1
|
Wang Y, Zhou Z, Broder JC, Woods RL, Orchard SG, Wolfe R, Ernst EJ, Ryan J, Ernst ME, Chan AT. Antibiotic Use and Subsequent Cognitive Decline and Dementia Risk in Healthy Older Adults. Neurology 2025; 104:e210129. [PMID: 39693592 DOI: 10.1212/wnl.0000000000210129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/25/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Antibiotics rapidly reduce intestinal bacterial diversity, leading to dysbiosis that persists for months to years. Although emerging evidence from retrospective and claims-based studies has linked dysbiosis to cognitive function, prospective data are lacking. We aim to examine the prospective association of antibiotics with cognitive aging among initially healthy older adults. METHODS We leveraged data from prospective follow-up and observational extension of ASPirin in Reducing Events in the Elderly, a completed randomized trial of community-based Australian older adults. Among participants whose prescription records were available and without dementia during the first 2 years of follow-up, we identified any or repeated antibiotic use based on the Anatomical Therapeutic Chemical code (J01). We assessed the associations of antibiotic use during the first 2 years with longitudinal changes in standardized composite and domain-specific cognitive scores (global cognition, episodic memory, language and executive function, and psychomotor speed) using linear mixed models, and with incident, clinically adjudicated dementia (Diagnostic and Statistical Manual for Mental Disorders, Fourth Edition criteria) and incident cognitive impairment, no dementia (CIND, without a dementia trigger but with significant, nontransient decline), using Cox proportional hazard models. RESULTS Over a median of 4.7 years after the second follow-up visit, we documented 461 dementia and 2,576 CIND cases among 13,571 participants (mean age ± SD 75.0 ± 4.1 years, 54.3% female). Compared with nonuse, antibiotic use was not associated with increased risks for dementia (hazard ratio [HR] 1.03; 95% CI 0.84-1.25), CIND (HR 1.02; 95% CI 0.94-1.11), or subsequent declines in cognitive scores, after adjusting for sociodemographic, lifestyle factors, family history of dementia, baseline cognitive function, and medications known to affect cognition. There were also no associations according to the cumulative frequency of antibiotic use, long-term use, specific antibiotic classes (e.g., beta-lactams, tetracyclines, and sulfonamides), and subgroups defined by risk factors. DISCUSSION Among initially healthy older adults, any or repeated antibiotic use was not associated with incident dementia, CIND, or accelerated cognitive decline. Although prescription data may not reflect the actual use, we examined the frequency of antibiotics within a defined period to capture the extent and duration of antibiotic exposure. Our results do not support an association between antibiotic-associated gut microbiome disruption and dementia risk.
Collapse
Affiliation(s)
- Yiqing Wang
- From the Clinical and Translational Epidemiology Unit (Y.W., A.T.C.), and Division of Gastroenterology (Y.W., A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston; School of Public Health and Preventive Medicine (Z.Z., J.C.B., R.L.W., S.G.O., R.W., J.R.), Monash University, Melbourne; Menzies Institute for Medical Research (Z.Z.), University of Tasmania, Australia; Department of Pharmacy Practice and Science (E.J.E.. M.E.E.), College of Pharmacy, and Department of Family Medicine (M.E.E.), Carver College of Medicine, University of Iowa, Iowa City; Department of Immunology and Infectious Diseases (A.T.C.), Harvard T.H. Chan School of Public Health, Boston; and Cancer Center (A.T.C.), Massachusetts General Hospital, Boston, MA
| | - Zhen Zhou
- From the Clinical and Translational Epidemiology Unit (Y.W., A.T.C.), and Division of Gastroenterology (Y.W., A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston; School of Public Health and Preventive Medicine (Z.Z., J.C.B., R.L.W., S.G.O., R.W., J.R.), Monash University, Melbourne; Menzies Institute for Medical Research (Z.Z.), University of Tasmania, Australia; Department of Pharmacy Practice and Science (E.J.E.. M.E.E.), College of Pharmacy, and Department of Family Medicine (M.E.E.), Carver College of Medicine, University of Iowa, Iowa City; Department of Immunology and Infectious Diseases (A.T.C.), Harvard T.H. Chan School of Public Health, Boston; and Cancer Center (A.T.C.), Massachusetts General Hospital, Boston, MA
| | - Jonathan C Broder
- From the Clinical and Translational Epidemiology Unit (Y.W., A.T.C.), and Division of Gastroenterology (Y.W., A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston; School of Public Health and Preventive Medicine (Z.Z., J.C.B., R.L.W., S.G.O., R.W., J.R.), Monash University, Melbourne; Menzies Institute for Medical Research (Z.Z.), University of Tasmania, Australia; Department of Pharmacy Practice and Science (E.J.E.. M.E.E.), College of Pharmacy, and Department of Family Medicine (M.E.E.), Carver College of Medicine, University of Iowa, Iowa City; Department of Immunology and Infectious Diseases (A.T.C.), Harvard T.H. Chan School of Public Health, Boston; and Cancer Center (A.T.C.), Massachusetts General Hospital, Boston, MA
| | - Robyn L Woods
- From the Clinical and Translational Epidemiology Unit (Y.W., A.T.C.), and Division of Gastroenterology (Y.W., A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston; School of Public Health and Preventive Medicine (Z.Z., J.C.B., R.L.W., S.G.O., R.W., J.R.), Monash University, Melbourne; Menzies Institute for Medical Research (Z.Z.), University of Tasmania, Australia; Department of Pharmacy Practice and Science (E.J.E.. M.E.E.), College of Pharmacy, and Department of Family Medicine (M.E.E.), Carver College of Medicine, University of Iowa, Iowa City; Department of Immunology and Infectious Diseases (A.T.C.), Harvard T.H. Chan School of Public Health, Boston; and Cancer Center (A.T.C.), Massachusetts General Hospital, Boston, MA
| | - Suzanne Gaye Orchard
- From the Clinical and Translational Epidemiology Unit (Y.W., A.T.C.), and Division of Gastroenterology (Y.W., A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston; School of Public Health and Preventive Medicine (Z.Z., J.C.B., R.L.W., S.G.O., R.W., J.R.), Monash University, Melbourne; Menzies Institute for Medical Research (Z.Z.), University of Tasmania, Australia; Department of Pharmacy Practice and Science (E.J.E.. M.E.E.), College of Pharmacy, and Department of Family Medicine (M.E.E.), Carver College of Medicine, University of Iowa, Iowa City; Department of Immunology and Infectious Diseases (A.T.C.), Harvard T.H. Chan School of Public Health, Boston; and Cancer Center (A.T.C.), Massachusetts General Hospital, Boston, MA
| | - Rory Wolfe
- From the Clinical and Translational Epidemiology Unit (Y.W., A.T.C.), and Division of Gastroenterology (Y.W., A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston; School of Public Health and Preventive Medicine (Z.Z., J.C.B., R.L.W., S.G.O., R.W., J.R.), Monash University, Melbourne; Menzies Institute for Medical Research (Z.Z.), University of Tasmania, Australia; Department of Pharmacy Practice and Science (E.J.E.. M.E.E.), College of Pharmacy, and Department of Family Medicine (M.E.E.), Carver College of Medicine, University of Iowa, Iowa City; Department of Immunology and Infectious Diseases (A.T.C.), Harvard T.H. Chan School of Public Health, Boston; and Cancer Center (A.T.C.), Massachusetts General Hospital, Boston, MA
| | - Erika J Ernst
- From the Clinical and Translational Epidemiology Unit (Y.W., A.T.C.), and Division of Gastroenterology (Y.W., A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston; School of Public Health and Preventive Medicine (Z.Z., J.C.B., R.L.W., S.G.O., R.W., J.R.), Monash University, Melbourne; Menzies Institute for Medical Research (Z.Z.), University of Tasmania, Australia; Department of Pharmacy Practice and Science (E.J.E.. M.E.E.), College of Pharmacy, and Department of Family Medicine (M.E.E.), Carver College of Medicine, University of Iowa, Iowa City; Department of Immunology and Infectious Diseases (A.T.C.), Harvard T.H. Chan School of Public Health, Boston; and Cancer Center (A.T.C.), Massachusetts General Hospital, Boston, MA
| | - Joanne Ryan
- From the Clinical and Translational Epidemiology Unit (Y.W., A.T.C.), and Division of Gastroenterology (Y.W., A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston; School of Public Health and Preventive Medicine (Z.Z., J.C.B., R.L.W., S.G.O., R.W., J.R.), Monash University, Melbourne; Menzies Institute for Medical Research (Z.Z.), University of Tasmania, Australia; Department of Pharmacy Practice and Science (E.J.E.. M.E.E.), College of Pharmacy, and Department of Family Medicine (M.E.E.), Carver College of Medicine, University of Iowa, Iowa City; Department of Immunology and Infectious Diseases (A.T.C.), Harvard T.H. Chan School of Public Health, Boston; and Cancer Center (A.T.C.), Massachusetts General Hospital, Boston, MA
| | - Michael E Ernst
- From the Clinical and Translational Epidemiology Unit (Y.W., A.T.C.), and Division of Gastroenterology (Y.W., A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston; School of Public Health and Preventive Medicine (Z.Z., J.C.B., R.L.W., S.G.O., R.W., J.R.), Monash University, Melbourne; Menzies Institute for Medical Research (Z.Z.), University of Tasmania, Australia; Department of Pharmacy Practice and Science (E.J.E.. M.E.E.), College of Pharmacy, and Department of Family Medicine (M.E.E.), Carver College of Medicine, University of Iowa, Iowa City; Department of Immunology and Infectious Diseases (A.T.C.), Harvard T.H. Chan School of Public Health, Boston; and Cancer Center (A.T.C.), Massachusetts General Hospital, Boston, MA
| | - Andrew T Chan
- From the Clinical and Translational Epidemiology Unit (Y.W., A.T.C.), and Division of Gastroenterology (Y.W., A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston; School of Public Health and Preventive Medicine (Z.Z., J.C.B., R.L.W., S.G.O., R.W., J.R.), Monash University, Melbourne; Menzies Institute for Medical Research (Z.Z.), University of Tasmania, Australia; Department of Pharmacy Practice and Science (E.J.E.. M.E.E.), College of Pharmacy, and Department of Family Medicine (M.E.E.), Carver College of Medicine, University of Iowa, Iowa City; Department of Immunology and Infectious Diseases (A.T.C.), Harvard T.H. Chan School of Public Health, Boston; and Cancer Center (A.T.C.), Massachusetts General Hospital, Boston, MA
| |
Collapse
|
2
|
Zhao X, Wu X, Ma T, Xiao J, Chen X, Tang M, Zhang L, Zhang T, Fan M, Liao J, Zhang B, Jiang X, Li J. Late-life physical activity, midlife-to-late-life activity patterns, APOE ε4 genotype, and cognitive impairment among Chinese older adults: a population-based observational study. Int J Behav Nutr Phys Act 2025; 22:5. [PMID: 39789564 PMCID: PMC11720804 DOI: 10.1186/s12966-024-01691-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/03/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Although physical activity (PA) has been linked to cognitive health, the nuanced relationships between different dimensions of PA and cognitive impairment remain inconclusive. This study investigated associations between late-life PA levels, midlife-to-late-life activity patterns, and cognitive impairment in Chinese older adults, considering potential moderation by apolipoprotein E (APOE) ε4 genotype. METHODS We analyzed baseline data from 6,899 participants (median age 68 years, 55.78% female) in the West China Health and Aging Cohort study, with 6,575 participants having APOE genotyping data. Late-life PA and midlife-to-late-life activity patterns were assessed using the Global Physical Activity Questionnaire and a standardized question, respectively. Cognitive function was evaluated using the Chinese version of Mini-Mental State Examination. Logistic regression models were used to examine associations. RESULTS Compared to low PA level, moderate (odds ratio [OR] = 0.74, 95% confidence interval [CI] = 0.55 ~ 0.99) and high PA levels (OR = 0.60, 95%CI = 0.48 ~ 0.75) were associated with lower risk of cognitive impairment. Engaging in work-, transport-, recreation-related, and moderate-intensity PA were each significantly associated with lower cognitive impairment risk. Maintaining activity levels from midlife to late life was associated with lower cognitive impairment risk compared to decreasing levels (OR = 0.75, 95%CI = 0.60 ~ 0.94). These associations were more pronounced in APOE ε4 non-carriers, with an interaction observed between APOE ε4 genotype and recreation-related PA (P-value = 0.04). CONCLUSIONS Our findings underscore the multifaceted benefits of PA in mitigating cognitive impairment risk among older Chinese adults. Public health strategies should focus on promoting overall late-life PA levels, especially moderate-intensity PA, and maintaining activity levels comparable to midlife, with potential for personalized interventions based on genetic risk profiles.
Collapse
Affiliation(s)
- Xunying Zhao
- Department of Epidemiology and Health Statistics and West China Institute of Preventive and Medical Integration for Major Diseases, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xueyao Wu
- Department of Epidemiology and Health Statistics and West China Institute of Preventive and Medical Integration for Major Diseases, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tianpei Ma
- Department of Epidemiology and Health Statistics and West China Institute of Preventive and Medical Integration for Major Diseases, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jinyu Xiao
- Department of Epidemiology and Health Statistics and West China Institute of Preventive and Medical Integration for Major Diseases, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xin Chen
- Department of Epidemiology and Health Statistics and West China Institute of Preventive and Medical Integration for Major Diseases, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Mingshuang Tang
- Department of Epidemiology and Health Statistics and West China Institute of Preventive and Medical Integration for Major Diseases, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Li Zhang
- Department of Epidemiology and Health Statistics and West China Institute of Preventive and Medical Integration for Major Diseases, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tao Zhang
- Department of Epidemiology and Health Statistics and West China Institute of Preventive and Medical Integration for Major Diseases, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Mengyu Fan
- Department of Epidemiology and Health Statistics and West China Institute of Preventive and Medical Integration for Major Diseases, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiaqiang Liao
- Department of Epidemiology and Health Statistics and West China Institute of Preventive and Medical Integration for Major Diseases, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ben Zhang
- Department of Epidemiology and Health Statistics and West China Institute of Preventive and Medical Integration for Major Diseases, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
- Hainan General Hospital and Hainan Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Xia Jiang
- Department of Epidemiology and Health Statistics and West China Institute of Preventive and Medical Integration for Major Diseases, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China.
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Solna, Stockholm, Sweden.
| | - Jiayuan Li
- Department of Epidemiology and Health Statistics and West China Institute of Preventive and Medical Integration for Major Diseases, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
3
|
Moore A, Ritchie MD. Is the Relationship Between Cardiovascular Disease and Alzheimer's Disease Genetic? A Scoping Review. Genes (Basel) 2024; 15:1509. [PMID: 39766777 PMCID: PMC11675426 DOI: 10.3390/genes15121509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES Cardiovascular disease (CVD) and Alzheimer's disease (AD) are two diseases highly prevalent in the aging population and often co-occur. The exact relationship between the two diseases is uncertain, though epidemiological studies have demonstrated that CVDs appear to increase the risk of AD and vice versa. This scoping review aims to examine the current identified overlapping genetics between CVDs and AD at the individual gene level and at the shared pathway level. METHODS Following PRISMA-ScR guidelines for a scoping review, we searched the PubMed and Scopus databases from 1990 to October 2024 for articles that involved (1) CVDs, (2) AD, and (3) used statistical methods to parse genetic relationships. RESULTS Our search yielded 2918 articles, of which 274 articles passed screening and were organized into two main sections: (1) evidence of shared genetic risk; and (2) shared mechanisms. The genes APOE, PSEN1, and PSEN2 reportedly have wide effects across the AD and CVD spectrum, affecting both cardiac and brain tissues. Mechanistically, changes in three main pathways (lipid metabolism, blood pressure regulation, and the breakdown of the blood-brain barrier (BBB)) contribute to subclinical and etiological changes that promote both AD and CVD progression. However, genetic studies continue to be limited by the availability of longitudinal data and lack of cohorts that are representative of diverse populations. CONCLUSIONS Highly penetrant familial genes simultaneously increase the risk of CVDs and AD. However, in most cases, sets of dysregulated genes within larger-scale mechanisms, like changes in lipid metabolism, blood pressure regulation, and BBB breakdown, increase the risk of both AD and CVDs and contribute to disease progression.
Collapse
Affiliation(s)
- Anni Moore
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Marylyn D. Ritchie
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
- Division of Informatics, Department of Biostatistics, Epidemiology & Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
4
|
Soldan A, Wang J, Pettigrew C, Davatzikos C, Erus G, Hohman TJ, Dumitrescu L, Bilgel M, Resnick SM, Rivera-Rivera LA, Langhough R, Johnson SC, Benzinger T, Morris JC, Laws SM, Fripp J, Masters CL, Albert MS. Alzheimer's disease genetic risk and changes in brain atrophy and white matter hyperintensities in cognitively unimpaired adults. Brain Commun 2024; 6:fcae276. [PMID: 39229494 PMCID: PMC11369827 DOI: 10.1093/braincomms/fcae276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 06/25/2024] [Accepted: 08/12/2024] [Indexed: 09/05/2024] Open
Abstract
Reduced brain volumes and more prominent white matter hyperintensities on MRI scans are commonly observed among older adults without cognitive impairment. However, it remains unclear whether rates of change in these measures among cognitively normal adults differ as a function of genetic risk for late-onset Alzheimer's disease, including APOE-ɛ4, APOE-ɛ2 and Alzheimer's disease polygenic risk scores (AD-PRS), and whether these relationships are influenced by other variables. This longitudinal study examined the trajectories of regional brain volumes and white matter hyperintensities in relationship to APOE genotypes (N = 1541) and AD-PRS (N = 1093) in a harmonized dataset of middle-aged and older individuals with normal cognition at baseline (mean baseline age = 66 years, SD = 9.6) and an average of 5.3 years of MRI follow-up (max = 24 years). Atrophy on volumetric MRI scans was quantified in three ways: (i) a composite score of regions vulnerable to Alzheimer's disease (SPARE-AD); (ii) hippocampal volume; and (iii) a composite score of regions indexing advanced non-Alzheimer's disease-related brain aging (SPARE-BA). Global white matter hyperintensity volumes were derived from fluid attenuated inversion recovery (FLAIR) MRI. Using linear mixed effects models, there was an APOE-ɛ4 gene-dose effect on atrophy in the SPARE-AD composite and hippocampus, with greatest atrophy among ɛ4/ɛ4 carriers, followed by ɛ4 heterozygouts, and lowest among ɛ3 homozygouts and ɛ2/ɛ2 and ɛ2/ɛ3 carriers, who did not differ from one another. The negative associations of APOE-ɛ4 with atrophy were reduced among those with higher education (P < 0.04) and younger baseline ages (P < 0.03). Higher AD-PRS were also associated with greater atrophy in SPARE-AD (P = 0.035) and the hippocampus (P = 0.014), independent of APOE-ɛ4 status. APOE-ɛ2 status (ɛ2/ɛ2 and ɛ2/ɛ3 combined) was not related to baseline levels or atrophy in SPARE-AD, SPARE-BA or the hippocampus, but was related to greater increases in white matter hyperintensities (P = 0.014). Additionally, there was an APOE-ɛ4 × AD-PRS interaction in relation to white matter hyperintensities (P = 0.038), with greater increases in white matter hyperintensities among APOE-ɛ4 carriers with higher AD-PRS. APOE and AD-PRS associations with MRI measures did not differ by sex. These results suggest that APOE-ɛ4 and AD-PRS independently and additively influence longitudinal declines in brain volumes sensitive to Alzheimer's disease and synergistically increase white matter hyperintensity accumulation among cognitively normal individuals. Conversely, APOE-ɛ2 primarily influences white matter hyperintensity accumulation, not brain atrophy. Results are consistent with the view that genetic factors for Alzheimer's disease influence atrophy in a regionally specific manner, likely reflecting preclinical neurodegeneration, and that Alzheimer's disease risk genes contribute to white matter hyperintensity formation.
Collapse
Affiliation(s)
- Anja Soldan
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jiangxia Wang
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Corinne Pettigrew
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Christos Davatzikos
- Centre for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Guray Erus
- Centre for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Timothy J Hohman
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37212, USA
| | - Logan Dumitrescu
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37212, USA
| | - Murat Bilgel
- Laboratory of Behavioral Neuroscience, National Institute on Aging Intramural Research Program, Baltimore, MD 21224, USA
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging Intramural Research Program, Baltimore, MD 21224, USA
| | - Leonardo A Rivera-Rivera
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53726, USA
| | - Rebecca Langhough
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53726, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53726, USA
| | - Tammie Benzinger
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John C Morris
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Simon M Laws
- Centre for Precision Health, Edith Cowan University, Joondalup, WA 6027, Australia
| | - Jurgen Fripp
- Australian E-Health Research Centre, CSIRO Health & Biosecurity, Herston, QLD 4029, Australia
| | - Colin L Masters
- The Florey Institute, University of Melbourne, Parkville, VIC 3052, Australia
| | - Marilyn S Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
5
|
Huq A, Thompson B, Winship I. Clinical application of whole genome sequencing in young onset dementia: challenges and opportunities. Expert Rev Mol Diagn 2024; 24:659-675. [PMID: 39135326 DOI: 10.1080/14737159.2024.2388765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 08/01/2024] [Indexed: 08/30/2024]
Abstract
INTRODUCTION Young onset dementia (YOD) by its nature is difficult to diagnose. Despite involvement of multidisciplinary neurogenetics services, patients with YOD and their families face significant diagnostic delays. Genetic testing for people with YOD currently involves a staggered, iterative approach. There is currently no optimal single genetic investigation that simultaneously identifies the different genetic variants resulting in YOD. AREAS COVERED This review discusses the advances in clinical genomic testing for people with YOD. Whole genome sequencing (WGS) can be employed as a 'one stop shop' genomic test for YOD. In addition to single nucleotide variants, WGS can reliably detect structural variants, short tandem repeat expansions, mitochondrial genetic variants as well as capture single nucleotide polymorphisms for the calculation of polygenic risk scores. EXPERT OPINION WGS, when used as the initial genetic test, can enhance the likelihood of a precision diagnosis and curtail the time taken to reach this. Finding a clinical diagnosis using WGS can reduce invasive and expensive investigations and could be cost effective. These advances need to be balanced against the limitations of the technology and the genetic counseling needs for these vulnerable patients and their families.
Collapse
Affiliation(s)
- Aamira Huq
- Department of Genomic Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
- Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - Bryony Thompson
- Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
- Department of Pathology, University of Melbourne, Parkville, Victoria, Australia
| | - Ingrid Winship
- Department of Genomic Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
- Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
6
|
Ogonowski NS, García-Marín LM, Fernando AS, Flores-Ocampo V, Rentería ME. Impact of genetic predisposition to late-onset neurodegenerative diseases on early life outcomes and brain structure. Transl Psychiatry 2024; 14:185. [PMID: 38605018 PMCID: PMC11009228 DOI: 10.1038/s41398-024-02898-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 03/27/2024] [Accepted: 04/03/2024] [Indexed: 04/13/2024] Open
Abstract
Most patients with late-onset neurodegenerative diseases such as Alzheimer's and Parkinson's have a complex aetiology resulting from numerous genetic risk variants of small effects located across the genome, environmental factors, and the interaction between genes and environment. Over the last decade, genome-wide association studies (GWAS) and post-GWAS analyses have shed light on the polygenic architecture of these diseases, enabling polygenic risk scores (PRS) to estimate an individual's relative genetic liability for presenting with the disease. PRS can screen and stratify individuals based on their genetic risk, potentially years or even decades before the onset of clinical symptoms. An emerging body of evidence from various research studies suggests that genetic susceptibility to late-onset neurodegenerative diseases might impact early life outcomes, including cognitive function, brain structure and function, and behaviour. This article summarises recent findings exploring the potential impact of genetic susceptibility to neurodegenerative diseases on early life outcomes. A better understanding of the impact of genetic susceptibility to neurodegenerative diseases early in life could be valuable in disease screening, detection, and prevention and in informing treatment strategies before significant neural damage has occurred. However, ongoing studies have limitations. Overall, our review found several studies focused on APOE haplotypes and Alzheimer's risk, but a limited number of studies leveraging polygenic risk scores or focused on genetic susceptibility to other late-onset conditions.
Collapse
Affiliation(s)
- Natalia S Ogonowski
- Mental Health & Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Luis M García-Marín
- Mental Health & Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Amali S Fernando
- Mental Health & Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Victor Flores-Ocampo
- Mental Health & Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Miguel E Rentería
- Mental Health & Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
7
|
Xu Y, Sun Z, Jonaitis E, Deming Y, Lu Q, Johnson SC, Engelman CD. Apolipoprotein E moderates the association between non-APOE polygenic risk score for Alzheimer's disease and aging on preclinical cognitive function. Alzheimers Dement 2024; 20:1063-1075. [PMID: 37858606 PMCID: PMC10916952 DOI: 10.1002/alz.13515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/01/2023] [Accepted: 09/25/2023] [Indexed: 10/21/2023]
Abstract
INTRODUCTION Variation in preclinical cognitive decline suggests additional genetic factors related to Alzheimer's disease (eg, a non-APOE polygenic risk score [PRS]) may interact with the APOE ε4 allele to influence cognitive decline. METHODS We tested the PRS × APOE ε4 × age interaction on preclinical cognition using longitudinal data from the Wisconsin Registry for Alzheimer's Prevention. All analyses were fitted using a linear mixed-effects model and adjusted for within individual/family correlation among 1190 individuals. RESULTS We found statistically significant PRS × APOE ε4 × age interactions on immediate learning (P = 0.038), delayed recall (P < 0.001), and Preclinical Alzheimer's Cognitive Composite 3 score (P = 0.026). PRS-related differences in overall and memory-related cognitive domains between people with and without APOE ε4 emerge around age 70, with a much stronger adverse PRS effect among APOE ε4 carriers. The findings were replicated in a population-based cohort. DISCUSSIONS APOE ε4 can modify the association between PRS and cognition decline. HIGHLIGHTS APOE ε4 can modify the association between polygenic risk scores (PRSs) and longitudinal cognition decline, with the modifying effects more pronounced when the PRS is constructed using a conservative P threshold (eg, P < 5e-8 ). The adverse genetic effect caused by the combined effect of the currently known genetic variants is more detrimental among APOE ε4 carriers around age 70. Individuals who are APOE ε4 carriers with high PRSs are the most vulnerable to the harmful effects caused by genetic burden.
Collapse
Affiliation(s)
- Yuexuan Xu
- Department of Population Health ScienceSchool of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Zhongxuan Sun
- Department of Biostatistics and Medical InformaticsSchool of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Erin Jonaitis
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Yuetiva Deming
- Department of Population Health ScienceSchool of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Department of MedicineSchool of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Qiongshi Lu
- Department of Biostatistics and Medical InformaticsSchool of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Sterling C. Johnson
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Department of MedicineSchool of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Corinne D. Engelman
- Department of Population Health ScienceSchool of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| |
Collapse
|
8
|
Zheng HT, Wu Z, Mielke MM, Murray AM, Ryan J. Plasma Biomarkers of Alzheimer's Disease and Neurodegeneration According to Sociodemographic Characteristics and Chronic Health Conditions. J Prev Alzheimers Dis 2024; 11:1189-1197. [PMID: 39350363 PMCID: PMC11436401 DOI: 10.14283/jpad.2024.142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 06/23/2024] [Indexed: 10/04/2024]
Abstract
Ultrasensitive assays have been developed which enable biomarkers of Alzheimer's disease pathology and neurodegeneration to be measured in blood. These biomarkers can aid in diagnosis, and have been used to predict risk of cognitive decline and Alzheimer's disease. The ease and cost-effectiveness of blood collections means that these biomarkers could be applied more broadly in population-based screening, however it is critical to first understand what other factors could affect blood biomarker levels. The aim of this review was to determine the extent that sociodemographic, lifestyle and health factors have been associated with blood biomarkers of Alzheimer's disease and neuropathology. Of the 32 studies included in this review, all but one measured biomarker levels in plasma, and age and sex were the most commonly investigated factors. The most consistent significant findings were a positive association between age and neurofilament light chain (NfL) and glial fibrillary acidic protein (GFAP), and females had higher GFAP than men. Apolipoprotein ε4 allele carriers had lower Aβ42 and Aβ42/40 ratio. Body mass index was negatively associated with GFAP and NfL, and chronic kidney disease with higher levels of all biomarkers. Too few studies have investigated other chronic health conditions and this requires further investigation. Given the potential for plasma biomarkers to enhance Alzheimer's disease diagnosis in primary care, it is important to understand how to interpret the biomarkers in light of factors that physiologically impact blood biomarker levels. This information will be critical for the establishment of reference ranges and thus the correct interpretation of these biomarkers in clinical screening.
Collapse
Affiliation(s)
- H T Zheng
- Joanne Ryan, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia,
| | | | | | | | | |
Collapse
|
9
|
Najar J, Thorvaldsson V, Kern S, Skoog J, Waern M, Zetterberg H, Blennow K, Skoog I, Zettergren A. Polygenic risk scores for Alzheimer's disease in relation to cognitive change: A representative sample from the general population followed over 16 years. Neurobiol Dis 2023; 189:106357. [PMID: 37977433 DOI: 10.1016/j.nbd.2023.106357] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 09/22/2023] [Accepted: 11/14/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Polygenic risk scores for Alzheimer's disease (AD-PRSs) have been associated with cognition. However, few studies have examined the effect of AD-PRS beyond the APOE gene, and the influence of genetic variants related to level of cognitive ability (COG-PRS) on cognitive performance over time in the general older population. METHOD A population-based sample of 965 individuals born in 1930, with genetic and standardized cognitive data on six psychometric tests (Thurstone's picture memory, immediate recall of 10 words, Block design, word fluency, figure identification, delayed recall of 12 items), were examined at age 70, 75, 79, and 85 years. Non-APOE AD-PRSs and COG-PRSs (P < 5e-8, P < 1e-5, P < 1e-3, P < 1e-1) were generated from recent genome-wide association studies. Linear mixed effect models with random intercepts and slope were used to analyze the effect of APOE ε4 allele, AD-PRSs, and COG-PRSs, on cognitive performance and rate of change. Analyses were repeated in samples excluding dementia. RESULTS APOE ε4 and AD-PRS predicted change in cognitive performance (APOE ε4*age: β = -0.03, P < 0.0001 and AD-PRS *age: β = -0.01, P = 0.02). The results remained similar in the sample excluding those with dementia. COG-PRS predicted level of cognitive performance, while APOE ε4 and AD-PRS did not. COG-PRSs did not predict change in cognitive performance. CONCLUSION We found that genetic predisposition of AD predicted cognitive decline among 70-year-olds followed over 16 years, regardless of dementia status, while polygenic risk for general cognitive performance did not.
Collapse
Affiliation(s)
- Jenna Najar
- Neuropsychiatric Epidemiology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy, Centre for Ageing and Health (AGECAP) at the University of Gothenburg, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Psychiatry, Cognition and Old Age Psychiatry Clinic, Gothenburg, Sweden; Department of Human Genetics, Genomics of Neurodegenerative Diseases and Aging at the Amsterdam University Medical Center, Amsterdam, the Netherlands.
| | - Valgeir Thorvaldsson
- Department of Psychology, and Centre for Ageing and Health (AGECAP), at the University of Gothenburg, Sweden.
| | - Silke Kern
- Neuropsychiatric Epidemiology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy, Centre for Ageing and Health (AGECAP) at the University of Gothenburg, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Psychiatry, Cognition and Old Age Psychiatry Clinic, Gothenburg, Sweden.
| | - Johan Skoog
- Neuropsychiatric Epidemiology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy, Centre for Ageing and Health (AGECAP) at the University of Gothenburg, Sweden.
| | - Margda Waern
- Neuropsychiatric Epidemiology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy, Centre for Ageing and Health (AGECAP) at the University of Gothenburg, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Psychosis Clinic, Gothenburg, Sweden.
| | - Henrik Zetterberg
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China; Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA.
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.
| | - Ingmar Skoog
- Neuropsychiatric Epidemiology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy, Centre for Ageing and Health (AGECAP) at the University of Gothenburg, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Psychiatry, Cognition and Old Age Psychiatry Clinic, Gothenburg, Sweden.
| | - Anna Zettergren
- Neuropsychiatric Epidemiology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy, Centre for Ageing and Health (AGECAP) at the University of Gothenburg, Sweden.
| |
Collapse
|
10
|
Yu C, Ryan J, Orchard SG, Robb C, Woods RL, Wolfe R, Renton AE, Goate AM, Brodtmann A, Shah RC, Chong TTJ, Sheets K, Kyndt C, Sood A, Storey E, Murray AM, McNeil JJ, Lacaze P. Validation of newly derived polygenic risk scores for dementia in a prospective study of older individuals. Alzheimers Dement 2023; 19:5333-5342. [PMID: 37177856 PMCID: PMC10640662 DOI: 10.1002/alz.13113] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 03/27/2023] [Accepted: 04/03/2023] [Indexed: 05/15/2023]
Abstract
INTRODUCTION Recent genome-wide association studies identified new dementia-associated variants. We assessed the performance of updated polygenic risk scores (PRSs) using these variants in an independent cohort. METHODS We used Cox models and area under the curve (AUC) to validate new PRSs (PRS-83SNP, PRS-SBayesR, and PRS-CS) compared with an older PRS-23SNP in 12,031 initially-healthy participants ≥70 years of age. Dementia was rigorously adjudicated according to Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition (DSM-IV) criteria. RESULTS PRS-83SNP, PRS-SBayesR, and PRS-CS were associated with incident dementia, with fully adjusted (including apolipoprotein E [APOE] ε4) hazard ratios per standard deviation (SD) of 1.35 (1.23-1.47), 1.37 (1.25-1.50), and 1.42 (1.30-1.56), respectively. The AUC of a model containing conventional/non-genetic factors and APOE was 74.7%. This was improved to 75.7% (p = 0.007), 76% (p = 0.004), and 76.1% (p = 0.003) with addition of PRS-83SNP, PRS-SBayesR, and PRS-CS, respectively. The PRS-23SNP did not improve AUC (74.7%, p = 0.95). CONCLUSION New PRSs for dementia significantly improve risk-prediction performance, but still account for less risk than APOE genotype overall.
Collapse
Affiliation(s)
- Chenglong Yu
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Joanne Ryan
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Suzanne G. Orchard
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Catherine Robb
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Robyn L. Woods
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Rory Wolfe
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Alan E. Renton
- Department Genetics and Genomic Sciences and Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Departments of Neurology and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Alison M. Goate
- Department Genetics and Genomic Sciences and Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Departments of Neurology and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Amy Brodtmann
- Cognitive Health Initiative, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia
| | - Raj C. Shah
- Department of Family & Preventive Medicine and the Rush Alzheimer’s Disease Center, Chicago, Illinois, USA
| | - Trevor T.-J. Chong
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Victoria, Australia
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
- Department of Clinical Neurosciences, St. Vincent’s Hospital, Melbourne, Victoria, Australia
| | - Kerry Sheets
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- Division of Geriatrics, Hennepin Healthcare, Minneapolis, Minnesota, USA
| | - Christopher Kyndt
- Department of Neurology, Melbourne Health, Parkville, Victoria, Australia
- Department of Neuroscience, Eastern Health, Box Hill, Victoria, Australia
| | - Ajay Sood
- Department of Neurology and the Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, Illinois, USA
| | - Elsdon Storey
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Anne M. Murray
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- Division of Geriatrics, Hennepin Healthcare, Minneapolis, Minnesota, USA
- Berman Center for Outcomes and Clinical Research, Hennepin Healthcare Research Institute, Hennepin Healthcare, and University of Minnesota, Minneapolis, Minnesota, USA
| | - John J. McNeil
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Paul Lacaze
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
11
|
Tsapanou A, Gacheru M, Lee S, Mourtzi N, Gazes Y, Habeck C, Belsky DW, Stern Y. Association of Cognitive Polygenic Index and Cognitive Performance with Age in Cognitively Healthy Adults. Genes (Basel) 2023; 14:1814. [PMID: 37761954 PMCID: PMC10531331 DOI: 10.3390/genes14091814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/08/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Genome-wide association studies have discovered common genetic variants associated with cognitive performance. Polygenic scores that summarize these discoveries explain up to 10% of the variance in cognitive test performance in samples of adults. However, the role these genetics play in cognitive aging is not well understood. We analyzed data from 168 cognitively healthy participants aged 23-77 years old, with data on genetics, neuropsychological assessment, and brain-imaging measurements from two large ongoing studies, the Reference Abilities Neural Networks, and the Cognitive Reserve study. We tested whether a polygenic index previously related to cognition (Cog PGI) would moderate the relationship between age and measurements of the cognitive domains extracted from a neuropsychological evaluation: fluid reasoning, memory, vocabulary, and speed of processing. We further explored the relationship of Cog PGI and age on cognition using Johnson-Neyman intervals for two-way interactions. Sex, education, and brain measures of cortical thickness, total gray matter volume, and white matter hyperintensity were considered covariates. The analysis controlled for population structure-ancestry. There was a significant interaction effect of Cog PGI on the association between age and the domains of memory (Standardized coefficient = -0.158, p-value = 0.022), fluid reasoning (Standardized coefficient = -0.146, p-value = 0.020), and vocabulary (Standardized coefficient = -0.191, p-value = 0.001). Higher PGI strengthened the negative relationship between age and the domains of memory and fluid reasoning while PGI weakened the positive relationship between age and vocabulary. Based on the Johnson-Neyman intervals, Cog PGI was significantly associated with domains of memory, reasoning, and vocabulary for younger adults. There is a significant moderation effect of genetic predisposition for cognition for the association between age and cognitive performance. Genetics discovered in genome-wide association studies of cognitive performance show a stronger association in young and midlife older adults.
Collapse
Affiliation(s)
- Angeliki Tsapanou
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA; (A.T.); (C.H.)
| | - Margaret Gacheru
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, NY 10032, USA;
| | - Seonjoo Lee
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Niki Mourtzi
- Department of Neurology, National and Kapodistrian University of Athens, 10679 Athens, Greece
| | - Yunglin Gazes
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA; (A.T.); (C.H.)
| | - Christian Habeck
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA; (A.T.); (C.H.)
| | - Daniel W. Belsky
- Department of Epidemiology and Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY 10032, USA
| | - Yaakov Stern
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA; (A.T.); (C.H.)
| |
Collapse
|
12
|
Juul Rasmussen I, Frikke-Schmidt R. Modifiable cardiovascular risk factors and genetics for targeted prevention of dementia. Eur Heart J 2023; 44:2526-2543. [PMID: 37224508 PMCID: PMC10481783 DOI: 10.1093/eurheartj/ehad293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 02/22/2023] [Accepted: 05/04/2023] [Indexed: 05/26/2023] Open
Abstract
Dementia is a major global challenge for health and social care in the 21st century. A third of individuals >65 years of age die with dementia, and worldwide incidence numbers are projected to be higher than 150 million by 2050. Dementia is, however, not an inevitable consequence of old age; 40% of dementia may theoretically be preventable. Alzheimer's disease (AD) accounts for approximately two-thirds of dementia cases and the major pathological hallmark of AD is accumulation of amyloid-β. Nevertheless, the exact pathological mechanisms of AD remain unknown. Cardiovascular disease and dementia share several risk factors and dementia often coexists with cerebrovascular disease. In a public health perspective, prevention is crucial, and it is suggested that a 10% reduction in prevalence of cardiovascular risk factors could prevent more than nine million dementia cases worldwide by 2050. Yet this assumes causality between cardiovascular risk factors and dementia and adherence to the interventions over decades for a large number of individuals. Using genome-wide association studies, the entire genome can be scanned for disease/trait associated loci in a hypothesis-free manner, and the compiled genetic information is not only useful for pinpointing novel pathogenic pathways but also for risk assessments. This enables identification of individuals at high risk, who likely will benefit the most from a targeted intervention. Further optimization of the risk stratification can be done by adding cardiovascular risk factors. Additional studies are, however, highly needed to elucidate dementia pathogenesis and potential shared causal risk factors between cardiovascular disease and dementia.
Collapse
Affiliation(s)
- Ida Juul Rasmussen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Ruth Frikke-Schmidt
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
13
|
Silvaieh S, König T, Wurm R, Parvizi T, Berger-Sieczkowski E, Goeschl S, Hotzy C, Wagner M, Berutti R, Sammler E, Stögmann E, Zimprich A. Comprehensive genetic screening of early-onset dementia patients in an Austrian cohort-suggesting new disease-contributing genes. Hum Genomics 2023; 17:55. [PMID: 37330543 PMCID: PMC10276391 DOI: 10.1186/s40246-023-00499-z] [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: 03/06/2023] [Accepted: 05/31/2023] [Indexed: 06/19/2023] Open
Abstract
Early-onset dementia (EOD), with symptom onset before age 65, has a strong genetic burden. Due to genetic and clinical overlaps between different types of dementia, whole-exome sequencing (WES) has emerged as an appropriate screening method for diagnostic testing and novel gene-finding approaches. We performed WES and C9orf72 repeat testing in 60 well-defined Austrian EOD patients. Seven patients (12%) carried likely disease-causing variants in monogenic genes, PSEN1, MAPT, APP, and GRN. Five patients (8%) were APOE4 homozygote carriers. Definite and possible risk variants were detected in the genes TREM2, SORL1, ABCA7 and TBK1. In an explorative approach, we cross-checked rare gene variants in our cohort with a curated neurodegeneration candidate gene list and identified DCTN1, MAPK8IP3, LRRK2, VPS13C and BACE1 as promising candidate genes. Conclusively, 12 cases (20%) carried variants relevant to patient counseling, comparable to previously reported studies, and can thus be considered genetically resolved. Reduced penetrance, oligogenic inheritance and not yet identified high-risk genes might explain the high number of unresolved cases. To address this issue, we provide complete genetic and phenotypic information (uploaded to the European Genome-phenome Archive), enabling other researchers to cross-check variants. Thereby, we hope to increase the chance of independently finding the same gene/variant-hit in other well-defined EOD patient cohorts, thus confirming new genetic risk variants or variant combinations.
Collapse
Affiliation(s)
- Sara Silvaieh
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Theresa König
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Raphael Wurm
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Tandis Parvizi
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Evelyn Berger-Sieczkowski
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Stella Goeschl
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Christoph Hotzy
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Matias Wagner
- Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
- Institute of Neurogenomics, Helmholtz Centrum, Munich, Germany
| | - Riccardo Berutti
- Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
| | - Esther Sammler
- Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - Elisabeth Stögmann
- Department of Neurology, Medical University of Vienna, Vienna, Austria.
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria.
| | - Alexander Zimprich
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
14
|
Xu Y, Sun Z, Jonaitis E, Deming Y, Lu Q, Johnson SC, Engelman CD. Apolipoprotein E moderates the association between Non- APOE Polygenic Risk Score for Alzheimer's Disease and Aging on Preclinical Cognitive Function. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.09.23291215. [PMID: 37398140 PMCID: PMC10312823 DOI: 10.1101/2023.06.09.23291215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
INTRODUCTION Variation in preclinical cognitive decline suggests additional genetic factors related to Alzheimer's disease (e.g., a non-APOE polygenic risk scores [PRS]) may interact with the APOE ε4 allele to influence cognitive decline. METHODS We tested the PRS×APOE ε4×age interaction on preclinical cognition using longitudinal data from the Wisconsin Registry for Alzheimer's Prevention. All analyses were fitted using a linear mixed-effects model and adjusted for within individual/family correlation among 1,190 individuals. RESULTS We found statistically significant PRS×APOE ε4×age interactions on immediate learning (P=0.038), delayed recall (P<0.001), and Preclinical Alzheimer's Cognitive Composite 3 score (P=0.026). PRS-related differences in overall and memory-related cognitive domains between people with and without APOE ε4 emerge around age 70, with a much stronger adverse PRS effect among APOE ε4 carriers. The findings were replicated in a population-based cohort. DISCUSSION APOE ε4 can modify the association between PRS and cognition decline.
Collapse
Affiliation(s)
- Yuexuan Xu
- Department of Population Health Science, School of Medicine and Public Health, University of Wisconsin-Madison
| | - Zhongxuan Sun
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin-Madison
| | - Erin Jonaitis
- Wisconsin Alzheimer’s Institute, University of Wisconsin-Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison
| | - Yuetiva Deming
- Department of Population Health Science, School of Medicine and Public Health, University of Wisconsin-Madison
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison
| | - Qiongshi Lu
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin-Madison
| | - Sterling C. Johnson
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison
- Wisconsin Alzheimer’s Institute, University of Wisconsin-Madison
| | - Corinne D. Engelman
- Department of Population Health Science, School of Medicine and Public Health, University of Wisconsin-Madison
| |
Collapse
|
15
|
Pettigrew C, Nazarovs J, Soldan A, Singh V, Wang J, Hohman T, Dumitrescu L, Libby J, Kunkle B, Gross AL, Johnson S, Lu Q, Engelman C, Masters CL, Maruff P, Laws SM, Morris JC, Hassenstab J, Cruchaga C, Resnick SM, Kitner-Triolo MH, An Y, Albert M. Alzheimer's disease genetic risk and cognitive reserve in relationship to long-term cognitive trajectories among cognitively normal individuals. Alzheimers Res Ther 2023; 15:66. [PMID: 36978190 PMCID: PMC10045505 DOI: 10.1186/s13195-023-01206-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 03/12/2023] [Indexed: 03/30/2023]
Abstract
BACKGROUND Both Alzheimer's disease (AD) genetic risk factors and indices of cognitive reserve (CR) influence risk of cognitive decline, but it remains unclear whether they interact. This study examined whether a CR index score modifies the relationship between AD genetic risk factors and long-term cognitive trajectories in a large sample of individuals with normal cognition. METHODS Analyses used data from the Preclinical AD Consortium, including harmonized data from 5 longitudinal cohort studies. Participants were cognitively normal at baseline (M baseline age = 64 years, 59% female) and underwent 10 years of follow-up, on average. AD genetic risk was measured by (i) apolipoprotein-E (APOE) genetic status (APOE-ε2 and APOE-ε4 vs. APOE-ε3; N = 1819) and (ii) AD polygenic risk scores (AD-PRS; N = 1175). A CR index was calculated by combining years of education and literacy scores. Longitudinal cognitive performance was measured by harmonized factor scores for global cognition, episodic memory, and executive function. RESULTS In mixed-effects models, higher CR index scores were associated with better baseline cognitive performance for all cognitive outcomes. APOE-ε4 genotype and AD-PRS that included the APOE region (AD-PRSAPOE) were associated with declines in all cognitive domains, whereas AD-PRS that excluded the APOE region (AD-PRSw/oAPOE) was associated with declines in executive function and global cognition, but not memory. There were significant 3-way CR index score × APOE-ε4 × time interactions for the global (p = 0.04, effect size = 0.16) and memory scores (p = 0.01, effect size = 0.22), indicating the negative effect of APOE-ε4 genotype on global and episodic memory score change was attenuated among individuals with higher CR index scores. In contrast, levels of CR did not attenuate APOE-ε4-related declines in executive function or declines associated with higher AD-PRS. APOE-ε2 genotype was unrelated to cognition. CONCLUSIONS These results suggest that APOE-ε4 and non-APOE-ε4 AD polygenic risk are independently associated with global cognitive and executive function declines among individuals with normal cognition at baseline, but only APOE-ε4 is associated with declines in episodic memory. Importantly, higher levels of CR may mitigate APOE-ε4-related declines in some cognitive domains. Future research is needed to address study limitations, including generalizability due to cohort demographic characteristics.
Collapse
Affiliation(s)
- Corinne Pettigrew
- Johns Hopkins University School of Medicine, 1600 McElderry St, Baltimore, MD, 21205, USA.
| | - Jurijs Nazarovs
- University of Wisconsin-Madison School of Medicine and Public Health, 750 Highland Ave, Madison, WI, 53726, USA
| | - Anja Soldan
- Johns Hopkins University School of Medicine, 1600 McElderry St, Baltimore, MD, 21205, USA
| | - Vikas Singh
- University of Wisconsin-Madison School of Medicine and Public Health, 750 Highland Ave, Madison, WI, 53726, USA
| | - Jiangxia Wang
- Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD, 21205, USA
| | - Timothy Hohman
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, 1207 17th Ave South, Nashville, TN, 37212, USA
| | - Logan Dumitrescu
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, 1207 17th Ave South, Nashville, TN, 37212, USA
| | - Julia Libby
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, 1207 17th Ave South, Nashville, TN, 37212, USA
| | - Brian Kunkle
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Alden L Gross
- Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD, 21205, USA
| | - Sterling Johnson
- University of Wisconsin-Madison School of Medicine and Public Health, 750 Highland Ave, Madison, WI, 53726, USA
| | - Qiongshi Lu
- University of Wisconsin-Madison School of Medicine and Public Health, 750 Highland Ave, Madison, WI, 53726, USA
| | - Corinne Engelman
- University of Wisconsin-Madison School of Medicine and Public Health, 750 Highland Ave, Madison, WI, 53726, USA
| | - Colin L Masters
- The Florey Institute, University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia
| | - Paul Maruff
- The Florey Institute, University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia
| | - Simon M Laws
- Centre for Precision Health and Collaborative Genomics and Translation Group, Edith Cowan University, 270 Jundaloop Drive, Jundaloop, WA, 6027, Australia
- Curtin Medical School, Curtin University, Kent Street, Bentley, WA, 6102, Australia
| | - John C Morris
- Washington University School of Medicine, 660 S Euclid Ave, St. Louis, MO, 63110, USA
| | - Jason Hassenstab
- Washington University School of Medicine, 660 S Euclid Ave, St. Louis, MO, 63110, USA
| | - Carlos Cruchaga
- Washington University School of Medicine, 660 S Euclid Ave, St. Louis, MO, 63110, USA
| | - Susan M Resnick
- National Institute on Aging Intramural Research Program, 251 Bayview Blvd, Baltimore, MD, 21224, USA
| | - Melissa H Kitner-Triolo
- National Institute on Aging Intramural Research Program, 251 Bayview Blvd, Baltimore, MD, 21224, USA
| | - Yang An
- National Institute on Aging Intramural Research Program, 251 Bayview Blvd, Baltimore, MD, 21224, USA
| | - Marilyn Albert
- Johns Hopkins University School of Medicine, 1600 McElderry St, Baltimore, MD, 21205, USA
| |
Collapse
|
16
|
Tomassen J, den Braber A, van der Lee SJ, Reus LM, Konijnenberg E, Carter SF, Yaqub M, van Berckel BNM, Collij LE, Boomsma DI, de Geus EJC, Scheltens P, Herholz K, Tijms BM, Visser PJ. Amyloid-β and APOE genotype predict memory decline in cognitively unimpaired older individuals independently of Alzheimer's disease polygenic risk score. BMC Neurol 2022; 22:484. [PMID: 36522743 PMCID: PMC9753236 DOI: 10.1186/s12883-022-02925-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 10/14/2022] [Accepted: 10/19/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND What combination of risk factors for Alzheimer's disease (AD) are most predictive of cognitive decline in cognitively unimpaired individuals remains largely unclear. We studied associations between APOE genotype, AD-Polygenic Risk Scores (AD-PRS), amyloid-β pathology and decline in cognitive functioning over time in a large sample of cognitively unimpaired older individuals. METHODS We included 276 cognitively unimpaired older individuals (75 ± 10 years, 63% female) from the EMIF-AD PreclinAD cohort. An AD-PRS was calculated including 83 genome-wide significant variants. The APOE gene was not included in the PRS and was analyzed separately. Baseline amyloid-β status was assessed by visual read of [18F]flutemetamol-PET standardized uptake value images. At baseline and follow-up (2.0 ± 0.4 years), the cognitive domains of memory, attention, executive function, and language were measured. We used generalized estimating equations corrected for age, sex and center to examine associations between APOE genotype and AD-PRS with amyloid-β status. Linear mixed models corrected for age, sex, center and education were used to examine associations between APOE genotype, AD-PRS and amyloid-β status, and their interaction on changes in cognitive functioning over time. RESULTS Fifty-two participants (19%) had abnormal amyloid-β, and 84 participants (31%) carried at least one APOE ε4 allele. APOE genotype and AD-PRS were both associated with abnormal amyloid-β status. Increasingly more risk-full APOE genotype, a high AD-PRS and an abnormal amyloid-β status were associated with steeper decline in memory functioning in separate models (all p ≤ 0.02). A model including 4-way interaction term (APOE×AD-PRS×amyloid-β×time) was not significant. When modelled together, both APOE genotype and AD-PRS predicted steeper decline in memory functioning (APOE β(SE)=-0.05(0.02); AD-PRS β(SE)=-0.04(0.01)). Additionally, when modelled together, both amyloid-β status and AD-PRS predicted a steeper decline in memory functioning (amyloid-β β(SE)=-0.07(0.04); AD-PRS β(SE)=-0.04(0.01)). Modelling both APOE genotype and amyloid-β status, we observed an interaction, in which APOE genotype was related to steeper decline in memory and language functioning in amyloid-β abnormal individuals only (β(SE)=-0.13(0.06); β(SE)=-0.22(0.07), respectively). CONCLUSION Our results suggest that APOE genotype is related to steeper decline in memory and language functioning in individuals with abnormal amyloid-β only. Furthermore, independent of amyloid-β status other genetic risk variants contribute to memory decline in initially cognitively unimpaired older individuals.
Collapse
Affiliation(s)
- Jori Tomassen
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands.
- Alzheimer Center Amsterdam, Neurology, Amsterdam UMC location VUmc, 1007 MB, Amsterdam, PO Box 7057, The Netherlands.
| | - Anouk den Braber
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Department of Biological Psychology, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Sven J van der Lee
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Genomics of Neurodegenerative Diseases and Aging, Human Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Lianne M Reus
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Elles Konijnenberg
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Stephen F Carter
- Wolfson Molecular Imaging Centre, Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, UK
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Maqsood Yaqub
- Department of Radiology & Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands
| | - Bart N M van Berckel
- Department of Radiology & Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands
| | - Lyduine E Collij
- Department of Radiology & Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands
| | - Dorret I Boomsma
- Department of Biological Psychology, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Eco J C de Geus
- Department of Biological Psychology, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Karl Herholz
- Wolfson Molecular Imaging Centre, Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, UK
| | - Betty M Tijms
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Pieter Jelle Visser
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Alzheimer Center Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
17
|
Kawada T. Which reduces the risk of cognitive impairment: physical activity or daytime nap? Psychogeriatrics 2022; 22:772. [PMID: 35662348 DOI: 10.1111/psyg.12864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/10/2022] [Accepted: 05/19/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Tomoyuki Kawada
- Department of Hygiene and Public Health, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
18
|
Svensson A, Garcia-Etxebarria K, Åkesson A, Borgfeldt C, Roth B, Ek M, D'Amato M, Ohlsson B. Applicability of polygenic risk scores in endometriosis clinical presentation. BMC Womens Health 2022; 22:208. [PMID: 35659226 PMCID: PMC9166598 DOI: 10.1186/s12905-022-01788-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 05/24/2022] [Indexed: 11/18/2022] Open
Abstract
Background Risk prediction is an essential part of preventative medicine and in recent years genomic information has become an interesting factor in risk models. Polygenic risk scores (PRS) combine the effect of many genetic variations into a single score which has been shown to have predictive value for many diseases. This study aimed to investigate the association between PRS for endometriosis and the clinical presentation of the disease. Methods Women with endometriosis (N = 172) were identified at the Department of Gynecology. All participants answered questionnaires regarding sociodemographic factors, lifestyle habits and medical history, registered bowel symptoms on the Visual Analog Scale for Irritable Bowel Syndrome and passed blood samples. DNA was extracted and samples were genotyped, and a PRS was calculated based on previous genome-wide association studies of endometriosis. Inflammatory proteins and TSH receptor antibodies (TRAb) in serum were analyzed. Results Inverse associations were identified between PRS and spread of endometriosis, involvement of the gastrointestinal tract and hormone treatment. However, significance was lost when calculated as p for trend and the specificity and sensitivity were low. There were no correlations between PRS and TRAb or inflammatory proteins. Conclusion The findings indicate that specific PRS should be developed to predict clinical presentations in patient with endometriosis. Supplementary Information The online version contains supplementary material available at 10.1186/s12905-022-01788-w.
Collapse
Affiliation(s)
- Agnes Svensson
- Department of Internal Medicine, Skåne University Hospital, Lund University, Malmö, Sweden.
| | - Koldo Garcia-Etxebarria
- Biodonostia, Gastrointestinal Genetics Group, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas Y Digestivas (CIBERehd), 20014, San Sebastian, Spain
| | - Anna Åkesson
- Clinical Studies Sweden - Forum South, Skåne University Hospital, Lund, Sweden
| | - Christer Borgfeldt
- Department of Obstetrics and Gynecology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Bodil Roth
- Department of Internal Medicine, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Malin Ek
- Department of Internal Medicine, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Mauro D'Amato
- Gastrointestinal Genetics Lab, CIC bioGUNE - BRTA, Derio, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao, Spain.,Department of Medicine and Surgery, LUM University, Casamassima, Italy
| | - Bodil Ohlsson
- Department of Internal Medicine, Skåne University Hospital, Lund University, Malmö, Sweden
| |
Collapse
|
19
|
Pushparaj PN, Kalamegam G, Wali Sait KH, Rasool M. Decoding the Role of Astrocytes in the Entorhinal Cortex in Alzheimer’s Disease Using High-Dimensional Single-Nucleus RNA Sequencing Data and Next-Generation Knowledge Discovery Methodologies: Focus on Drugs and Natural Product Remedies for Dementia. Front Pharmacol 2022; 12:720170. [PMID: 35295737 PMCID: PMC8918735 DOI: 10.3389/fphar.2021.720170] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 12/10/2021] [Indexed: 12/14/2022] Open
Abstract
Introduction: Alzheimer’s disease (AD) is a major cause of the development of cognitive decline and dementia. AD and associated dementias (ADRD) are the major contributors to the enormous burden of morbidity and mortality worldwide. To date, there are no robust therapies to alleviate or cure this debilitating disease. Most drug treatments focus on restoring the normal function of neurons and the cells that cause inflammation, such as microglia in the brain. However, the role of astrocytes, the brain’s housekeeping cells, in the development of AD and the initiation of dementia is still not well understood. Objective: To decipher the role of astrocytes in the entorhinal cortex of AD patients using single nuclear RNA sequencing (snRNASeq) datasets from the Single Cell RNA-seq Database for Alzheimer’s Disease (scREAD). The datasets were originally derived from astrocytes, isolated from the entorhinal cortex of AD brain and healthy brain to decipher disease-specific signaling pathways as well as drugs and natural products that reverse AD-specific signatures in astrocytes. Methods: We used snRNASeq datasets from the scREAD database originally derived from astrocytes isolated from the entorhinal cortex of AD and healthy brains from the Gene Expression Omnibus (GEO) (GSE138852 and GSE147528) and analyzed them using next-generation knowledge discovery (NGKD) platforms. scREAD is a user-friendly open-source interface available at https://bmbls.bmi.osumc.edu/scread/that enables more discovery-oriented strategies. snRNASeq data and metadata can also be visualized and downloaded via an interactive web application at adsn.ddnetbio.com. Differentially expressed genes (DEGs) for each snRNASeq dataset were analyzed using iPathwayGuide to compare and derive disease-specific pathways, gene ontologies, and in silico predictions of drugs and natural products that regulate AD -specific signatures in astrocytes. In addition, DEGs were analyzed using the L1000FWD and L1000CDS2 signature search programming interfaces (APIs) to identify additional drugs and natural products that mimic or reverse AD-specific gene signatures in astrocytes. Results: We found that PI3K/AKT signaling, Wnt signaling, neuroactive ligand-receptor interaction pathways, neurodegeneration pathways, etc. were significantly impaired in astrocytes from the entorhinal cortex of AD patients. Biological processes such as glutamate receptor signaling pathway, regulation of synapse organization, cell-cell adhesion via plasma membrane adhesion molecules, and chylomicrons were negatively enriched in the astrocytes from the entorhinal cortex of AD patients. Gene sets involved in cellular components such as postsynaptic membrane, synaptic membrane, postsynapse, and synapse part were negatively enriched (p < 0.01). Moreover, molecular functions such as glutamate receptor activity, neurotransmitter receptor activity, and extracellular ligand-gated ion channels were negatively regulated in the astrocytes of the entorhinal cortex of AD patients (p < 0.01). Moreover, the application of NGKD platforms revealed that antirheumatic drugs, vitamin-E, emetine, narciclasine, cephaeline, trichostatin A, withaferin A, dasatinib, etc. can potentially reverse gene signatures associated with AD. Conclusions: The present study highlights an innovative approach to use NGKD platforms to find unique disease-associated signaling pathways and specific synthetic drugs and natural products that can potentially reverse AD and ADRD-associated gene signatures.
Collapse
Affiliation(s)
- Peter Natesan Pushparaj
- Center of Excellence in Genomic Medicine Research, Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, India
- *Correspondence: Peter Natesan Pushparaj, ; Mahmood Rasool,
| | - Gauthaman Kalamegam
- Center for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Khalid Hussain Wali Sait
- Department of Obstetrics and Gynaecology, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mahmood Rasool
- Center of Excellence in Genomic Medicine Research, Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- *Correspondence: Peter Natesan Pushparaj, ; Mahmood Rasool,
| |
Collapse
|
20
|
Genetic risk scores and dementia risk across different ethnic groups in UK Biobank. PLoS One 2022; 17:e0277378. [PMID: 36477264 PMCID: PMC9728885 DOI: 10.1371/journal.pone.0277378] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 10/26/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Genetic Risk Scores (GRS) for predicting dementia risk have mostly been used in people of European ancestry with limited testing in other ancestry groups. METHODS We conducted a logistic regression with all-cause dementia as the outcome and z-standardised GRS as the exposure across diverse ethnic groups. FINDINGS There was variation in frequency of APOE alleles across ethnic groups. Per standard deviation (SD) increase in z-GRS including APOE, the odds ratio (OR) for dementia was 1.73 (95%CI 1.69-1.77). Z-GRS excluding APOE also increased dementia risk (OR 1.21 per SD increase, 95% CI 1.18-1.24) and there was no evidence that ethnicity modified this association. Prediction of secondary outcomes was less robust in those not of European ancestry when APOE was excluded from the GRS. INTERPRETATION z-GRS derived from studies in people of European ancestry can be used to quantify genetic risk in people from more diverse ancestry groups. Urgent work is needed to include people from diverse ancestries in future genetic risk studies to make this field more inclusive.
Collapse
|
21
|
Abstract
Over the past decade, substantial progress has been made in the discovery of alleles contributing to the risk of coronary artery disease. In addition to providing causal insights into disease, these endeavours have yielded and enabled the refinement of polygenic risk scores. These scores can be used to predict incident coronary artery disease in multiple cohorts and indicate the clinical response to some preventive therapies in post hoc analyses of clinical trials. These observations and the widespread ability to calculate polygenic risk scores from direct-to-consumer and health-care-associated biobanks have raised many questions about responsible clinical adoption. In this Review, we describe technical and downstream considerations for the derivation and validation of polygenic risk scores and current evidence for their efficacy and safety. We discuss the implementation of these scores in clinical medicine for uses including risk prediction and screening algorithms for coronary artery disease, prioritization of patient subgroups that are likely to derive benefit from treatment, and efficient prospective clinical trial designs.
Collapse
|
22
|
Bakshi A, Riaz M, Orchard SG, Carr PR, Joshi AD, Cao Y, Rebello R, Nguyen-Dumont T, Southey MC, Millar JL, Gately L, Gibbs P, Ford LG, Parnes HL, Chan AT, McNeil JJ, Lacaze P. A Polygenic Risk Score Predicts Incident Prostate Cancer Risk in Older Men but Does Not Select for Clinically Significant Disease. Cancers (Basel) 2021; 13:5815. [PMID: 34830967 PMCID: PMC8616400 DOI: 10.3390/cancers13225815] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/24/2022] Open
Abstract
Despite the high prevalence of prostate cancer in older men, the predictive value of a polygenic risk score (PRS) remains uncertain in men aged ≥70 years. We used a 6.6 million-variant PRS to predict the risk of incident prostate cancer in a prospective study of 5701 men of European descent aged ≥70 years (mean age 75 years) enrolled in the ASPirin in Reducing Events in the Elderly (ASPREE) clinical trial. The study endpoint was prostate cancer, including metastatic or non-metastatic disease, confirmed by an expert panel. After excluding participants with a history of prostate cancer at enrolment, we used a multivariable Cox proportional hazards model to assess the association between the PRS and incident prostate cancer risk, adjusting for covariates. Additionally, we examined the distribution of Gleason grade groups by PRS group to determine if a higher PRS was associated with higher grade disease. We tested for interaction between the PRS and aspirin treatment. Logistic regression was used to independently assess the association of the PRS with prevalent (pre-trial) prostate cancer, reported in medical histories. During a median follow-up time of 4.6 years, 218 of the 5701 participants (3.8%) were diagnosed with prostate cancer. The PRS predicted incident risk with a hazard ratio (HR) of 1.52 per standard deviation (SD) (95% confidence interval (CI) 1.33-1.74, p < 0.001). Men in the top quintile of the PRS distribution had an almost three times higher risk of prostate cancer than men in the lowest quintile (HR = 2.99 (95% CI 1.90-4.27), p < 0.001). However, a higher PRS was not associated with a higher Gleason grade groups. We found no interaction between aspirin treatment and the PRS for prostate cancer risk. The PRS was also associated with prevalent prostate cancer (odds ratio = 1.80 per SD (95% CI 1.65-1.96), p < 0.001).While a PRS for prostate cancer is strongly associated with incident risk in men aged ≥70 years, the clinical utility of the PRS as a biomarker is currently limited by its inability to select for clinically significant disease.
Collapse
Affiliation(s)
- Andrew Bakshi
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC 3004, Australia; (M.R.); (S.G.O.); (P.R.C.); (J.L.M.); (J.J.M.); (P.L.)
| | - Moeen Riaz
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC 3004, Australia; (M.R.); (S.G.O.); (P.R.C.); (J.L.M.); (J.J.M.); (P.L.)
| | - Suzanne G. Orchard
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC 3004, Australia; (M.R.); (S.G.O.); (P.R.C.); (J.L.M.); (J.J.M.); (P.L.)
| | - Prudence R. Carr
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC 3004, Australia; (M.R.); (S.G.O.); (P.R.C.); (J.L.M.); (J.J.M.); (P.L.)
| | - Amit D. Joshi
- Clinical and Translational Epidemiology Unit, MGH Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02108, USA; (A.D.J.); (A.T.C.)
| | - Yin Cao
- Alvin J. Siteman Cancer Center, Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA;
| | - Richard Rebello
- Centre for Cancer Research, Department of Clinical Pathology, University of Melbourne, Melbourne, VIC 3010, Australia;
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Tú Nguyen-Dumont
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Melbourne, VIC 3168, Australia; (T.N.-D.); (M.C.S.)
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Melissa C. Southey
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Melbourne, VIC 3168, Australia; (T.N.-D.); (M.C.S.)
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC 3010, Australia
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC 3004, Australia
| | - Jeremy L. Millar
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC 3004, Australia; (M.R.); (S.G.O.); (P.R.C.); (J.L.M.); (J.J.M.); (P.L.)
- Alfred Health Radiation Oncology, Alfred Hospital, Melbourne, VIC 3004, Australia
- Central Clinical School, Monash University, Melbourne, VIC 3168, Australia
| | - Lucy Gately
- Personalised Oncology Division, Walter and Eliza Hall Institute Medical Research, Faculty of Medicine, University of Melbourne, Melbourne, VIC 3052, Australia; (L.G.); (P.G.)
| | - Peter Gibbs
- Personalised Oncology Division, Walter and Eliza Hall Institute Medical Research, Faculty of Medicine, University of Melbourne, Melbourne, VIC 3052, Australia; (L.G.); (P.G.)
| | - Leslie G. Ford
- Division of Cancer Prevention, National Cancer Institute, Rockville, MD 20892, USA; (L.G.F.); (H.L.P.)
| | - Howard L. Parnes
- Division of Cancer Prevention, National Cancer Institute, Rockville, MD 20892, USA; (L.G.F.); (H.L.P.)
| | - Andrew T. Chan
- Clinical and Translational Epidemiology Unit, MGH Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02108, USA; (A.D.J.); (A.T.C.)
| | - John J. McNeil
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC 3004, Australia; (M.R.); (S.G.O.); (P.R.C.); (J.L.M.); (J.J.M.); (P.L.)
| | - Paul Lacaze
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC 3004, Australia; (M.R.); (S.G.O.); (P.R.C.); (J.L.M.); (J.J.M.); (P.L.)
- Clinical and Translational Epidemiology Unit, MGH Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02108, USA; (A.D.J.); (A.T.C.)
| |
Collapse
|