging-related diseases are the most prevalent diseases in advanced countries nowadays, accounting for a substantial proportion of mortality. We describe the explanatory properties of an evolution-based model of causation (EBMC) applicable to aging-related diseases and intrinsic mortality. The EBMC takes the sufficient and component causes model of causation as a starting point and develops it using evolutionary and statistical theories. Genetic component causes are classified as “early-onset” or “late-onset” and environmental component causes as “evolutionarily conserved” or “evolutionarily recent.” Genetic and environmental component causes are considered to occur as random events following time-to-event distributions, and sufficient causes are classified according to whether or not their time-to-event distributions are “molded” by the declining force of natural selection with increasing age. We obtain for each of these two groups different time-to-event distributions for disease incidence or intrinsic mortality asymptotically (i.e., for a large number of sufficient causes). The EBMC provides explanations for observations about aging-related diseases concerning the penetrance of genetic risk variants, the age of onset of monogenic vs. sporadic forms, the meaning of “age as a risk factor,” the relation between frequency and age of onset, and the emergence of diseases associated with the modern Western lifestyle. The EBMC also provides an explanation of the Gompertz mortality model at the fundamental level of genetic causes and involving evolutionary biology. Implications for healthy aging are examined under the scenarios of health promotion and postponed aging. Most importantly from a public health standpoint, the EBMC implies that primary prevention through changes in lifestyle and reduction of environmental exposures is paramount in promoting healthy aging.
Collapse
Affiliation(s)
- Gilberto Levy
- Independent Researcher, Rio de Janeiro, Brazil
- *Correspondence: Gilberto Levy
| | - Bruce Levin
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, United States
| |
Collapse
|
24
|
Troutwine BR, Hamid L, Lysaker CR, Strope TA, Wilkins HM. Apolipoprotein E and Alzheimer's disease. Acta Pharm Sin B 2022; 12:496-510. [PMID: 35256931 PMCID: PMC8897057 DOI: 10.1016/j.apsb.2021.10.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/30/2021] [Accepted: 10/07/2021] [Indexed: 12/14/2022] Open
Abstract
Genetic variation in apolipoprotein E (APOE) influences Alzheimer's disease (AD) risk. APOE ε4 alleles are the strongest genetic risk factor for late onset sporadic AD. The AD risk is dose dependent, as those carrying one APOE ε4 allele have a 2-3-fold increased risk, while those carrying two ε4 alleles have a 10-15-fold increased risk. Individuals carrying APOE ε2 alleles have lower AD risk and those carrying APOE ε3 alleles have neutral risk. APOE is a lipoprotein which functions in lipid transport, metabolism, and inflammatory modulation. Isoform specific effects of APOE within the brain include alterations to Aβ, tau, neuroinflammation, and metabolism. Here we review the association of APOE with AD, the APOE isoform specific effects within brain and periphery, and potential therapeutics.
Collapse
Affiliation(s)
- Benjamin R. Troutwine
- Department of Neurology University of Kansas Medical Center, Kansas City, KS 66160, USA
- University of Kansas Alzheimer's Disease Center, Kansas City, KS 66160, USA
| | - Laylan Hamid
- University of Kansas Alzheimer's Disease Center, Kansas City, KS 66160, USA
| | - Colton R. Lysaker
- University of Kansas Alzheimer's Disease Center, Kansas City, KS 66160, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Taylor A. Strope
- University of Kansas Alzheimer's Disease Center, Kansas City, KS 66160, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Heather M. Wilkins
- Department of Neurology University of Kansas Medical Center, Kansas City, KS 66160, USA
- University of Kansas Alzheimer's Disease Center, Kansas City, KS 66160, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
25
|
Bin-Jumah MN, Nadeem MS, Gilani SJ, Al-Abbasi FA, Ullah I, Alzarea SI, Ghoneim MM, Alshehri S, Uddin A, Murtaza BN, Kazmi I. Genes and Longevity of Lifespan. Int J Mol Sci 2022; 23:1499. [PMID: 35163422 PMCID: PMC8836117 DOI: 10.3390/ijms23031499] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/04/2022] [Accepted: 01/26/2022] [Indexed: 12/12/2022] Open
Abstract
Aging is a complex process indicated by low energy levels, declined physiological activity, stress induced loss of homeostasis leading to the risk of diseases and mortality. Recent developments in medical sciences and an increased availability of nutritional requirements has significantly increased the average human lifespan worldwide. Several environmental and physiological factors contribute to the aging process. However, about 40% human life expectancy is inherited among generations, many lifespan associated genes, genetic mechanisms and pathways have been demonstrated during last decades. In the present review, we have evaluated many human genes and their non-human orthologs established for their role in the regulation of lifespan. The study has included more than fifty genes reported in the literature for their contributions to the longevity of life. Intact genomic DNA is essential for the life activities at the level of cell, tissue, and organ. Nucleic acids are vulnerable to oxidative stress, chemotherapies, and exposure to radiations. Efficient DNA repair mechanisms are essential for the maintenance of genomic integrity, damaged DNA is not replicated and transferred to next generations rather the presence of deleterious DNA initiates signaling cascades leading to the cell cycle arrest or apoptosis. DNA modifications, DNA methylation, histone methylation, histone acetylation and DNA damage can eventually lead towards apoptosis. The importance of calorie restriction therapy in the extension of lifespan has also been discussed. The role of pathways involved in the regulation of lifespan such as DAF-16/FOXO (forkhead box protein O1), TOR and JNK pathways has also been particularized. The study provides an updated account of genetic factors associated with the extended lifespan and their interactive contributory role with cellular pathways.
Collapse
Affiliation(s)
- May Nasser Bin-Jumah
- Biology Department, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh 11671, Saudi Arabia;
- Environment and Biomaterial Unit, Health Sciences Research Center, Princess Nourah Bint Abdulrahman University, Riyadh 11671, Saudi Arabia
| | - Muhammad Shahid Nadeem
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Sadaf Jamal Gilani
- Department of Basic Health Sciences, Princess Nourah Bint Abdulrahman University, Riyadh 11671, Saudi Arabia;
| | - Fahad A. Al-Abbasi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Inam Ullah
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore 54000, Pakistan;
| | - Sami I. Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka 72341, Saudi Arabia;
| | - Mohammed M. Ghoneim
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Ad Diriyah 13713, Saudi Arabia;
| | - Sultan Alshehri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Aziz Uddin
- Department of Biotechnology and Genetic Engineering, Hazara University, Mansehra 21300, Pakistan;
| | - Bibi Nazia Murtaza
- Department of Zoology, Abbottabad University of Science and Technology (AUST), Abbottabad 22310, Pakistan;
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| |
Collapse
|
26
|
Garcia AR, Finch C, Gatz M, Kraft T, Eid Rodriguez D, Cummings D, Charifson M, Buetow K, Beheim BA, Allayee H, Thomas GS, Stieglitz J, Gurven MD, Kaplan H, Trumble BC. APOE4 is associated with elevated blood lipids and lower levels of innate immune biomarkers in a tropical Amerindian subsistence population. eLife 2021; 10:68231. [PMID: 34586066 PMCID: PMC8480980 DOI: 10.7554/elife.68231] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 08/23/2021] [Indexed: 12/17/2022] Open
Abstract
In post-industrial settings, apolipoprotein E4 (APOE4) is associated with increased cardiovascular and neurological disease risk. However, the majority of human evolutionary history occurred in environments with higher pathogenic diversity and low cardiovascular risk. We hypothesize that in high-pathogen and energy-limited contexts, the APOE4 allele confers benefits by reducing innate inflammation when uninfected, while maintaining higher lipid levels that buffer costs of immune activation during infection. Among Tsimane forager-farmers of Bolivia (N = 1266, 50% female), APOE4 is associated with 30% lower C-reactive protein, and higher total cholesterol and oxidized LDL. Blood lipids were either not associated, or negatively associated with inflammatory biomarkers, except for associations of oxidized LDL and inflammation which were limited to obese adults. Further, APOE4 carriers maintain higher levels of total and LDL cholesterol at low body mass indices (BMIs). These results suggest that the relationship between APOE4 and lipids may be beneficial for pathogen-driven immune responses and unlikely to increase cardiovascular risk in an active subsistence population.
Collapse
Affiliation(s)
- Angela R Garcia
- Center for Evolution and Medicine, Arizona State University, Tempe, United States.,Department of Anthropology, Emory University, Atlanta, United States
| | - Caleb Finch
- Leonard Davis School of Gerontology, Dornsife College, University of Southern California, Los Angeles, Los Angeles, United States
| | - Margaret Gatz
- Center for Economic and Social Research, University of Southern California, Los Angeles, Los Angeles, United States
| | - Thomas Kraft
- Department of Anthropology, University of California, Santa Barbara, Santa Barbara, United States
| | | | - Daniel Cummings
- Institute for Economics and Society, Chapman University, Orange, United States
| | - Mia Charifson
- Vilcek Institute of Graduate Biomedical Sciences, New York University, New York, United States
| | - Kenneth Buetow
- Center for Evolution and Medicine, Arizona State University, Tempe, United States.,School of Life Sciences, Arizona State University, Tempe, United States
| | - Bret A Beheim
- Department of Human Behavior, Ecology and Culture, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Hooman Allayee
- Department of Preventive Medicine and Biochemistry & Molecular Medicine, Keck School of Medicine, University of Southern California, Irvine, Irvine, United States.,Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Irvine, Irvine, United States
| | - Gregory S Thomas
- Long Beach Memorial, Long Beach and University of California Irvine, Irvine, United States
| | - Jonathan Stieglitz
- Institute for Advanced Study in Toulouse, Universite Toulouse, Toulouse, France
| | - Michael D Gurven
- Department of Anthropology, University of California, Santa Barbara, Santa Barbara, United States
| | - Hillard Kaplan
- Institute for Economics and Society, Chapman University, Orange, United States
| | - Benjamin C Trumble
- School of Human Evolution and Social Change, Arizona State University, Tempe, United States
| |
Collapse
|
27
|
Carmichael J, Hicks AJ, Spitz G, Gould KR, Ponsford J. Moderators of gene-outcome associations following traumatic brain injury. Neurosci Biobehav Rev 2021; 130:107-124. [PMID: 34411558 DOI: 10.1016/j.neubiorev.2021.08.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 07/04/2021] [Accepted: 08/13/2021] [Indexed: 12/14/2022]
Abstract
The field of genomics is the principal avenue in the ongoing development of precision/personalised medicine for a variety of health conditions. However, relating genes to outcomes is notoriously complex, especially when considering that other variables can change, or moderate, gene-outcome associations. Here, we comprehensively discuss moderation of gene-outcome associations in the context of traumatic brain injury (TBI), a common, chronically debilitating, and costly neurological condition that is under complex polygenic influence. We focus our narrative review on single nucleotide polymorphisms (SNPs) of three of the most studied genes (apolipoprotein E, brain-derived neurotrophic factor, and catechol-O-methyltransferase) and on three demographic variables believed to moderate associations between these SNPs and TBI outcomes (age, biological sex, and ethnicity). We speculate on the mechanisms which may underlie these moderating effects, drawing widely from biomolecular and behavioural research (n = 175 scientific reports) within the TBI population (n = 72) and other neurological, healthy, ageing, and psychiatric populations (n = 103). We conclude with methodological recommendations for improved exploration of moderators in future genetics research in TBI and other populations.
Collapse
Affiliation(s)
- Jai Carmichael
- Monash-Epworth Rehabilitation Research Centre, Epworth HealthCare, Melbourne, Australia; Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Australia.
| | - Amelia J Hicks
- Monash-Epworth Rehabilitation Research Centre, Epworth HealthCare, Melbourne, Australia; Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Australia
| | - Gershon Spitz
- Monash-Epworth Rehabilitation Research Centre, Epworth HealthCare, Melbourne, Australia; Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Australia
| | - Kate Rachel Gould
- Monash-Epworth Rehabilitation Research Centre, Epworth HealthCare, Melbourne, Australia; Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Australia
| | - Jennie Ponsford
- Monash-Epworth Rehabilitation Research Centre, Epworth HealthCare, Melbourne, Australia; Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Australia
| |
Collapse
|
28
|
Identification of pleiotropy at the gene level between psychiatric disorders and related traits. Transl Psychiatry 2021; 11:410. [PMID: 34326310 PMCID: PMC8322263 DOI: 10.1038/s41398-021-01530-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/08/2021] [Accepted: 06/21/2021] [Indexed: 01/22/2023] Open
Abstract
Major mental disorders are highly prevalent and make a substantial contribution to the global disease burden. It is known that mental disorders share clinical characteristics, and genome-wide association studies (GWASs) have recently provided evidence for shared genetic factors as well. Genetic overlaps are usually identified at the single-marker level. Here, we aimed to identify genetic overlaps at the gene level between 7 mental disorders (schizophrenia, autism spectrum disorder, major depressive disorder, anorexia nervosa, ADHD, bipolar disorder and anxiety), 8 brain morphometric traits, 2 cognitive traits (educational attainment and general cognitive function) and 9 personality traits (subjective well-being, depressive symptoms, neuroticism, extraversion, openness to experience, agreeableness and conscientiousness, children's aggressive behaviour, loneliness) based on publicly available GWASs. We performed systematic conditional regression analyses to identify independent signals and select loci associated with more than one trait. We identified 48 genes containing independent markers associated with several traits (pleiotropy at the gene level). We also report 9 genes with different markers that show independent associations with single traits (allelic heterogeneity). This study demonstrates that mental disorders and related traits do show pleiotropy at the gene level as well as the single-marker level. The identification of these genes might be important for prioritizing further deep genotyping, functional studies, or drug targeting.
Collapse
|
29
|
Qin W, Li W, Wang Q, Gong M, Li T, Shi Y, Song Y, Li Y, Li F, Jia J. Race-Related Association between APOE Genotype and Alzheimer's Disease: A Systematic Review and Meta-Analysis. J Alzheimers Dis 2021; 83:897-906. [PMID: 34334408 DOI: 10.3233/jad-210549] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The global race-dependent association of Alzheimer's disease (AD) and apolipoprotein E (APOE) genotype is not well understood. Transethnic analysis of APOE could clarify the role of genetics in AD risk across populations. OBJECTIVE This study aims to determine how race and APOE genotype affect the risks for AD. METHODS We performed a systematic search of PubMed, Embase, Web of Science, and the Cochrane Library since 1993 to Aug 25, 2020. A total of 10,395 reports were identified, and 133 were eligible for analysis with data on 77,402 participants. Studies contained AD clinical diagnostic and APOE genotype data. Homogeneous data sets were pooled in case-control analyses. Odds ratios and 95% confidence intervals for developing AD were calculated for populations of different races and APOE genotypes. RESULTS The proportion of APOE genotypes and alleles differed between populations of different races. Results showed that APOEɛ4 was a risk factor for AD, whereas APOEɛ2 protected against it. The effects of APOEɛ4 and ɛ2 on AD risk were distinct in various races, they were substantially attenuated among Black people. Sub-group analysis found a higher frequency of APOEɛ4/ɛ4 and lower frequency of APOEɛ3/ɛ3 among early-onset AD than late-onset AD in a combined group and different races. CONCLUSION Our meta-analysis suggests that the association of APOE genotypes and AD differ between races. These results enhance our understanding of APOE-related risk for AD across race backgrounds and provide new insights into precision medicine for AD.
Collapse
Affiliation(s)
- Wei Qin
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Wenwen Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Qi Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Min Gong
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Tingting Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yuqing Shi
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yang Song
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ying Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Fangyu Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Geriatric Cognitive Disorders, Beijing, China.,Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, Beijing, China.,Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
30
|
Curtis D. Analysis of whole genome sequenced cases and controls shows that the association of variants in TOMM40, BCAM, NECTIN2 and APOC1 with late onset Alzheimer's disease is driven by linkage disequilibrium with APOE ε2/ε3/ε4 alleles. J Neurogenet 2021; 35:59-66. [PMID: 33970751 DOI: 10.1080/01677063.2020.1866569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 12/16/2020] [Indexed: 10/21/2022]
Abstract
Variants in APOE are associated with risk of late onset Alzheimer's disease (LOAD) but the magnitude of the effect has been reported to vary across ancestries. Also, other variants in the region have been reported to show association though it has been unclear whether this was secondary to their linkage disequilibrium with the APOE variants rs429358 and rs7412. Previous analyses of exome-sequenced samples have identified other genes in which rare variants impact risk of disease. In this study 2000 whole genome sequenced cases and controls with different ancestries were subjected to gene-based weighted burden analysis to identify risk genes. Additionally, individual variants in the APOE region were tested for association with LOAD. When using the APOE variants as covariates no individual genes showed statistically significant evidence for association after Bonferroni correction for multiple testing, which may well be a consequence of the modest sample size. Likewise, for those variants initially showing evidence of association with LOAD incorporating the APOE variants as covariates dramatically reduced the strength of association. These results demonstrate that the differential association of APOE across ancestries does not appear to be driven by another variant in the region. It seems likely that no other genes in the region have a direct effect on LOAD risk.
Collapse
Affiliation(s)
- David Curtis
- UCL Genetics Institute, University College London, London, UK
- Centre for Psychiatry, Queen Mary University of London, London, UK
| |
Collapse
|
31
|
Finch CE, Kulminski AM. The ApoE Locus and COVID-19: Are We Going Where We Have Been? J Gerontol A Biol Sci Med Sci 2021; 76:e1-e3. [PMID: 32777042 PMCID: PMC7454416 DOI: 10.1093/gerona/glaa200] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Indexed: 12/26/2022] Open
Affiliation(s)
- Caleb E Finch
- Center with Leonard Davis School of Gerontology and Dornsife College, University of Southern California, Los Angeles
| | - Alexander M Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina
| |
Collapse
|
32
|
Henson RN, Suri S, Knights E, Rowe JB, Kievit RA, Lyall DM, Chan D, Eising E, Fisher SE. Effect of apolipoprotein E polymorphism on cognition and brain in the Cambridge Centre for Ageing and Neuroscience cohort. Brain Neurosci Adv 2020; 4:2398212820961704. [PMID: 33088920 PMCID: PMC7545750 DOI: 10.1177/2398212820961704] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 08/27/2020] [Indexed: 01/01/2023] Open
Abstract
Polymorphisms in the apolipoprotein E (APOE) gene have been associated with individual differences in cognition, brain structure and brain function. For example, the ε4 allele has been associated with cognitive and brain impairment in old age and increased risk of dementia, while the ε2 allele has been claimed to be neuroprotective. According to the ‘antagonistic pleiotropy’ hypothesis, these polymorphisms have different effects across the lifespan, with ε4, for example, postulated to confer benefits on cognitive and brain functions earlier in life. In this stage 2 of the Registered Report – https://osf.io/bufc4, we report the results from the cognitive and brain measures in the Cambridge Centre for Ageing and Neuroscience cohort (www.cam-can.org). We investigated the antagonistic pleiotropy hypothesis by testing for allele-by-age interactions in approximately 600 people across the adult lifespan (18–88 years), on six outcome variables related to cognition, brain structure and brain function (namely, fluid intelligence, verbal memory, hippocampal grey-matter volume, mean diffusion within white matter and resting-state connectivity measured by both functional magnetic resonance imaging and magnetoencephalography). We found no evidence to support the antagonistic pleiotropy hypothesis. Indeed, Bayes factors supported the null hypothesis in all cases, except for the (linear) interaction between age and possession of the ε4 allele on fluid intelligence, for which the evidence for faster decline in older ages was ambiguous. Overall, these pre-registered analyses question the antagonistic pleiotropy of APOE polymorphisms, at least in healthy adults.
Collapse
Affiliation(s)
- Richard N Henson
- MRC Cognition & Brain Sciences Unit, University of Cambridge, Cambridge, UK.,Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Sana Suri
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, UK.,Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK
| | - Ethan Knights
- MRC Cognition & Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - James B Rowe
- MRC Cognition & Brain Sciences Unit, University of Cambridge, Cambridge, UK.,Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Rogier A Kievit
- MRC Cognition & Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - Donald M Lyall
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Dennis Chan
- Institute of Cognitive Neuroscience, University College London, London, UK
| | - Else Eising
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands
| | - Simon E Fisher
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands.,Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| |
Collapse
|
33
|
Smith CJ, Ashford JW, Perfetti TA. Putative Survival Advantages in Young Apolipoprotein ɛ4 Carriers are Associated with Increased Neural Stress. J Alzheimers Dis 2020; 68:885-923. [PMID: 30814349 PMCID: PMC6484250 DOI: 10.3233/jad-181089] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Inheritance of a single copy of the apolipoprotein E (APOE) ɛ4 allele increases risk of Alzheimer’s disease (AD) by 3-4-fold, with homozygosity associated with a 12-16-fold increase in risk, relative to ɛ3 allele homozygosity. There is a decreased risk associated with the APOE ɛ2 allele. The pathological consequence of APOE genotype has led to intense efforts to understand the mechanistic basis of the interplay between APOE status and loss of synapses. Numerous ɛ4 allele-related associations have been reported with the potential relevance of these associations to the pathogenesis of AD unknown at this time. In primarily young subjects, we have reviewed a representative body of literature on ɛ4 allele-associations related to the following: cardiovascular responses; impacts on reproduction and fetal development; co-morbidities; resistance to infectious disease; responses to head injury; biochemical differences possibly related to neural stress; and brain structure-function differences. In addition, the literature on the association between the ɛ4 allele and cognitive performance has been reviewed comprehensively. The weight-of-the-evidence supports the hypothesis that possession of the ancestral ɛ4 allele in youth is associated with improved fitness during fetal development, infancy, and youth relative to the more recently appearing ɛ3 allele, at the expense of decreased fitness in old age, which is substantially improved by the ɛ3 allele. However, possession of the ɛ4 allele is also associated with higher levels of synaptic macromolecular turnover, which likely stresses basic cellular neuroplasticity mechanisms. Clinical trials of potential AD therapeutics should consider APOE status as an enrollment criterion.
Collapse
Affiliation(s)
- Carr J Smith
- Florida State University, Department of Nurse Anesthesia, Panama City, FL, USA
| | - J Wesson Ashford
- Stanford University and VA Palo Alto Health Care System, Palo Alto, CA, USA
| | | |
Collapse
|
34
|
Choong WK, Wang JH, Sung TY. MinProtMaxVP: Generating a minimized number of protein variant sequences containing all possible variant peptides for proteogenomic analysis. J Proteomics 2020; 223:103819. [PMID: 32407886 DOI: 10.1016/j.jprot.2020.103819] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 05/04/2020] [Accepted: 05/09/2020] [Indexed: 12/12/2022]
Abstract
Identifying single-amino-acid variants (SAVs) from mass spectrometry-based experiments is critical for validating single-nucleotide variants (SNVs) at the protein level to facilitate biomedical research. Currently, two approaches are usually applied to convert SNV annotations into SAV-harboring protein sequences. One approach generates one sequence containing exactly one SAV, and the other all SAVs. However, they may neglect the possibility of SAV combinations, e.g., haplotypes, existing in bio-samples. Therefore, it is necessary to consider all SAV combinations of a protein when generating SAV-harboring protein sequences. In this paper, we propose MinProtMaxVP, a novel approach which selects a minimized number of SAV-harboring protein sequences generated from the exhaustive approach, while still accommodating all possible variant peptides, by solving a classic set covering problem. Our study on known haplotype variations of TAS2R38 justifies the necessity for MinProtMaxVP to consider all combinations of SAVs. The performance of MinProtMaxVP is demonstrated by an in silico study on OR2T27 with five SAVs and real experimental data of the HEK293 cell line. Furthermore, assuming simulated somatic and germline variants of OR2T27 in tumor and normal tissues demonstrates that when adopting the appropriate somatic and germline SAV integration strategy, MinProtMaxVP is adaptable to labeling and label-free mass spectrometry-based experiments. SIGNIFICANCE: We present MinProtMaxVP, a novel approach to generate SAV-harboring protein sequences for constructing a customized protein sequence database, which is used in database searching for variant peptide identification. This approach outperforms the existing approaches in generating all possible variant peptides to be included in protein sequences and possibly leading to identification of more variant peptides in proteogenomic analysis.
Collapse
Affiliation(s)
- Wai-Kok Choong
- Institute of Information Science, Academia Sinica, Nankang, Taipei 11529, Taiwan
| | - Jen-Hung Wang
- Institute of Information Science, Academia Sinica, Nankang, Taipei 11529, Taiwan
| | - Ting-Yi Sung
- Institute of Information Science, Academia Sinica, Nankang, Taipei 11529, Taiwan.
| |
Collapse
|
35
|
Iacono D, Feltis GC. Impact of Apolipoprotein E gene polymorphism during normal and pathological conditions of the brain across the lifespan. Aging (Albany NY) 2020; 11:787-816. [PMID: 30677746 PMCID: PMC6366964 DOI: 10.18632/aging.101757] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 01/05/2019] [Indexed: 12/12/2022]
Abstract
The central nervous system (CNS) is the cellular substrate for the integration of complex, dynamic, constant, and simultaneous interactions among endogenous and exogenous stimuli across the entire human lifespan. Numerous studies on aging-related brain diseases show that some genes identified as risk factors for some of the most common neurodegenerative diseases - such as the allele 4 of APOE gene (APOE4) for Alzheimer's disease (AD) - have a much earlier neuro-anatomical and neuro-physiological impact. The impact of APOE polymorphism appears in fact to start as early as youth and early-adult life. Intriguingly, though, those same genes associated with aging-related brain diseases seem to influence different aspects of the brain functioning much earlier actually, that is, even from the neonatal periods and earlier. The APOE4, an allele classically associated with later-life neurodegenerative disorders as AD, seems in fact to exert a series of very early effects on phenomena of neuroplasticity and synaptogenesis that begin from the earliest periods of life such as the fetal ones.We reviewed some of the findings supporting the hypothesis that APOE polymorphism is an early modifier of various neurobiological aspects across the entire human lifespan - from the in-utero to the centenarian life - during both normal and pathological conditions of the brain.
Collapse
Affiliation(s)
- Diego Iacono
- Neuropathology Research, Biomedical Research Institute of New Jersey (BRInj), Cedar Knolls, NJ 07927, USA.,MidAtlantic Neonatology Associates (MANA), Morristown, NJ 07960, USA.,Atlantic Neuroscience Institute, Atlantic Health System (AHS), Overlook Medical Center, Summit, NJ 07901, USA
| | - Gloria C Feltis
- Neuropathology Research, Biomedical Research Institute of New Jersey (BRInj), Cedar Knolls, NJ 07927, USA
| |
Collapse
|
36
|
|
37
|
Pratt J, Boreham C, Ennis S, Ryan AW, De Vito G. Genetic Associations with Aging Muscle: A Systematic Review. Cells 2019; 9:E12. [PMID: 31861518 PMCID: PMC7016601 DOI: 10.3390/cells9010012] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/17/2019] [Accepted: 12/18/2019] [Indexed: 12/24/2022] Open
Abstract
The age-related decline in skeletal muscle mass, strength and function known as 'sarcopenia' is associated with multiple adverse health outcomes, including cardiovascular disease, stroke, functional disability and mortality. While skeletal muscle properties are known to be highly heritable, evidence regarding the specific genes underpinning this heritability is currently inconclusive. This review aimed to identify genetic variants known to be associated with muscle phenotypes relevant to sarcopenia. PubMed, Embase and Web of Science were systematically searched (from January 2004 to March 2019) using pre-defined search terms such as "aging", "sarcopenia", "skeletal muscle", "muscle strength" and "genetic association". Candidate gene association studies and genome wide association studies that examined the genetic association with muscle phenotypes in non-institutionalised adults aged ≥50 years were included. Fifty-four studies were included in the final analysis. Twenty-six genes and 88 DNA polymorphisms were analysed across the 54 studies. The ACTN3, ACE and VDR genes were the most frequently studied, although the IGF1/IGFBP3, TNFα, APOE, CNTF/R and UCP2/3 genes were also shown to be significantly associated with muscle phenotypes in two or more studies. Ten DNA polymorphisms (rs154410, rs2228570, rs1800169, rs3093059, rs1800629, rs1815739, rs1799752, rs7412, rs429358 and 192 bp allele) were significantly associated with muscle phenotypes in two or more studies. Through the identification of key gene variants, this review furthers the elucidation of genetic associations with muscle phenotypes associated with sarcopenia.
Collapse
Affiliation(s)
- Jedd Pratt
- Institute for Sport and Health, University College Dublin, Dublin, Ireland; (C.B.); (G.D.V.)
- Genomics Medicine Ireland, Dublin, Ireland; (S.E.); (A.W.R.)
| | - Colin Boreham
- Institute for Sport and Health, University College Dublin, Dublin, Ireland; (C.B.); (G.D.V.)
| | - Sean Ennis
- Genomics Medicine Ireland, Dublin, Ireland; (S.E.); (A.W.R.)
- UCD ACoRD, Academic Centre on Rare Diseases, University College Dublin, Dublin, Ireland
| | - Anthony W. Ryan
- Genomics Medicine Ireland, Dublin, Ireland; (S.E.); (A.W.R.)
| | - Giuseppe De Vito
- Institute for Sport and Health, University College Dublin, Dublin, Ireland; (C.B.); (G.D.V.)
- Department of Biomedical Sciences, University of Padova, Via F. Marzolo 3, 35131 Padova, Italy
| |
Collapse
|
38
|
Blue EE, Horimoto ARVR, Mukherjee S, Wijsman EM, Thornton TA. Local ancestry at APOE modifies Alzheimer's disease risk in Caribbean Hispanics. Alzheimers Dement 2019; 15:1524-1532. [PMID: 31606368 PMCID: PMC6925639 DOI: 10.1016/j.jalz.2019.07.016] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 07/22/2019] [Accepted: 07/25/2019] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Although the relationship between APOE and Alzheimer's disease (AD) is well established in populations of European descent, the effects of APOE and ancestry on AD risk in diverse populations is not well understood. METHODS Logistic mixed model regression and survival analyses were performed in a sample of 3067 Caribbean Hispanics and 3028 individuals of European descent to assess the effects of APOE genotype, local ancestry, and genome-wide ancestry on AD risk and age at onset. RESULTS Among the Caribbean Hispanics, individuals with African-derived ancestry at APOE had 39% lower odds of AD than individuals with European-derived APOE, after adjusting for APOE genotype, age, and genome-wide ancestry. While APOE E2 and E4 effects on AD risk and age at onset were significant in the Caribbean Hispanics, they were substantially attenuated compared with those in European ancestry individuals. DISCUSSION These results suggest that additional genetic variation in the APOE region influences AD risk beyond APOE E2/E3/E4.
Collapse
Affiliation(s)
- Elizabeth E Blue
- Division of Medical Genetics, University of Washington, Seattle, WA, USA.
| | | | | | - Ellen M Wijsman
- Division of Medical Genetics, University of Washington, Seattle, WA, USA; Department of Biostatistics, University of Washington, Seattle, WA, USA; Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Timothy A Thornton
- Department of Biostatistics, University of Washington, Seattle, WA, USA; Department of Statistics, University of Washington, Seattle, WA, USA.
| |
Collapse
|
39
|
Trumble BC, Finch CE. THE EXPOSOME IN HUMAN EVOLUTION: FROM DUST TO DIESEL. THE QUARTERLY REVIEW OF BIOLOGY 2019; 94:333-394. [PMID: 32269391 PMCID: PMC7141577 DOI: 10.1086/706768] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Global exposures to air pollution and cigarette smoke are novel in human evolutionary history and are associated with about 16 million premature deaths per year. We investigate the history of the human exposome for relationships between novel environmental toxins and genetic changes during human evolution in six phases. Phase I: With increased walking on savannas, early human ancestors inhaled crustal dust, fecal aerosols, and spores; carrion scavenging introduced new infectious pathogens. Phase II: Domestic fire exposed early Homo to novel toxins from smoke and cooking. Phases III and IV: Neolithic to preindustrial Homo sapiens incurred infectious pathogens from domestic animals and dense communities with limited sanitation. Phase V: Industrialization introduced novel toxins from fossil fuels, industrial chemicals, and tobacco at the same time infectious pathogens were diminishing. Thereby, pathogen-driven causes of mortality were replaced by chronic diseases driven by sterile inflammogens, exogenous and endogenous. Phase VI: Considers future health during global warming with increased air pollution and infections. We hypothesize that adaptation to some ancient toxins persists in genetic variations associated with inflammation and longevity.
Collapse
Affiliation(s)
- Benjamin C Trumble
- School of Human Evolution & Social Change and Center for Evolution and Medicine, Arizona State University Tempe, Arizona 85287 USA
| | - Caleb E Finch
- Leonard Davis School of Gerontology and Dornsife College, University of Southern California Los Angeles, California 90089-0191 USA
| |
Collapse
|
40
|
Giuliani C, Garagnani P, Franceschi C. Genetics of Human Longevity Within an Eco-Evolutionary Nature-Nurture Framework. Circ Res 2019; 123:745-772. [PMID: 30355083 DOI: 10.1161/circresaha.118.312562] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Human longevity is a complex trait, and to disentangle its basis has a great theoretical and practical consequences for biomedicine. The genetics of human longevity is still poorly understood despite several investigations that used different strategies and protocols. Here, we argue that such rather disappointing harvest is largely because of the extraordinary complexity of the longevity phenotype in humans. The capability to reach the extreme decades of human lifespan seems to be the result of an intriguing mixture of gene-environment interactions. Accordingly, the genetics of human longevity is here described as a highly context-dependent phenomenon, within a new integrated, ecological, and evolutionary perspective, and is presented as a dynamic process, both historically and individually. The available literature has been scrutinized within this perspective, paying particular attention to factors (sex, individual biography, family, population ancestry, social structure, economic status, and education, among others) that have been relatively neglected. The strength and limitations of the most powerful and used tools, such as genome-wide association study and whole-genome sequencing, have been discussed, focusing on prominently emerged genes and regions, such as apolipoprotein E, Forkhead box O3, interleukin 6, insulin-like growth factor-1, chromosome 9p21, 5q33.3, and somatic mutations among others. The major results of this approach suggest that (1) the genetics of longevity is highly population specific; (2) small-effect alleles, pleiotropy, and the complex allele timing likely play a major role; (3) genetic risk factors are age specific and need to be integrated in the light of the geroscience perspective; (4) a close relationship between genetics of longevity and genetics of age-related diseases (especially cardiovascular diseases) do exist. Finally, the urgent need of a global approach to the largely unexplored interactions between the 3 genetics of human body, that is, nuclear, mitochondrial, and microbiomes, is stressed. We surmise that the comprehensive approach here presented will help in increasing the above-mentioned harvest.
Collapse
Affiliation(s)
- Cristina Giuliani
- From the Department of Biological, Geological, and Environmental Sciences (BiGeA), Laboratory of Molecular Anthropology and Centre for Genome Biology (C.G.), University of Bologna, Italy.,School of Anthropology and Museum Ethnography, University of Oxford, United Kingdom (C.G.).,Interdepartmental Centre 'L. Galvani' (CIG), University of Bologna, Italy (C.G.)
| | - Paolo Garagnani
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES) (P.G.), University of Bologna, Italy.,Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet at Huddinge University Hospital, Stockholm, Sweden (P.G.)
| | | |
Collapse
|
41
|
The Genetic Variability of APOE in Different Human Populations and Its Implications for Longevity. Genes (Basel) 2019; 10:genes10030222. [PMID: 30884759 PMCID: PMC6471373 DOI: 10.3390/genes10030222] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/01/2019] [Accepted: 03/12/2019] [Indexed: 12/11/2022] Open
Abstract
Human longevity is a complex phenotype resulting from the combinations of context-dependent gene-environment interactions that require analysis as a dynamic process in a cohesive ecological and evolutionary framework. Genome-wide association (GWAS) and whole-genome sequencing (WGS) studies on centenarians pointed toward the inclusion of the apolipoprotein E (APOE) polymorphisms ε2 and ε4, as implicated in the attainment of extreme longevity, which refers to their effect in age-related Alzheimer's disease (AD) and cardiovascular disease (CVD). In this case, the available literature on APOE and its involvement in longevity is described according to an anthropological and population genetics perspective. This aims to highlight the evolutionary history of this gene, how its participation in several biological pathways relates to human longevity, and which evolutionary dynamics may have shaped the distribution of APOE haplotypes across the globe. Its potential adaptive role will be described along with implications for the study of longevity in different human groups. This review also presents an updated overview of the worldwide distribution of APOE alleles based on modern day data from public databases and ancient DNA samples retrieved from literature in the attempt to understand the spatial and temporal frame in which present-day patterns of APOE variation evolved.
Collapse
|
42
|
Belloy ME, Napolioni V, Greicius MD. A Quarter Century of APOE and Alzheimer's Disease: Progress to Date and the Path Forward. Neuron 2019; 101:820-838. [PMID: 30844401 PMCID: PMC6407643 DOI: 10.1016/j.neuron.2019.01.056] [Citation(s) in RCA: 353] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 01/08/2019] [Accepted: 01/27/2019] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is considered a polygenic disorder. This view is clouded, however, by lingering uncertainty over how to treat the quasi "monogenic" role of apolipoprotein E (APOE). The APOE4 allele is not only the strongest genetic risk factor for AD, it also affects risk for cardiovascular disease, stroke, and other neurodegenerative disorders. This review, based mostly on data from human studies, ranges across a variety of APOE-related pathologies, touching on evolutionary genetics and risk mitigation by ethnicity and sex. The authors also address one of the most fundamental question pertaining to APOE4 and AD: does APOE4 increase AD risk via a loss or gain of function? The answer will be of the utmost importance in guiding future research in AD.
Collapse
Affiliation(s)
- Michaël E Belloy
- Department of Neurology and Neurological Sciences, FIND Lab, Stanford University, Stanford, CA 94304, USA
| | - Valerio Napolioni
- Department of Neurology and Neurological Sciences, FIND Lab, Stanford University, Stanford, CA 94304, USA
| | - Michael D Greicius
- Department of Neurology and Neurological Sciences, FIND Lab, Stanford University, Stanford, CA 94304, USA.
| |
Collapse
|
43
|
Menta BW, Swerdlow RH. An Integrative Overview of Non-Amyloid and Non-Tau Pathologies in Alzheimer's Disease. Neurochem Res 2019; 44:12-21. [PMID: 30084096 PMCID: PMC6347553 DOI: 10.1007/s11064-018-2603-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 07/27/2018] [Accepted: 07/30/2018] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that devastates the lives of its victims, and challenges the family members and health care infrastructures that care for them. Clinically, attempts to understand AD have focused on trying to predict the presence of, and more recently demonstrate the presence of, its characteristic amyloid plaque and neurofibrillary tangle pathologies. Fundamental research has also traditionally focused on understanding the generation, content, and pathogenicity of plaques and tangles, but in addition to this there is now an emerging independent interest in other molecular phenomena including apolipoprotein E, lipid metabolism, neuroinflammation, and mitochondrial function. While studies emphasizing the role of these phenomena have provided valuable AD insights, it is interesting that at the molecular level these entities extensively intertwine and interact. In this review, we provide a brief overview of why apolipoprotein E, lipid metabolism, neuroinflammation, and mitochondrial research have become increasingly ascendant in the AD research field, and present the case for studying these phenomena from an integrated perspective.
Collapse
Affiliation(s)
- Blaise W Menta
- University of Kansas Alzheimer's Disease Center, Fairway, KS, USA
- Neuroscience Graduate Program, University of Kansas Medical Center, Lawrence, KS, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Lawrence, KS, USA
| | - Russell H Swerdlow
- University of Kansas Alzheimer's Disease Center, Fairway, KS, USA.
- Neuroscience Graduate Program, University of Kansas Medical Center, Lawrence, KS, USA.
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Lawrence, KS, USA.
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Lawrence, KS, USA.
- Department of Neurology, University of Kansas Medical Center, Lawrence, KS, USA.
- Landon Center on Aging, MS 2012, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA.
| |
Collapse
|
44
|
de Frutos-Lucas J, López-Sanz D, Zuluaga P, Rodríguez-Rojo IC, Luna R, López ME, Delgado-Losada ML, Marcos A, Barabash A, López-Higes R, Maestú F, Fernández A. Physical activity effects on the individual alpha peak frequency of older adults with and without genetic risk factors for Alzheimer’s Disease: A MEG study. Clin Neurophysiol 2018; 129:1981-1989. [DOI: 10.1016/j.clinph.2018.06.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 05/29/2018] [Accepted: 06/25/2018] [Indexed: 11/30/2022]
|
45
|
Lee JH, Hong SM, Shin YA. Effects of exercise training on stroke risk factors, homocysteine concentration, and cognitive function according the APOE genotype in stroke patients. J Exerc Rehabil 2018; 14:267-274. [PMID: 29740562 PMCID: PMC5931164 DOI: 10.12965/jer.1836108.054] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 03/28/2018] [Indexed: 11/22/2022] Open
Abstract
The apolipoprotein E (APOE) gene has been suggested to be associated with stroke and dementia. However, the effects of exercise training on dementia according to the APOE genotype are not consistent to those reported by previous studies. Therefore, we examined the effects of exercise training on stroke risk factors including blood pressure, lipid profiles, homocysteine (Hcy) concentrations, and cognitive function according to the APOE genotype in stroke patients. We examined the stroke risk factors, Hcy, and cognitive function in 28 ischemic stroke patients before and after 6 months of exercise training. After exercise training, body weight, body mass index (BMI), and percent body fat decreased significantly in both APOE groups. According to the APOE genotype, the changes in BMI in the APOE ɛ4 group higher than those in APOE ɛ3 group significantly. Total cholesterol (TC), low-density lipoprotein (LDL)-C, triglyceride (TG), and Hcy decreased and high-density lipoprotein (HDL)-C increased significantly in the both groups. According to the APOE genotype, systolic blood pressure in the APOE ɛ4 group decreased, but in the APOE ɛ3 group increased after exercise training. TC, LDL-C, and TG in the APOE ɛ4 group decreased more extensively than those in the APOE ɛ3 group after exercise training. VO2max (maximal oxygen consumption) and cognition increased significantly in both groups. Folate acid intake also increased significantly in both groups. The APOE genotype affects variations in the risk factors of stroke after exercise training. However, the Hcy and cognitive function did not differ based on the APOE genotype.
Collapse
Affiliation(s)
- Joon-Hee Lee
- Department of Physical Education, Centre for Sport Medicine, Kyung Hee University, Seoul, Korea
| | - Sang-Min Hong
- Department of Physical Education, College of Education, Dongguk University, Seoul, Korea
| | - Yun-A Shin
- Department of Prescription & Rehabilitation of Exercise, College of Physical Exercise, and Department of Kinesiologic Medical Science, Graduate School Dankook University, Cheonan, Korea
| |
Collapse
|
46
|
Babenko VN, Afonnikov DA, Ignatieva EV, Klimov AV, Gusev FE, Rogaev EI. Haplotype analysis of APOE intragenic SNPs. BMC Neurosci 2018; 19:16. [PMID: 29745836 PMCID: PMC5998902 DOI: 10.1186/s12868-018-0413-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Background APOE ε4 allele is most common genetic risk factor for Alzheimer’s disease (AD) and cognitive decline. However, it remains poorly understood why only some carriers of APOE ε4 develop AD and how ethnic variabilities in APOE locus contribute to AD risk. Here, to address the role of APOE haplotypes, we reassessed the diversity of APOE locus in major ethnic groups and in Alzheimer’s Disease Neuroimaging Initiative (ADNI) dataset on patients with AD, and subjects with mild cognitive impairment (MCI), and control non-demented individuals. Results We performed APOE gene haplotype analysis for a short block of five SNPs across the gene using the ADNI whole genome sequencing dataset. The compilation of ADNI data with 1000 Genomes identified the APOE ε4 linked haplotypes, which appeared to be distant for the Asian, African and European populations. The common European ε4-bearing haplotype is associated with AD but not with MCI, and the Africans lack this haplotype. Haplotypic inference revealed alleles that may confer protection against AD. By assessing the DNA methylation profile of the APOE haplotypes, we found that the AD-associated haplotype features elevated APOE CpG content, implying that this locus can also be regulated by genetic-epigenetic interactions. Conclusions We showed that SNP frequency profiles within APOE locus are highly skewed to population-specific haplotypes, suggesting that the ancestral background within different sites at APOE gene may shape the disease phenotype. We propose that our results can be utilized for more specific risk assessment based on population descent of the individuals and on higher specificity of five site haplotypes associated with AD. Electronic supplementary material The online version of this article (10.1186/s12868-018-0413-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vladimir N Babenko
- The Federal Research Center Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, Center of Neurobiology and Neurogenetics, Lavrentieva str. 10, Novosibirsk, Russia, 630090. .,Novosibirsk State University, Pirogova Str, 2, Novosibirsk, Russia, 630090.
| | - Dmitry A Afonnikov
- The Federal Research Center Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, Center of Neurobiology and Neurogenetics, Lavrentieva str. 10, Novosibirsk, Russia, 630090.,Novosibirsk State University, Pirogova Str, 2, Novosibirsk, Russia, 630090
| | - Elena V Ignatieva
- The Federal Research Center Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, Center of Neurobiology and Neurogenetics, Lavrentieva str. 10, Novosibirsk, Russia, 630090.,Novosibirsk State University, Pirogova Str, 2, Novosibirsk, Russia, 630090
| | - Anton V Klimov
- The Federal Research Center Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, Center of Neurobiology and Neurogenetics, Lavrentieva str. 10, Novosibirsk, Russia, 630090.,Novosibirsk State University, Pirogova Str, 2, Novosibirsk, Russia, 630090
| | - Fedor E Gusev
- Vavilov Institute of General Genetics RAS, Gubkina str. 3, Moscow, Russia, 119991
| | - Evgeny I Rogaev
- The Federal Research Center Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, Center of Neurobiology and Neurogenetics, Lavrentieva str. 10, Novosibirsk, Russia, 630090.,Vavilov Institute of General Genetics RAS, Gubkina str. 3, Moscow, Russia, 119991.,Department of Psychiatry, University of Massachusetts Medical School, BNRI, Worcester, MA, 15604, USA.,Faculty of Biology, Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia, 119234
| |
Collapse
|
47
|
Soko ND, Masimirembwa C, Dandara C. Pharmacogenomics of Rosuvastatin: A Glocal (Global+Local) African Perspective and Expert Review on a Statin Drug. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2018; 20:498-509. [PMID: 27631189 DOI: 10.1089/omi.2016.0114] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The incidence of cardiovascular diseases (CVDs) in African populations residing in the African continent is on the rise fueled by both a steady increase in CVD risk factors and comorbidities such as human immunodeficiency virus/acquired immune deficiency syndrome (HIV/AIDS), tuberculosis, and parasitic diseases such as bilharzia. Statins are recommended together with lifestyle changes in the treatment of hypercholesterolemia and overall reduction of cardiovascular events. Rosuvastatin in particular is an attractive candidate in the management of CVDs in African populations often plagued with multimorbidities owing to both its potency and low drug-to-drug interaction potential. In this expert review, we describe the pharmacogenetics of rosuvastatin and how it may instrumentally affect the African populations. We describe polymorphisms in the candidate genes, ABCG2, SLCO1B1, CYP2C9, APOE, PCSK9, LDLR, LPA, and HMGCR, and their role in the potency and safety of rosuvastatin therapy. We report on qualitative and quantitative differences in the distribution of genetic variants that affect efficacy and toxicity of rosuvastatin. These differences are observed across world populations (Caucasian, European, and Asian) as well as within African populations. Finally, we advocate for extensive pharmacogenetic studies in African populations that take into account the genetic diversity of intra-African ethnic groups and the genetic differences between African populations and other global populations, with a collaborative and collective aim to provide effective and safe use of rosuvastatin in management of CVD in Africa. Our key thesis presented in this innovation field analysis is that rosuvastatin precision medicine can serve as a veritable Glocal (Global and Local) model to offer pharmacogenetic-guided optimal therapeutics for the public in both developing and developed regions of the world.
Collapse
Affiliation(s)
- Nyarai D Soko
- 1 Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, and University of Cape Town , Cape Town, South Africa
| | - Collen Masimirembwa
- 2 African Institute of Biomedical Science and Technology (AiBST) , Wilkins Hospital, Harare, Zimbabwe .,3 Clinical Pharmacology, Department of Medicine, University of Cape Town , Cape Town, South Africa
| | - Collet Dandara
- 1 Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, and University of Cape Town , Cape Town, South Africa
| |
Collapse
|
48
|
Esquerda-Canals G, Montoliu-Gaya L, Güell-Bosch J, Villegas S. Mouse Models of Alzheimer's Disease. J Alzheimers Dis 2018; 57:1171-1183. [PMID: 28304309 DOI: 10.3233/jad-170045] [Citation(s) in RCA: 191] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that nowadays affects more than 40 million people worldwide and it is predicted to exponentially increase in the coming decades. Because no curative treatment exists, research on the pathophysiology of the disease, as well as the testing of new drugs, are mandatory. For these purposes, animal models constitute a valuable, although perfectible tool. This review takes a tour through several aspects of mouse models of AD, such as the generation of transgenic models, the relevance of the promoter driving the expression of the transgenes, and the concrete transgenes used to simulate AD pathophysiology. Then, transgenic mouse lines harboring mutated human genes at several loci such as APP, PSEN1, APOEɛ4, and ob (leptin) are reviewed. Therefore, not only the accumulation of the Aβ peptide is emulated but also cholesterol and insulin metabolism. Further novel information about the disease will allow for the development of more accurate animal models, which in turn will undoubtedly be helpful for bringing preclinical research closer to clinical trials in humans.
Collapse
Affiliation(s)
- Gisela Esquerda-Canals
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.,Departament de Biologia Cel·lular, Fisiologia i Immunologia, Unitat de Citologia i Histologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Laia Montoliu-Gaya
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Jofre Güell-Bosch
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Sandra Villegas
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| |
Collapse
|
49
|
Genetic Risk Factors for Complex Forms of Alzheimer’s Disease. NEURODEGENER DIS 2018. [DOI: 10.1007/978-3-319-72938-1_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
50
|
Davies JMS, Cillard J, Friguet B, Cadenas E, Cadet J, Cayce R, Fishmann A, Liao D, Bulteau AL, Derbré F, Rébillard A, Burstein S, Hirsch E, Kloner RA, Jakowec M, Petzinger G, Sauce D, Sennlaub F, Limon I, Ursini F, Maiorino M, Economides C, Pike CJ, Cohen P, Salvayre AN, Halliday MR, Lundquist AJ, Jakowec NA, Mechta-Grigoriou F, Mericskay M, Mariani J, Li Z, Huang D, Grant E, Forman HJ, Finch CE, Sun PY, Pomatto LCD, Agbulut O, Warburton D, Neri C, Rouis M, Cillard P, Capeau J, Rosenbaum J, Davies KJA. The Oxygen Paradox, the French Paradox, and age-related diseases. GeroScience 2017; 39:499-550. [PMID: 29270905 PMCID: PMC5745211 DOI: 10.1007/s11357-017-0002-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 11/09/2017] [Indexed: 02/06/2023] Open
Abstract
A paradox is a seemingly absurd or impossible concept, proposition, or theory that is often difficult to understand or explain, sometimes apparently self-contradictory, and yet ultimately correct or true. How is it possible, for example, that oxygen "a toxic environmental poison" could be also indispensable for life (Beckman and Ames Physiol Rev 78(2):547-81, 1998; Stadtman and Berlett Chem Res Toxicol 10(5):485-94, 1997)?: the so-called Oxygen Paradox (Davies and Ursini 1995; Davies Biochem Soc Symp 61:1-31, 1995). How can French people apparently disregard the rule that high dietary intakes of cholesterol and saturated fats (e.g., cheese and paté) will result in an early death from cardiovascular diseases (Renaud and de Lorgeril Lancet 339(8808):1523-6, 1992; Catalgol et al. Front Pharmacol 3:141, 2012; Eisenberg et al. Nat Med 22(12):1428-1438, 2016)?: the so-called, French Paradox. Doubtless, the truth is not a duality and epistemological bias probably generates apparently self-contradictory conclusions. Perhaps nowhere in biology are there so many apparently contradictory views, and even experimental results, affecting human physiology and pathology as in the fields of free radicals and oxidative stress, antioxidants, foods and drinks, and dietary recommendations; this is particularly true when issues such as disease-susceptibility or avoidance, "healthspan," "lifespan," and ageing are involved. Consider, for example, the apparently paradoxical observation that treatment with low doses of a substance that is toxic at high concentrations may actually induce transient adaptations that protect against a subsequent exposure to the same (or similar) toxin. This particular paradox is now mechanistically explained as "Adaptive Homeostasis" (Davies Mol Asp Med 49:1-7, 2016; Pomatto et al. 2017a; Lomeli et al. Clin Sci (Lond) 131(21):2573-2599, 2017; Pomatto and Davies 2017); the non-damaging process by which an apparent toxicant can activate biological signal transduction pathways to increase expression of protective genes, by mechanisms that are completely different from those by which the same agent induces toxicity at high concentrations. In this review, we explore the influences and effects of paradoxes such as the Oxygen Paradox and the French Paradox on the etiology, progression, and outcomes of many of the major human age-related diseases, as well as the basic biological phenomenon of ageing itself.
Collapse
Affiliation(s)
- Joanna M S Davies
- The Medical Group, Internal Medicine, Rheumatology & Osteoporosis, Dermatology, Pulmonology, Ophthalmology, and Cardiology; the Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
| | - Josiane Cillard
- Lab de Biologie Cellulaire et Végétale, Faculté de Pharmacie, Université de Rennes, 35043, Rennes Cedex, France
| | - Bertrand Friguet
- Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, Sorbonne Universités, UPMC Univ Paris 06, 75005, Paris, France
- INSERM ERL U1164, 75005, Paris, France
| | - Enrique Cadenas
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
- School of Pharmacy, University of Southern California, Los Angeles, CA, 90089-9121, USA
- Department of Biochemistry & Molecular Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, 90033, USA
| | - Jean Cadet
- Département de Médecine nucléaire et Radiobiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec, J1H 5N4, Canada
| | - Rachael Cayce
- The Medical Group, Internal Medicine, Rheumatology & Osteoporosis, Dermatology, Pulmonology, Ophthalmology, and Cardiology; the Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
| | - Andrew Fishmann
- The Medical Group, Internal Medicine, Rheumatology & Osteoporosis, Dermatology, Pulmonology, Ophthalmology, and Cardiology; the Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
| | - David Liao
- The Medical Group, Internal Medicine, Rheumatology & Osteoporosis, Dermatology, Pulmonology, Ophthalmology, and Cardiology; the Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
| | - Anne-Laure Bulteau
- Institut de Génomique Fonctionnelle de Lyon,ENS de Lyon, CNRS, 69364, Lyon Cedex 07, France
| | - Frédéric Derbré
- Laboratory for Movement, Sport and Health Sciences-EA 1274, M2S, Université de Rennes 2-ENS, Bruz, 35170, Rennes, France
| | - Amélie Rébillard
- Laboratory for Movement, Sport and Health Sciences-EA 1274, M2S, Université de Rennes 2-ENS, Bruz, 35170, Rennes, France
| | - Steven Burstein
- The Medical Group, Internal Medicine, Rheumatology & Osteoporosis, Dermatology, Pulmonology, Ophthalmology, and Cardiology; the Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
| | - Etienne Hirsch
- INSERM UMR 1127-CNRS UMR 7225, Institut du cerveau et de la moelle épinière-ICM Thérapeutique Expérimentale de la Maladie de Parkinson, Université Pierre et Marie Curie, 75651, Paris Cedex 13, France
| | - Robert A Kloner
- Huntington Medical Research Institutes, Pasadena, CA, 91105, USA
| | - Michael Jakowec
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Giselle Petzinger
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Delphine Sauce
- Chronic infections and Immune ageing, INSERM U1135, Hopital Pitie-Salpetriere, Pierre et Marie Curie University, 75013, Paris, France
| | | | - Isabelle Limon
- Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, Sorbonne Universités, UPMC Univ Paris 06, 75005, Paris, France
| | - Fulvio Ursini
- Department of Molecular Medicine, University of Padova, 35121, Padova, Italy
| | - Matilde Maiorino
- Department of Molecular Medicine, University of Padova, 35121, Padova, Italy
| | - Christina Economides
- Los Angeles Cardiology Associates, Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
| | - Christian J Pike
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
- Division of Neurobiology, Department of Biological Sciences of the Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, 90089-0191, USA
| | - Pinchas Cohen
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
- Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, 90033, USA
| | - Anne Negre Salvayre
- Lipid peroxidation, Signalling and Vascular Diseases INSERM U1048, 31432, Toulouse Cedex 4, France
| | - Matthew R Halliday
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Adam J Lundquist
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Nicolaus A Jakowec
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | | | - Mathias Mericskay
- Laboratoire de Signalisation et Physiopathologie Cardiovasculaire-Inserm UMR-S 1180, Faculté de Pharmacie, Université Paris-Sud, 92296 Châtenay-Malabry, Paris, France
| | - Jean Mariani
- Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, Sorbonne Universités, UPMC Univ Paris 06, 75005, Paris, France
| | - Zhenlin Li
- Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, Sorbonne Universités, UPMC Univ Paris 06, 75005, Paris, France
- INSERM ERL U1164, 75005, Paris, France
| | - David Huang
- Department of Radiation Oncology, Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
| | - Ellsworth Grant
- Department of Oncology & Hematology, Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
| | - Henry J Forman
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
| | - Caleb E Finch
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
- Los Angeles Cardiology Associates, Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
- Division of Molecular & Computational Biology, Department of Biological Sciences of the Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, 90089-0191, USA
| | - Patrick Y Sun
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
- Division of Molecular & Computational Biology, Department of Biological Sciences of the Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, 90089-0191, USA
| | - Laura C D Pomatto
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
- Division of Molecular & Computational Biology, Department of Biological Sciences of the Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, 90089-0191, USA
| | - Onnik Agbulut
- Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, Sorbonne Universités, UPMC Univ Paris 06, 75005, Paris, France
| | - David Warburton
- Children's Hospital of Los Angeles, Developmental Biology, Regenerative Medicine and Stem Cell Therapeutics program and the Center for Environmental Impact on Global Health Across the Lifespan at The Saban Research Institute, Los Angeles, CA, 90027, USA
- Department of Pediatrics, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, 90033, USA
| | - Christian Neri
- Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, Sorbonne Universités, UPMC Univ Paris 06, 75005, Paris, France
| | - Mustapha Rouis
- Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, Sorbonne Universités, UPMC Univ Paris 06, 75005, Paris, France
- INSERM ERL U1164, 75005, Paris, France
| | - Pierre Cillard
- Lab de Biologie Cellulaire et Végétale, Faculté de Pharmacie, Université de Rennes, 35043, Rennes Cedex, France
| | - Jacqueline Capeau
- DR Saint-Antoine UMR_S938, UPMC, Inserm Faculté de Médecine, Université Pierre et Marie Curie, 75012, Paris, France
| | - Jean Rosenbaum
- Scientific Service of the Embassy of France in the USA, Consulate General of France in Los Angeles, Los Angeles, CA, 90025, USA
| | - Kelvin J A Davies
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA.
- Department of Biochemistry & Molecular Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, 90033, USA.
- Division of Molecular & Computational Biology, Department of Biological Sciences of the Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, 90089-0191, USA.
| |
Collapse
|