1
|
Li J, Li J, Chen S, Liu Z, Dai J, Wang Y, Cui M, Suo C, Xu K, Jin L, Chen X, Jiang Y. Prospective Investigation Unravels Plasma Proteomic Links to Dementia. Mol Neurobiol 2025; 62:7345-7360. [PMID: 39885106 DOI: 10.1007/s12035-025-04716-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 01/19/2025] [Indexed: 02/01/2025]
Abstract
Investigating plasma proteomic signatures of dementia offers insights into its pathology, aids biomarker discovery, supports disease monitoring, and informs drug development. Here, we analyzed data from 48,367 UK Biobank participants with proteomic profiling. Using Cox and generalized linear models, we examined the longitudinal associations between proteomic signatures and dementia-related phenotypes. Mendelian randomization analysis was employed to identify causal associations, and machine learning algorithms were applied to develop protein-based models for dementia prediction. We identified 74 proteins significantly associated with the risk of various types of dementia and cognitive functions after Bonferroni correction. Among these, strong associations were observed for growth/differentiation factor 15 (GDF15), glial fibrillary acidic protein (GFAP), and neurofilament light polypeptide (NEFL), across all types of dementia. Additionally, 15 proteins demonstrated significant associations with neuroimaging-defined dementia endophenotypes. Two-sample Mendelian randomization analyses further substantiated causal relationships between dementia-associated proteins and Alzheimer's disease, particularly involving GDF15, proto-oncogene tyrosine-protein kinase receptor Ret (RET), and GFAP. Moreover, we identified three protein modules associated with dementia, primarily linked to immune system processes, angiogenesis, and energy metabolism, providing insights into potential biological pathways underlying the disease. Furthermore, we proposed a ten-protein panel capable of forecasting dementia over a median follow-up period of 8.6 years, achieving an area under the curve (AUC) of 0.857 (95% confidence interval (CI), 0.837-0.876). Our results revealed dementia-associated plasma proteomic signatures, and their causal relationships, notably GDF15-RET signaling with Alzheimer's disease, and proposed a promising protein panel for high-risk dementia screening.
Collapse
Affiliation(s)
- Jincheng Li
- Research and Innovation Center, Shanghai Pudong Hospital, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, 200433, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, 225300, Jiangsu, China
| | - Jialin Li
- Research and Innovation Center, Shanghai Pudong Hospital, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, 200433, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, 225300, Jiangsu, China
| | - Shuaizhou Chen
- Research and Innovation Center, Shanghai Pudong Hospital, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, 200433, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, 225300, Jiangsu, China
| | - Zhenqiu Liu
- Research and Innovation Center, Shanghai Pudong Hospital, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, 200433, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, 225300, Jiangsu, China
| | - Jiacheng Dai
- Research and Innovation Center, Shanghai Pudong Hospital, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, 200433, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, 225300, Jiangsu, China
| | - Yingzhe Wang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Mei Cui
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Chen Suo
- Fudan University Taizhou Institute of Health Sciences, Taizhou, 225300, Jiangsu, China
- Ministry of Education Key Laboratory of Public Health Safety, Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China
| | - Kelin Xu
- Fudan University Taizhou Institute of Health Sciences, Taizhou, 225300, Jiangsu, China
- Ministry of Education Key Laboratory of Public Health Safety, Department of Biostatistics, School of Public Health, Fudan University, Shanghai, 200032, China
| | - Li Jin
- Fudan University Taizhou Institute of Health Sciences, Taizhou, 225300, Jiangsu, China
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200433, China
| | - Xingdong Chen
- Fudan University Taizhou Institute of Health Sciences, Taizhou, 225300, Jiangsu, China.
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200433, China.
- Yiwu Research Institute of Fudan University, Yiwu, 322000, Zhejiang, China.
| | - Yanfeng Jiang
- Research and Innovation Center, Shanghai Pudong Hospital, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, 200433, China.
- Fudan University Taizhou Institute of Health Sciences, Taizhou, 225300, Jiangsu, China.
- International Human Phenome Institute (Shanghai), Shanghai, 201210, China.
| |
Collapse
|
2
|
Beydoun MA, Beydoun HA, Li Z, Hu YH, Noren Hooten N, Ding J, Hossain S, Maino Vieytes CA, Launer LJ, Evans MK, Zonderman AB. Alzheimer's Disease polygenic risk, the plasma proteome, and dementia incidence among UK older adults. GeroScience 2025; 47:2507-2523. [PMID: 39586964 PMCID: PMC11978584 DOI: 10.1007/s11357-024-01413-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 10/23/2024] [Indexed: 11/27/2024] Open
Abstract
Alzheimer's Disease (AD) is a complex polygenic neurodegenerative disorder. Its genetic risk's relationship with all-cause dementia may be influenced by the plasma proteome. Up to 40,139 UK Biobank participants aged ≥ 50y at baseline assessment (2006-2010) were followed-up for ≤ 15 y for dementia incidence. Plasma proteomics were performed on a sub-sample of UK Biobank participants (k = 1,463 plasma proteins). AD polygenic risk scores (PRS) were used as the primary exposure and Cox proportional hazards models were conducted to examine the AD PRS-dementia relationship. A four-way decomposition model then partitioned the total effect (TE) of AD PRS on dementia into an effect due to mediation only, an effect due to interaction only, neither or both. The study found that AD PRS tertiles significantly increased the risk for all-cause dementia, particularly among women. The study specifically found that AD PRS was associated with a 79% higher risk for all-cause dementia for each unit increase (HR = 1.79, 95% CI: 1.70-1.87, P < 0.001). Eighty-six plasma proteins were significantly predicted by AD PRS, including a positive association with PLA2G7, BRK1, the glial acidic fibrillary protein (GFAP), neurofilament light chain (NfL), and negative with TREM2. Both GFAP and NfL significantly interacted synergistically with AD PRS to increase all-dementia risk (> 10% of TE is pure interaction), while GFAP was also an important consistent mediator in the AD PRS-dementia relationship. In summary, we detected significant interactions of NfL and GFAP with AD PRS, in relation to dementia incidence, suggesting potential for personalized dementia prevention and management.
Collapse
Affiliation(s)
- May A Beydoun
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA.
| | - Hind A Beydoun
- VA National Center On Homelessness Among Veterans, U.S. Department of Veterans Affairs, Washington, DC, 20420, USA
- Department of Management, Policy, and Community Health, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Zhiguang Li
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Yi-Han Hu
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Jun Ding
- Translational Gerontology Branch, National Institute On Aging, NIA/NIH/IRP, Baltimore, MD, 21224, USA
| | - Sharmin Hossain
- Department of Human Services (DHS), State of Maryland, Baltimore, MD, 21202, USA
| | - Christian A Maino Vieytes
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| |
Collapse
|
3
|
Yousefpour Shahrivar R, Karami F, Karami E. Differential gene expression patterns in Niemann-Pick Type C and Tay-Sachs diseases: Implications for neurodegenerative mechanisms. PLoS One 2025; 20:e0319401. [PMID: 40106490 PMCID: PMC11922228 DOI: 10.1371/journal.pone.0319401] [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: 09/30/2024] [Accepted: 02/01/2025] [Indexed: 03/22/2025] Open
Abstract
Lysosomal storage disorders (LSDs) are a group of rare genetic conditions characterized by the impaired function of enzymes responsible for lipid digestion. Among these LSDs, Tay-Sachs disease (TSD) and Niemann-Pick type C (NPC) may share a common gene expression profile. In this study, we conducted a bioinformatics analysis to explore the gene expression profile overlap between TSD and NPC. Analyses were performed on RNA-seq datasets for both TSD and NPC from the Gene Expression Omnibus (GEO) database. Datasets were subjected to differential gene expression analysis utilizing the DESeq2 package in the R programming language. A total of 147 differentially expressed genes (DEG) were found to be shared between the TSD and NPC datasets. Enrichment analysis was then performed on the DEGs. We found that the common DEGs are predominantly associated with processes such as cell adhesion mediated by integrin, cell-substrate adhesion, and urogenital system development. Furthermore, construction of protein-protein interaction (PPI) networks using the Cytoscape software led to the identification of four hub genes: APOE, CD44, SNCA, and ITGB5. Those hub genes not only can unravel the pathogenesis of related neurologic diseases with common impaired pathways, but also may pave the way towards targeted gene therapy of LSDs.In addition, they serve as the potential biomarkers for related neurodegenerative diseases warranting further investigations.
Collapse
Affiliation(s)
- Ramin Yousefpour Shahrivar
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Fatemeh Karami
- Department of Medical Genetics, Applied Biophotonics Research Center, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Ebrahim Karami
- Department of Electrical and Computer Engineering, Faculty of Engineering and Applied Sciences, Memorial University of Newfoundland, St. John's, Canada
| |
Collapse
|
4
|
Ivarsson Orrelid C, Rosberg O, Weiner S, Johansson FD, Gobom J, Zetterberg H, Mwai N, Stempfle L. Applying machine learning to high-dimensional proteomics datasets for the identification of Alzheimer's disease biomarkers. Fluids Barriers CNS 2025; 22:23. [PMID: 40033432 PMCID: PMC11874791 DOI: 10.1186/s12987-025-00634-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 02/11/2025] [Indexed: 03/05/2025] Open
Abstract
PURPOSE This study explores the application of machine learning to high-dimensional proteomics datasets for identifying Alzheimer's disease (AD) biomarkers. AD, a neurodegenerative disorder affecting millions worldwide, necessitates early and accurate diagnosis for effective management. METHODS We leverage Tandem Mass Tag (TMT) proteomics data from the cerebrospinal fluid (CSF) samples from the frontal cortex of patients with idiopathic normal pressure hydrocephalus (iNPH), a condition often comorbid with AD, with rare access to both lumbar and ventricular samples. Our methodology includes extensive data preprocessing to address batch effects and missing values, followed by the use of the Synthetic Minority Over-sampling Technique (SMOTE) for data augmentation to overcome the small sample size. We apply linear, and non-linear machine learning models, and ensemble methods, to compare iNPH patients with and without biomarker evidence of AD pathology ( A β - T - or A β + T + ) in a classification task. RESULTS We present a machine learning workflow for working with high-dimensional TMT proteomics data that addresses their inherent data characteristics. Our results demonstrate that batch effect correction has no or minor impact on the models' performance and robust feature selection is critical for model stability and performance, especially in the high-dimensional proteomics data setting for AD diagnostics. The results further indicated that removing features with missing values produced stronger models than imputing them, and the batch effect had minimal impact on the models Our best-performing disease-progression detection model, a random forest, achieves an AUC of 0.84 (± 0.03). CONCLUSION We identify several novel protein biomarkers candidates, such as FABP3 and GOT1, with potential diagnostic value for AD pathology detection, suggesting the necessity of different biomarkers for AD diagnoses for patients with iNPH, and considering different biomarkers for ventricular and lumbar CSF samples. This work underscores the importance of a meticulous machine learning process in enhancing biomarker discovery. Our study also provides insights in translating biomarkers from other central nervous system diseases like iNPH, and both ventricular and lumbar CSF samples for biomarker discovery, providing a foundation for future research and clinical applications.
Collapse
Affiliation(s)
- Christoffer Ivarsson Orrelid
- Computer Science and Engineering, Chalmers University of Technology and University of Gothenburg, Rännvägen 6b, 41296, Gothenburg, Västra Götalandsregionen, Sweden.
| | - Oscar Rosberg
- Computer Science and Engineering, Chalmers University of Technology and University of Gothenburg, Rännvägen 6b, 41296, Gothenburg, Västra Götalandsregionen, Sweden
| | - Sophia Weiner
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Wallinsgatan 6, 43141, Möndal, Västra Götalandsregionen, Sweden
| | - Fredrik D Johansson
- Computer Science and Engineering, Chalmers University of Technology and University of Gothenburg, Rännvägen 6b, 41296, Gothenburg, Västra Götalandsregionen, Sweden
| | - Johan Gobom
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Wallinsgatan 6, 43141, Möndal, Västra Götalandsregionen, Sweden
- Clinical Neurochemistry Lab, Clinical Neurochemistry Lab, Institute of Neuroscience and Physiology, Sahlgrenska University Hospital, Wallinsgatan 6, 43141, Möndal, Västra Götalandsregionen, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Wallinsgatan 6, 43141, Möndal, Västra Götalandsregionen, Sweden
- Clinical Neurochemistry Lab, Clinical Neurochemistry Lab, Institute of Neuroscience and Physiology, Sahlgrenska University Hospital, Wallinsgatan 6, 43141, Möndal, Västra Götalandsregionen, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, UCL Institute of Neurology, Queen Square, London, UK
- 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
| | - Newton Mwai
- Computer Science and Engineering, Chalmers University of Technology and University of Gothenburg, Rännvägen 6b, 41296, Gothenburg, Västra Götalandsregionen, Sweden
| | - Lena Stempfle
- Computer Science and Engineering, Chalmers University of Technology and University of Gothenburg, Rännvägen 6b, 41296, Gothenburg, Västra Götalandsregionen, Sweden
| |
Collapse
|
5
|
Miao J, Zhang Y, Su C, Zheng Q, Guo J. Insulin-Like Growth Factor Signaling in Alzheimer's Disease: Pathophysiology and Therapeutic Strategies. Mol Neurobiol 2025; 62:3195-3225. [PMID: 39240280 PMCID: PMC11790777 DOI: 10.1007/s12035-024-04457-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 08/27/2024] [Indexed: 09/07/2024]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia among the elderly population, posing a significant public health challenge due to limited therapeutic options that merely delay cognitive decline. AD is associated with impaired energy metabolism and reduced neurotrophic signaling. The insulin-like growth factor (IGF) signaling pathway, crucial for central nervous system (CNS) development, metabolism, repair, cognition, and emotion regulation, includes IGF-1, IGF-2, IGF-1R, IGF-2R, insulin receptor (IR), and six insulin-like growth factor binding proteins (IGFBPs). Research has identified abnormalities in IGF signaling in individuals with AD and AD models. Dysregulated expression of IGFs, receptors, IGFBPs, and disruptions in downstream phosphoinositide 3-kinase-protein kinase B (PI3K/AKT) and mitogen-activated protein kinase (MAPK) pathways collectively increase AD susceptibility. Studies suggest modulating the IGF pathway may ameliorate AD pathology and cognitive decline. This review explores the CNS pathophysiology of IGF signaling in AD progression and assesses the potential of targeting the IGF system as a novel therapeutic strategy. Further research is essential to elucidate how aberrant IGF signaling contributes to AD development, understand underlying molecular mechanisms, and evaluate the safety and efficacy of IGF-based treatments.
Collapse
Affiliation(s)
- Jie Miao
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Yanli Zhang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
- Department of Neurology, Sixth Hospital of Shanxi Medical University (General Hospital of Tisco), Taiyuan, 030001, Shanxi, China
| | - Chen Su
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Qiandan Zheng
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Junhong Guo
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
6
|
Sim MA, Doecke JD, Liew OW, Wong LL, Tan ESJ, Chan SP, Chong JRF, Cai Y, Hilal S, Venketasubramanian N, Tan BY, Lai MKP, Choi H, Masters CL, Richards AM, Chen CLH. Plasma proteomics for cognitive decline and dementia-A Southeast Asian cohort study. Alzheimers Dement 2025; 21:e14577. [PMID: 39998981 PMCID: PMC11854348 DOI: 10.1002/alz.14577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/27/2024] [Accepted: 01/12/2025] [Indexed: 02/27/2025]
Abstract
INTRODUCTION The prognostic utility of plasma proteomics for cognitive decline and dementia in a Southeast Asian population characterized by high cerebrovascular disease (CeVD) burden is underexplored. METHODS We examined this in a Singaporean memory clinic cohort of 528 subjects (n = 300, CeVD; n = 167, incident cognitive decline) followed-up for 4 years. RESULTS Of 1441 plasma proteins surveyed, a 12-protein signature significantly predicted cognitive decline (q-value < .05). Sixteen diverse biological processes were implicated in cognitive decline. Ten proteins independently predicted incident dementia (q-value < .05). A unified prediction model combining plasma proteins with clinical risk factors increased the area under the curve for outcome prediction from 0.62 to 0.85. External validation in the cerebrospinal fluid proteome of an independent Caucasian cohort replicated four of the significantly predictive plasma markers for cognitive decline namely: GFAP, NEFL, AREG, and PPY. DISCUSSION The prognostic proteins prioritized in our study provide robust signals in two different biological matrices, representing potential mechanistic targets for dementia and cognitive decline. HIGHLIGHTS A total of 1441 plasma proteins were profiled in a Singaporean memory clinic cohort. We report prognostic plasma protein signatures for cognitive decline and dementia. External validation was performed in the cerebrospinal fluid proteome of a Caucasian cohort. A concordant proteomic signature was identified across both biofluids and cohorts. Further studies are needed to explore the therapeutic implications of these proteins for dementia.
Collapse
Affiliation(s)
- Ming Ann Sim
- Departments of Pharmacology and Psychological Medicine, Memory Aging and Cognition CentreNational University of SingaporeSingaporeSingapore
- Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of AnesthesiaNational University Health SystemSingaporeSingapore
| | - James D. Doecke
- Australian E‐Health Research CentreCSIROHerstonQueenslandAustralia
| | - Oi Wah Liew
- Department of CardiologyNational University Heart CentreSingaporeSingapore
- Cardiovascular Research InstituteNational University of SingaporeSingaporeSingapore
| | - Lee Lee Wong
- Department of CardiologyNational University Heart CentreSingaporeSingapore
- Cardiovascular Research InstituteNational University of SingaporeSingaporeSingapore
| | - Eugene S. J. Tan
- Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of CardiologyNational University Heart CentreSingaporeSingapore
| | - Siew Pang Chan
- Department of CardiologyNational University Heart CentreSingaporeSingapore
- Cardiovascular Research InstituteNational University of SingaporeSingaporeSingapore
| | - Joyce R. F. Chong
- Departments of Pharmacology and Psychological Medicine, Memory Aging and Cognition CentreNational University of SingaporeSingaporeSingapore
| | - Yuan Cai
- Department of Medicine and Therapeutics, Faculty of MedicineDivision of NeurologyThe Chinese University of Hong KongMa Liu ShuiHong Kong
| | - Saima Hilal
- Saw Swee Hock School of Public HealthNational University of Singapore and National University Health SystemSingaporeSingapore
| | | | - Boon Yeow Tan
- Department of MedicineSt Luke's HospitalSingaporeSingapore
| | | | - Mitchell K. P Lai
- Departments of Pharmacology and Psychological Medicine, Memory Aging and Cognition CentreNational University of SingaporeSingaporeSingapore
| | - Hyungwon Choi
- Cardiovascular Research InstituteNational University of SingaporeSingaporeSingapore
- Department of MedicineYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Singapore Lipidomics IncubatorLife Sciences InstituteNational University of SingaporeSingaporeSingapore
| | - Colin L. Masters
- The Florey Institute of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
| | - Arthur Mark Richards
- Department of CardiologyNational University Heart CentreSingaporeSingapore
- Cardiovascular Research InstituteNational University of SingaporeSingaporeSingapore
- Christchurch Heart InstituteUniversity of OtagoDunedinNew Zealand
| | - Christopher L. H. Chen
- Departments of Pharmacology and Psychological Medicine, Memory Aging and Cognition CentreNational University of SingaporeSingaporeSingapore
| |
Collapse
|
7
|
Beydoun MA, Beydoun HA, Hu YH, Li Z, Georgescu MF, Noren Hooten N, Bouhrara M, Weiss J, Launer LJ, Evans MK, Zonderman AB. Mediating and moderating effects of plasma proteomic biomarkers on the association between poor oral health problems and brain white matter microstructural integrity: the UK Biobank study. Mol Psychiatry 2025; 30:388-401. [PMID: 39080466 PMCID: PMC11746130 DOI: 10.1038/s41380-024-02678-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 01/22/2025]
Abstract
The plasma proteome can mediate associations between periodontal disease (Pd) and brain white matter integrity (WMI). We screened 5089 UK Biobank participants aged 40-70 years for poor oral health problems (POHP). We examined the association between POHP and WMI (fractional anisotropy (FA), mean diffusivity (MD), Intracellular Volume Fraction (ICVF), Isotropic Volume Fraction (ISOVF) and Orientation Diffusion (OD)), decomposing the total effect through the plasma proteome of 1463 proteins into pure mediation, pure interaction, neither, while adjusting for socio-demographic and cardiovascular health factors. Similarly, structural equations modeling (SEM) was conducted. POHP was more prevalent among men (12.3% vs. 9.6%), and was associated with lower WMI on most metrics, in a sex-specific manner. Of 15 proteins strongly associated with POHP, growth differentiation factor 15 (GDF15) and WAP four-disulfide core domain 2 (WFDC2; also known as human epididymis protein 4; HE4) were consistent mediators. Both proteins mediated 7-8% of total POHP effect on FAmean. SEM yielded significant total effects for FAmean, MDmean and ISOVFmean in full models, with %mediated by common latent factor (GDF15 and WFDC2) ranging between 13% (FAmean) and 19% (ISOVFmean). For FA, mediation by this common factor was found for 16 of 49 tract-specific and global mean metrics. Protein metabolism, immune system, and signal transduction were the most common pathways for mediational effects. POHP was associated with poorer WMI, which was partially mediated by GDF15 and WFDC2.
Collapse
Affiliation(s)
- May A Beydoun
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD, 21224, USA.
| | - Hind A Beydoun
- VA National Center on Homelessness Among Veterans, U.S. Department of Veterans Affairs, Washington, DC, 20420, USA
- Department of Management, Policy, and Community Health, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Yi-Han Hu
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD, 21224, USA
| | - Zhiguang Li
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD, 21224, USA
| | - Michael F Georgescu
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD, 21224, USA
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD, 21224, USA
| | - Mustapha Bouhrara
- Laboratory of Clinical Investigation, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD, 21224, USA
| | - Jordan Weiss
- Stanford Center on Longevity, Stanford University, Stanford, CA, 94301, USA
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD, 21224, USA
| | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD, 21224, USA
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD, 21224, USA
| |
Collapse
|
8
|
Farnum Z, Mani R, Bindoff A, Wilson R, Fiotakis A, Stephens J, Cho E, Mackay-Sim A, Sinclair D. Convergent effects of synthetic glucocorticoid dexamethasone and amyloid beta in human olfactory neurosphere-derived cells. J Neurochem 2025; 169:e16263. [PMID: 39556451 DOI: 10.1111/jnc.16263] [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: 06/18/2024] [Revised: 10/10/2024] [Accepted: 10/22/2024] [Indexed: 11/20/2024]
Abstract
Stressful life events and glucocorticoid (stress) hormones appear to increase the risk of Alzheimer's disease and hasten its progression, but the reasons for this remain unclear. One potential explanation is that when amyloid β (Aβ) pathology is accumulating in the preclinical disease stage, glucocorticoid receptor signalling during stressful events exacerbates cellular dysfunction caused by Aβ. Alternatively, Aβ may disrupt glucocorticoid receptor signalling. To explore these possibilities, we investigated whether the synthetic glucocorticoid dexamethasone and Aβ have overlapping effects on the cellular proteome and whether Aβ influences canonical glucocorticoid receptor function. Human olfactory neurosphere-derived (ONS) cells, collected from the olfactory mucosa of six adult donors, were treated with soluble Aβ40 or Aβ42 followed by dexamethasone. Proteins were quantified by mass spectrometry. After 32 h treatment, Aβ40 and Aβ42 both induced profound changes in innate immunity-related proteins. After 72 h, Aβ42 formed widespread aggregates and induced few proteomic changes, whereas Aβ40 remained soluble and altered expression of mitochondrial and innate immunity-related proteins. ONS cells revealed overlapping impacts of Aβ40 and dexamethasone, with 23 proteins altered by both treatments. For 16 proteins (including eight mitochondrial proteins) dexamethasone counteracted the effects of Aβ40. For example, caspase 4 and methylmalonate-semialdehyde dehydrogenase were increased by Aβ40 and decreased by dexamethasone. Consistent with this finding, Aβ40 increased, but dexamethasone decreased, ONS cell proliferation. For seven proteins, including superoxide dismutase [Mn] mitochondrial, dexamethasone exacerbated the effects of Aβ40. For some proteins, including complement C3, the effects of dexamethasone differed depending on whether Aβ40 was present or absent. Neither Aβ species influenced glucocorticoid receptor nuclear translocation. Overall, this study revealed that glucocorticoid receptor signalling modifies the intracellular effects of Aß40, counteracting some effects and exacerbating others. It suggests that cellular mechanisms through which glucocorticoid receptor signalling influences Alzheimer's disease risk/progression are complex and determined by the balance of beneficial and detrimental glucocorticoid effects.
Collapse
Affiliation(s)
- Zane Farnum
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Radhika Mani
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Aidan Bindoff
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Richard Wilson
- Central Science Laboratory, University of Tasmania, Hobart, Tasmania, Australia
| | - Adoni Fiotakis
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Jessica Stephens
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Ellie Cho
- Biological Optical Microscopy Platform, The University of Melbourne, Melbourne, Victoria, Australia
| | - Alan Mackay-Sim
- Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland, Australia
| | - Duncan Sinclair
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
9
|
Beydoun MA, Beydoun HA, Noren Hooten N, Li Z, Hu Y, Georgescu MF, Hossain S, Tanaka T, Bouhrara M, Maino Vieytes CA, Fanelli‐Kuczmarski MT, Launer LJ, Evans MK, Zonderman AB. Plasma proteomic biomarkers as mediators or moderators for the association between poor cardiovascular health and white matter microstructural integrity: The UK Biobank study. Alzheimers Dement 2025; 21:e14507. [PMID: 39822062 PMCID: PMC11864230 DOI: 10.1002/alz.14507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 11/16/2024] [Accepted: 12/03/2024] [Indexed: 01/19/2025]
Abstract
INTRODUCTION The plasma proteome's mediating or moderating roles in the association between poor cardiovascular health (CVH) and brain white matter (WM) microstructural integrity are largely unknown. METHODS Data from 3953 UK Biobank participants were used (40-70 years, 2006-2010), with a neuroimaging visit between 2014 and 2021. Poor CVH was determined using Life's Essential 8 (LE8) and reversing standardized z-scores (LE8z _rev). The plasma proteome was examined as a potential mediator or moderator of LE8z _rev's effects on quantitative diffusion-weighted magnetic resonance imaging (dMRI) metrics. RESULTS LE8z_rev was significantly associated with deteriorated WM microstructural integrity, as reflected by lower tract-averaged fractional anisotropy (dMRI-FAmean), (β ± standared error (SE): -0.00152 ± 0.0003, p < 0.001) and higher tract-averaged orientation dispersion (dMRI-ODmean), (β ± SE:+0.00081 ± 0.00017, p < 0.001). Ten strongly mediating plasma proteins of 1463 were identified, with leptin as the principal driver. DISCUSSION Poor CVH is linked to poor WM microstructural integrity measures (lower FAmean and higher ODmean), mostly mediated through leptin. HIGHLIGHTS Up to 3953 UK Biobank participants were selected for this study. Poor cardiovascular health (CVH) was determined using Life's Essential 8. The plasma proteome was examined as a potential mediator or moderator of poor CVH's effect on dMRI metrics. Ten plasma proteins were identified with strong mediating effects, with leptin being the principal driver.
Collapse
Affiliation(s)
- May A. Beydoun
- Laboratory of Epidemiology and Population SciencesNational Institute on Aging, NIA/NIH/IRPBaltimoreMarylandUSA
| | - Hind A. Beydoun
- VA National Center on Homelessness Among VeteransU.S. Department of Veterans AffairsWashington, DCUSA
- Department of Management, Policy, and Community Health, School of Public HealthUniversity of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population SciencesNational Institute on Aging, NIA/NIH/IRPBaltimoreMarylandUSA
| | - Zhiguang Li
- Laboratory of Epidemiology and Population SciencesNational Institute on Aging, NIA/NIH/IRPBaltimoreMarylandUSA
| | - Yi‐Han Hu
- Laboratory of Epidemiology and Population SciencesNational Institute on Aging, NIA/NIH/IRPBaltimoreMarylandUSA
| | - Michael F. Georgescu
- Laboratory of Epidemiology and Population SciencesNational Institute on Aging, NIA/NIH/IRPBaltimoreMarylandUSA
| | - Sharmin Hossain
- Department of Human Services (DHS)State of MarylandBaltimoreMarylandUSA
| | - Toshiko Tanaka
- Translational Gerontology BranchNational Institute on Aging, NIA/NIH/IRPBaltimoreMarylandUSA
| | - Mustapha Bouhrara
- Laboratory of Clinical InvestigationNational Institute on Aging, NIA/NIH/IRPBaltimoreMarylandUSA
| | - Christian A. Maino Vieytes
- Laboratory of Epidemiology and Population SciencesNational Institute on Aging, NIA/NIH/IRPBaltimoreMarylandUSA
| | - Marie T. Fanelli‐Kuczmarski
- Laboratory of Epidemiology and Population SciencesNational Institute on Aging, NIA/NIH/IRPBaltimoreMarylandUSA
| | - Lenore J. Launer
- Laboratory of Epidemiology and Population SciencesNational Institute on Aging, NIA/NIH/IRPBaltimoreMarylandUSA
| | - Michele K. Evans
- Laboratory of Epidemiology and Population SciencesNational Institute on Aging, NIA/NIH/IRPBaltimoreMarylandUSA
| | - Alan B. Zonderman
- Laboratory of Epidemiology and Population SciencesNational Institute on Aging, NIA/NIH/IRPBaltimoreMarylandUSA
| |
Collapse
|
10
|
Zhang Y, Guo Y, He Y, You J, Zhang Y, Wang L, Chen S, He X, Yang L, Huang Y, Kang J, Ge Y, Dong Q, Feng J, Cheng W, Yu J. Large-scale proteomic analyses of incident Alzheimer's disease reveal new pathophysiological insights and potential therapeutic targets. Mol Psychiatry 2024:10.1038/s41380-024-02840-x. [PMID: 39562718 DOI: 10.1038/s41380-024-02840-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 11/07/2024] [Accepted: 11/08/2024] [Indexed: 11/21/2024]
Abstract
Pathophysiological evolutions in early-stage Alzheimer's disease (AD) are not well understood. We used data of 2923 Olink plasma proteins from 51,296 non-demented middle-aged adults. During a follow-up of 15 years, 689 incident AD cases occurred. Cox-proportional hazard models were applied to identify AD-associated proteins in different time intervals. Through linking to protein categories, changing sequences of protein z-scores can reflect pathophysiological evolutions. Mendelian randomization using blood protein quantitative loci data provided causal evidence for potentially druggable proteins. We identified 48 AD-related proteins, with CEND1, GFAP, NEFL, and SYT1 being top hits in both near-term (HR:1.15-1.77; P:9.11 × 10-65-2.78 × 10-6) and long-term AD risk (HR:1.20-1.54; P:2.43 × 10-21-3.95 × 10-6). These four proteins increased 15 years before AD diagnosis and progressively escalated, indicating early and sustained dysfunction in synapse and neurons. Proteins related to extracellular matrix organization, apoptosis, innate immunity, coagulation, and lipid homeostasis showed early disturbances, followed by malfunctions in metabolism, adaptive immunity, and final synaptic and neuronal loss. Combining CEND1, GFAP, NEFL, and SYT1 with demographics generated desirable predictions for 10-year (AUC = 0.901) and over-10-year AD (AUC = 0.864), comparable to full model. Mendelian randomization supports potential genetic link between CEND1, SYT1, and AD as outcome. Our findings highlight the importance of exploring the pathophysiological evolutions in early stages of AD, which is essential for the development of early biomarkers and precision therapeutics.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu Guo
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu He
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jia You
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - YaRu Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - LinBo Wang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
| | - ShiDong Chen
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - XiaoYu He
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liu Yang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - YuYuan Huang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - JuJiao Kang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
| | - YiJun Ge
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - JianFeng Feng
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Wei Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China.
| | - JinTai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
11
|
Lee KH, Assassi S, Mohan C, Pedroza C. Addressing statistical challenges in the analysis of proteomics data with extremely small sample size: a simulation study. BMC Genomics 2024; 25:1086. [PMID: 39543503 PMCID: PMC11566501 DOI: 10.1186/s12864-024-11018-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 11/08/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND One of the most promising approaches for early and more precise disease prediction and diagnosis is through the inclusion of proteomics data augmented with clinical data. Clinical proteomics data is often characterized by its high dimensionality and extremely limited sample size, posing a significant challenge when employing machine learning techniques for extracting only the most relevant information. Although there is a wide array of statistical techniques and numerous analysis pipelines employed in proteomics data analysis, it is unclear which of these methods produce the most efficient, reproducible, and clinically meaningful results. RESULTS In this study, we compared 9 unique analysis schemes comprised of different machine learning and dimensionality reduction methods for the analysis of simulated proteomics data consisting of 1317 proteins measured in 26 subjects (i.e., 13 controls and 13 cases). In scenarios where the sample size is extremely small (i.e., n < 30), all schemes resulted in an exceptionally high level of performance metrics, indicating potential overfitting. While performance metrics did not exhibit significant differences across schemes, the set of proteins selected to be discriminatory between groups demonstrated a substantial level of heterogeneity. However, despite heterogeneity in the selected proteins, their biological pathways and genetic diseases exhibited similarities. A sensitivity analysis conducted using varying sample sizes indicated that the stability of a set of selected biomarkers improves with larger sample sizes within a scheme. CONCLUSIONS When the aim of the study is to identify a statistical model that best distinguishes between cohort groups using proteomics data and to uncover the biological pathways and disorders common among the selected proteins, the majority of widely used analysis pipelines perform similarly. However, if the main objective is to pinpoint a set of selected proteins that wield significant influence in discriminating cohort groups and utilize them for subsequent investigations, meticulous consideration is necessary when opting for statistical models, due to the possibility of heterogeneity in the sets of selected proteins.
Collapse
Affiliation(s)
- Kyung Hyun Lee
- Institute for Clinical Research and Learning Health Care, Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Shervin Assassi
- Department of Internal Medicine - Rheumatology, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Claudia Pedroza
- Institute for Clinical Research and Learning Health Care, Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
12
|
Cristiani CM, Calomino C, Scaramuzzino L, Murfuni MS, Parrotta EI, Bianco MG, Cuda G, Quattrone A, Quattrone A. Proximity Elongation Assay and ELISA for the Identification of Serum Diagnostic Biomarkers in Parkinson's Disease and Progressive Supranuclear Palsy. Int J Mol Sci 2024; 25:11663. [PMID: 39519214 PMCID: PMC11546529 DOI: 10.3390/ijms252111663] [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: 10/01/2024] [Revised: 10/27/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Clinical differentiation of progressive supranuclear palsy (PSP) from Parkinson's disease (PD) is challenging due to overlapping phenotypes and late onset of PSP specific symptoms, highlighting the need for easily assessable biomarkers. We used proximity elongation assay (PEA) to analyze 460 proteins in serum samples from 46 PD, 30 PSP patients, and 24 healthy controls. ANCOVA was used to identify the most promising proteins and machine learning (ML) XGBoost and random forest algorithms to assess their classification performance. Promising proteins were also quantified by ELISA. Moreover, correlations between serum biomarkers and biological and clinical features were investigated. We identified five proteins (TFF3, CPB1, OPG, CNTN1, TIMP4) showing different levels between PSP and PD, which achieved good performance (AUC: 0.892) when combined by ML. On the other hand, when the three most significant biomarkers (TFF3, CPB1 and OPG) were analyzed by ELISA, there was no difference between groups. Serum levels of TFF3 positively correlated with age in all subjects' groups, while for OPG and CPB1 such a correlation occurred in PSP patients only. Moreover, CPB1 positively correlated with disease severity in PD, while no correlations were observed in the PSP group. Overall, we identified CPB1 correlating with PD severity, which may support clinical staging of PD. In addition, our results showing discrepancy between PEA and ELISA technology suggest that caution should be used when translating proteomic findings into clinical practice.
Collapse
Affiliation(s)
| | - Camilla Calomino
- Neuroscience Research Center, University “Magna Graecia”, 88100 Catanzaro, Italy
| | - Luana Scaramuzzino
- Neuroscience Research Center, University “Magna Graecia”, 88100 Catanzaro, Italy
| | - Maria Stella Murfuni
- Research Centre for Advanced Biochemistry and Molecular Biology, Department of Clinical and Experimental Medicine, University “Magna Graecia”, 88100 Catanzaro, Italy
| | - Elvira Immacolata Parrotta
- Institute of Molecular Biology, Department of Medical and Surgical Sciences, University “Magna Graecia”, 88100 Catanzaro, Italy
| | | | - Giovanni Cuda
- Research Centre for Advanced Biochemistry and Molecular Biology, Department of Clinical and Experimental Medicine, University “Magna Graecia”, 88100 Catanzaro, Italy
| | - Aldo Quattrone
- Neuroscience Research Center, University “Magna Graecia”, 88100 Catanzaro, Italy
| | - Andrea Quattrone
- Neuroscience Research Center, University “Magna Graecia”, 88100 Catanzaro, Italy
- Institute of Neurology, Department of Medical and Surgical Sciences, University “Magna Graecia”, 88100 Catanzaro, Italy
| |
Collapse
|
13
|
Beydoun HA, Beydoun MA, Noren Hooten N, Weiss J, Li Z, Georgescu MF, Maino Vieytes CA, Meirelles O, Launer LJ, Evans MK, Zonderman AB. Mediating and moderating effects of plasma proteomic biomarkers on the association between poor oral health problems and incident dementia: The UK Biobank study. GeroScience 2024; 46:5343-5363. [PMID: 38809392 PMCID: PMC11336161 DOI: 10.1007/s11357-024-01202-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 05/12/2024] [Indexed: 05/30/2024] Open
Abstract
The plasma proteome can mediate poor oral health problems (POHP)'s link to incident dementia. We screened 37,269 UK Biobank participants 50-74 years old (2006-2010) for prevalent POHP, further tested against 1463 plasma proteins and incident dementia over up to 15 years of follow-up. Total effect (TE) of POHP-dementia through plasma proteomic markers was decomposed into pure indirect effect (PIE), interaction referent (INTREF), controlled direct effect (CDE), or mediated interaction (INTMED). POHP increased the risk of all-cause dementia by 17% (P < 0.05). Growth differentiation factor 15 (GDF15) exhibited the strongest mediating effects (PIE > 0, P < 0.001), explaining 28% the total effect of POHP on dementia, as a pure indirect effect. A first principal component encompassing top 4 mediators (GDF15, IL19, MMP12, and ACVRL1), explained 11% of the POHP-dementia effect as a pure indirect effect. Pathway analysis including all mediators (k = 173 plasma proteins) revealed the involvement of the immune system, signal transduction, metabolism, disease, and gene expression, while STRING analysis indicated that top mediators within the first principal component were also represented in the two largest proteomic clusters. The dominant biological GO pathway for the GDF15 cluster was GO:0007169 labeled as "transmembrane receptor protein tyrosine kinase signaling pathway." Dementia is linked to POHP mediated by GDF15 among several proteomic markers.
Collapse
Affiliation(s)
- Hind A Beydoun
- US Department of Veterans Affairs, VA National Center On Homelessness Among Veterans, Washington, DC, 20420, USA
- Department of Management, Policy, and Community Health, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - May A Beydoun
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA.
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Jordan Weiss
- Stanford Center on Longevity, Stanford University, Palo Alto, CA, 94305, USA
| | - Zhiguang Li
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Michael F Georgescu
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Christian A Maino Vieytes
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Osorio Meirelles
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| |
Collapse
|
14
|
Green ZD, John CS, Kueck PJ, Blankenship AE, Kemna RE, Johnson CN, Yoksh LE, Best SR, Donald JS, Mahnken JD, Burns JM, Vidoni ED, Morris JK. Acute exercise alters brain glucose metabolism in aging and Alzheimer's disease. J Physiol 2024. [PMID: 39258961 DOI: 10.1113/jp286923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/22/2024] [Indexed: 09/12/2024] Open
Abstract
There is evidence that aerobic exercise improves brain health. Benefits may be modulated by acute physiological responses to exercise, but this has not been well characterized in older or cognitively impaired adults. The randomized controlled trial 'AEROBIC' (NCT04299308) enrolled 60 older adults who were cognitively healthy (n = 30) or cognitively impaired (n = 30) to characterize the acute brain responses to moderate [45-55% heart rate reserve (HRR)] and higher (65-75% HRR) intensity acute exercise. Each participant received two fluorodeoxyglucose positron emission tomography (FDG-PET) scans, one at rest and one following acute exercise. Change in cerebral glucose metabolism from rest to exercise was the primary outcome. Blood biomarker responses were also characterized as secondary outcomes. Whole grey matter FDG-PET standardized uptake value ratio (SUVR) differed between exercise (1.045 ± 0.082) and rest (0.985 ± 0.077) across subjects [Diff = -0.060, t(58) = 13.8, P < 0.001] regardless of diagnosis. Exercise increased lactate area under the curve (AUC) [F(1,56) = 161.99, P < 0.001] more in the higher intensity group [mean difference (MD) = 97.0 ± 50.8] than the moderate intensity group (MD = 40.3 ± 27.5; t = -5.252, P < 0.001). Change in lactate AUC and FDG-PET SUVR correlated significantly (R2 = 0.179, P < 0.001). Acute exercise decreased whole grey matter cerebral glucose metabolism. This effect tracked with the systemic lactate response, suggesting that lactate may serve as a key brain fuel during exercise. Direct measurements of brain lactate metabolism in response to exercise are warranted. KEY POINTS: Acute exercise is associated with a drop in global brain glucose metabolism in both cognitively healthy older adults and those with Alzheimer's disease. Blood lactate levels increase following acute exercise. Change in brain metabolism tracks with blood lactate, suggesting it may be an important brain fuel. Acute exercise stimulates changes in brain-derived neurotrophic factor and other blood biomarkers.
Collapse
Affiliation(s)
- Zachary D Green
- University of Kansas Alzheimer's Disease Research Center, Fairway, KS, USA
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Casey S John
- University of Kansas Alzheimer's Disease Research Center, Fairway, KS, USA
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Paul J Kueck
- University of Kansas Alzheimer's Disease Research Center, Fairway, KS, USA
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Anneka E Blankenship
- University of Kansas Alzheimer's Disease Research Center, Fairway, KS, USA
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Riley E Kemna
- University of Kansas Alzheimer's Disease Research Center, Fairway, KS, USA
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Chelsea N Johnson
- University of Kansas Alzheimer's Disease Research Center, Fairway, KS, USA
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Lauren E Yoksh
- Department of Radiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Shaun R Best
- Department of Radiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Joseph S Donald
- Department of Radiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jonathan D Mahnken
- University of Kansas Alzheimer's Disease Research Center, Fairway, KS, USA
- Department of Radiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jeffrey M Burns
- University of Kansas Alzheimer's Disease Research Center, Fairway, KS, USA
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Eric D Vidoni
- University of Kansas Alzheimer's Disease Research Center, Fairway, KS, USA
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jill K Morris
- University of Kansas Alzheimer's Disease Research Center, Fairway, KS, USA
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
15
|
Beydoun MA, Beydoun HA, Hu YH, Maino Vieytes CA, Noren Hooten N, Song M, Georgescu MF, Fanelli-Kuczmarski MT, Meirelles O, Launer LJ, Evans MK, Zonderman AB. Plasma proteomic biomarkers and the association between poor cardiovascular health and incident dementia: The UK Biobank study. Brain Behav Immun 2024; 119:995-1007. [PMID: 38710337 PMCID: PMC11285716 DOI: 10.1016/j.bbi.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/04/2024] [Accepted: 05/02/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND The study examined how plasma proteome indicators may explain the link between poor cardiovascular health (CVH) and dementia risk. METHODS The present study involved 28,974 UK Biobank participants aged 50-74y at baseline (2006-2010) who were followed-up for ≤ 15 y for incidence of dementia. CVH was calculated using Life's Essential 8 (LE8) total scores. The scores were standardized and reverse coded to reflect poor CVH (LE8z_rev). OLINK proteomics was available on this sample (k = 1,463 plasma proteins). The study primarily tested the mediating effects of the plasma proteome in LE8z_rev-dementia effect. The total effect was decomposed into "mediation only" or pure indirect effect (PIE), "interaction only" or interaction referent (INTREF), "neither mediation nor interaction" or controlled direct effect (CDE), and "both mediation and interaction" or mediated interaction (INTMED). RESULTS The study found poorer CVH assessed by LE8z_rev increased the risk of all-cause dementia by 11 % [per 1 SD, hazard ratio, (HR) = 1.11, 95 % CI: 1.03-1.20, p = 0.005). The study identified 11 plasma proteins with strong mediating effects, with GDF15 having the strongest association with dementia risk (per 1 SD, HR = 1.24, 95 % CI: 1.16, 1.33, P < 0.001 when LE8z_rev is set at its mean value) and the largest proportion mediated combining PIE and INTMED (62.6 %; 48 % of TE is PIE), followed by adrenomedullin or ADM. A first principal component with 10 top mediators (TNFRSF1A, GDF15, FSTL3, COL6A3, PLAUR, ADM, GFRAL, ACVRL1, TNFRSF6B, TGFA) mediated 53.6 % of the LE8z_rev-dementia effect. Using all the significant PIE (k = 526) proteins, we used OLINK Insight pathway analysis to identify key pathways, which revealed the involvement of the immune system, signal transduction, metabolism, disease, protein metabolism, hemostasis, membrane trafficking, extracellular matrix organization, developmental biology, and gene expression among others. STRING analysis revealed that five top consistent proteomic mediators were represented in two larger clusters reflecting numerous interconnected biological gene ontology pathways, most notably cytokine-mediated signaling pathway for GDF15 cluster (GO:0019221) and regulation of peptidyl-tyrosine phosphorylation for the ADM cluster (GO:0050730). CONCLUSION Dementia is linked to poor CVH mediated by GDF15 and ADM among several key proteomic markers which collectively explained ∼ 54 % of the total effect.
Collapse
Affiliation(s)
- May A Beydoun
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States.
| | - Hind A Beydoun
- VA National Center on Homelessness Among Veterans, U.S. Department of Veterans Affairs, Washington, DC 20420, United States; Department of Management, Policy, and Community Health, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX 77030, United States
| | - Yi-Han Hu
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States
| | - Christian A Maino Vieytes
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States
| | - Minkyo Song
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States
| | - Michael F Georgescu
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States
| | - Marie T Fanelli-Kuczmarski
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States
| | - Osorio Meirelles
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States
| | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States
| |
Collapse
|
16
|
Beuthner BE, Elkenani M, Evert K, Mustroph J, Jacob CF, Paul NB, Beißbarth T, Zeisberg EM, Schnelle M, Puls M, Hasenfuß G, Toischer K. Histological assessment of cardiac amyloidosis in patients undergoing transcatheter aortic valve replacement. ESC Heart Fail 2024; 11:1636-1646. [PMID: 38407567 PMCID: PMC11098657 DOI: 10.1002/ehf2.14709] [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: 06/27/2023] [Revised: 11/28/2023] [Accepted: 01/19/2024] [Indexed: 02/27/2024] Open
Abstract
AIMS Studies have reported a strongly varying co-prevalence of aortic stenosis (AS) and cardiac amyloidosis (CA). We sought to histologically determine the co-prevalence of AS and CA in patients undergoing transcatheter aortic valve replacement (TAVR). Consequently, we aimed to derive an algorithm to identify cases in which to suspect the co-prevalence of AS and CA. METHODS AND RESULTS In this prospective, monocentric study, endomyocardial biopsies of 162 patients undergoing TAVR between January 2017 and March 2021 at the University Medical Centre Göttingen were analysed by one pathologist blinded to clinical data using haematoxylin-eosin staining, Elastica van Gieson staining, and Congo red staining of endomyocardial biopsies. CA was identified in only eight patients (4.9%). CA patients had significantly higher N-terminal pro-brain natriuretic peptide (NT-proBNP) levels (4356.20 vs. 1938.00 ng/L, P = 0.034), a lower voltage-to-mass ratio (0.73 vs. 1.46 × 10-2 mVm2/g, P = 0.022), and lower transaortic gradients (Pmean 17.5 vs. 38.0 mmHg, P = 0.004) than AS patients. Concomitant CA was associated with a higher prevalence of post-procedural acute kidney injury (50.0% vs. 13.1%, P = 0.018) and sudden cardiac death [SCD; P (log-rank test) = 0.017]. Following propensity score matching, 184 proteins were analysed to identify serum biomarkers of concomitant CA. CA patients expressed lower levels of chymotrypsin (P = 0.018) and carboxypeptidase 1 (P = 0.027). We propose an algorithm using commonly documented parameters-stroke volume index, ejection fraction, NT-proBNP levels, posterior wall thickness, and QRS voltage-to-mass ratio-to screen for CA in AS patients, reaching a sensitivity of 66.6% with a specificity of 98.1%. CONCLUSIONS The co-prevalence of AS and CA was lower than expected, at 4.9%. Despite excellent 1 year mortality, AS + CA patients died significantly more often from SCD. We propose a multimodal algorithm to facilitate more effective screening for CA containing parameters commonly documented during clinical routine. Proteomic biomarkers may yield additional information in the future.
Collapse
Affiliation(s)
- Bo Eric Beuthner
- Department of Cardiology and PneumologyUniversity Medical Centre Göttingen, Georg August University of GöttingenRobert‐Koch‐Straße 4037075GöttingenGermany
- German Centre for Cardiovascular Research (DZHK), partner site GöttingenGöttingenGermany
| | - Manar Elkenani
- Department of Cardiology and PneumologyUniversity Medical Centre Göttingen, Georg August University of GöttingenRobert‐Koch‐Straße 4037075GöttingenGermany
- German Centre for Cardiovascular Research (DZHK), partner site GöttingenGöttingenGermany
| | - Katja Evert
- Institute of PathologyUniversity of RegensburgRegensburgGermany
| | - Julian Mustroph
- Department of Internal Medicine IIUniversity Medical Centre RegensburgRegensburgGermany
| | - Christoph Friedemann Jacob
- Department of Cardiology and PneumologyUniversity Medical Centre Göttingen, Georg August University of GöttingenRobert‐Koch‐Straße 4037075GöttingenGermany
- German Centre for Cardiovascular Research (DZHK), partner site GöttingenGöttingenGermany
| | - Niels Benjamin Paul
- Department of Cardiology and PneumologyUniversity Medical Centre Göttingen, Georg August University of GöttingenRobert‐Koch‐Straße 4037075GöttingenGermany
- Department of Medical BioinformaticsUniversity Medical Centre Göttingen, Georg August University of GöttingenGöttingenGermany
| | - Tim Beißbarth
- Department of Medical BioinformaticsUniversity Medical Centre Göttingen, Georg August University of GöttingenGöttingenGermany
| | - Elisabeth Maria Zeisberg
- Department of Cardiology and PneumologyUniversity Medical Centre Göttingen, Georg August University of GöttingenRobert‐Koch‐Straße 4037075GöttingenGermany
- German Centre for Cardiovascular Research (DZHK), partner site GöttingenGöttingenGermany
| | - Moritz Schnelle
- German Centre for Cardiovascular Research (DZHK), partner site GöttingenGöttingenGermany
- Department of Clinical ChemistryUniversity Medical Centre Göttingen, Georg August University of GöttingenGöttingenGermany
| | - Miriam Puls
- Department of Cardiology and PneumologyUniversity Medical Centre Göttingen, Georg August University of GöttingenRobert‐Koch‐Straße 4037075GöttingenGermany
- German Centre for Cardiovascular Research (DZHK), partner site GöttingenGöttingenGermany
| | - Gerd Hasenfuß
- Department of Cardiology and PneumologyUniversity Medical Centre Göttingen, Georg August University of GöttingenRobert‐Koch‐Straße 4037075GöttingenGermany
- German Centre for Cardiovascular Research (DZHK), partner site GöttingenGöttingenGermany
| | - Karl Toischer
- Department of Cardiology and PneumologyUniversity Medical Centre Göttingen, Georg August University of GöttingenRobert‐Koch‐Straße 4037075GöttingenGermany
- German Centre for Cardiovascular Research (DZHK), partner site GöttingenGöttingenGermany
| |
Collapse
|
17
|
Guo Y, You J, Zhang Y, Liu WS, Huang YY, Zhang YR, Zhang W, Dong Q, Feng JF, Cheng W, Yu JT. Plasma proteomic profiles predict future dementia in healthy adults. NATURE AGING 2024; 4:247-260. [PMID: 38347190 DOI: 10.1038/s43587-023-00565-0] [Citation(s) in RCA: 73] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 12/22/2023] [Indexed: 02/22/2024]
Abstract
The advent of proteomics offers an unprecedented opportunity to predict dementia onset. We examined this in data from 52,645 adults without dementia in the UK Biobank, with 1,417 incident cases and a follow-up time of 14.1 years. Of 1,463 plasma proteins, GFAP, NEFL, GDF15 and LTBP2 consistently associated most with incident all-cause dementia (ACD), Alzheimer's disease (AD) and vascular dementia (VaD), and ranked high in protein importance ordering. Combining GFAP (or GDF15) with demographics produced desirable predictions for ACD (area under the curve (AUC) = 0.891) and AD (AUC = 0.872) (or VaD (AUC = 0.912)). This was also true when predicting over 10-year ACD, AD and VaD. Individuals with higher GFAP levels were 2.32 times more likely to develop dementia. Notably, GFAP and LTBP2 were highly specific for dementia prediction. GFAP and NEFL began to change at least 10 years before dementia diagnosis. Our findings strongly highlight GFAP as an optimal biomarker for dementia prediction, even more than 10 years before the diagnosis, with implications for screening people at high risk for dementia and for early intervention.
Collapse
Affiliation(s)
- Yu Guo
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jia You
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
| | - Yi Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei-Shi Liu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu-Yuan Huang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ya-Ru Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Zhang
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China.
| | - Wei Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China.
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
18
|
Kerminen H, Marzetti E, D’Angelo E. Biological and Physical Performance Markers for Early Detection of Cognitive Impairment in Older Adults. J Clin Med 2024; 13:806. [PMID: 38337499 PMCID: PMC10856537 DOI: 10.3390/jcm13030806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/26/2024] [Accepted: 01/28/2024] [Indexed: 02/12/2024] Open
Abstract
Dementia is a major cause of poor quality of life, disability, and mortality in old age. According to the geroscience paradigm, the mechanisms that drive the aging process are also involved in the pathogenesis of chronic degenerative diseases, including dementia. The dissection of such mechanisms is therefore instrumental in providing biological targets for interventions and new sources for biomarkers. Within the geroscience paradigm, several biomarkers have been discovered that can be measured in blood and that allow early identification of individuals at risk of cognitive impairment. Examples of such markers include inflammatory biomolecules, markers of neuroaxonal damage, extracellular vesicles, and DNA methylation. Furthermore, gait speed, measured at a usual and fast pace and as part of a dual task, has been shown to detect individuals at risk of future dementia. Here, we provide an overview of available biomarkers that may be used to gauge the risk of cognitive impairment in apparently healthy older adults. Further research should establish which combination of biomarkers possesses the highest predictive accuracy toward incident dementia. The implementation of currently available markers may allow the identification of a large share of at-risk individuals in whom preventive interventions should be implemented to maintain or increase cognitive reserves, thereby reducing the risk of progression to dementia.
Collapse
Affiliation(s)
- Hanna Kerminen
- Faculty of Medicine and Health Technology, Gerontology Research Center (GEREC), Tampere University, Arvo Ylpön katu 34, 33520 Tampere, Finland;
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy
| | - Emanuele Marzetti
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy;
| | - Emanuela D’Angelo
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy;
| |
Collapse
|
19
|
Roberts JA, Basu-Roy S, Shin J, Varma VR, Williamson A, Blackshear C, Griswold ME, Candia J, Elango P, Karikkineth AC, Tanaka T, Ferrucci L, Thambisetty M. Serum Proteomic Signatures of Common Health Outcomes among Older Adults. Gerontology 2024; 70:269-278. [PMID: 38219723 DOI: 10.1159/000534753] [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: 04/30/2023] [Accepted: 10/09/2023] [Indexed: 01/16/2024] Open
Abstract
INTRODUCTION In aging populations, the coexistence of multiple health comorbidities represents a significant challenge for clinicians and researchers. Leveraging advances in omics techniques to characterize these health conditions may provide insight into disease pathogenesis as well as reveal biomarkers for monitoring, prognostication, and diagnosis. Researchers have previously established the utility of big data approaches with respect to comprehensive health outcome measurements in younger populations, identifying protein markers that may provide significant health information with a single blood sample. METHODS Here, we employed a similar approach in two cohorts of older adults, the Baltimore Longitudinal Study of Aging (mean age = 76.12 years) and InCHIANTI Study (mean age = 66.05 years), examining the relationship between levels of serum proteins and 5 key health outcomes: kidney function, fasting glucose, physical activity, lean body mass, and percent body fat. RESULTS Correlations between proteins and health outcomes were primarily shared across both older adult cohorts. We further identified that most proteins associated with health outcomes in the older adult cohorts were not associated with the same outcomes in a prior study of a younger population. A subset of proteins, adiponectin, MIC-1, and NCAM-120, were associated with at least three health outcomes in both older adult cohorts but not in the previously published younger cohort, suggesting that they may represent plausible markers of general health in older adult populations. CONCLUSION Taken together, these findings suggest that comprehensive protein health markers have utility in aging populations and are distinct from those identified in younger adults, indicating unique mechanisms of disease with aging.
Collapse
Affiliation(s)
- Jackson A Roberts
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA,
- Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA,
| | - Sayantani Basu-Roy
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Jong Shin
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Vijay R Varma
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Andrew Williamson
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Chad Blackshear
- University of Mississippi Medical Center, Jackson, Mississippi, USA
| | | | - Julián Candia
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Palchamy Elango
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Ajoy C Karikkineth
- Clinical Research Core, National Institute on Aging, National Institutes of Health Intramural Research Program, Baltimore, Maryland, USA
| | - Toshiko Tanaka
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Madhav Thambisetty
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| |
Collapse
|
20
|
You J, Guo Y, Zhang Y, Kang JJ, Wang LB, Feng JF, Cheng W, Yu JT. Plasma proteomic profiles predict individual future health risk. Nat Commun 2023; 14:7817. [PMID: 38016990 PMCID: PMC10684756 DOI: 10.1038/s41467-023-43575-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 11/13/2023] [Indexed: 11/30/2023] Open
Abstract
Developing a single-domain assay to identify individuals at high risk of future events is a priority for multi-disease and mortality prevention. By training a neural network, we developed a disease/mortality-specific proteomic risk score (ProRS) based on 1461 Olink plasma proteins measured in 52,006 UK Biobank participants. This integrative score markedly stratified the risk for 45 common conditions, including infectious, hematological, endocrine, psychiatric, neurological, sensory, circulatory, respiratory, digestive, cutaneous, musculoskeletal, and genitourinary diseases, cancers, and mortality. The discriminations witnessed high accuracies achieved by ProRS for 10 endpoints (e.g., cancer, dementia, and death), with C-indexes exceeding 0.80. Notably, ProRS produced much better or equivalent predictive performance than established clinical indicators for almost all endpoints. Incorporating clinical predictors with ProRS enhanced predictive power for most endpoints, but this combination only exhibited limited improvement when compared to ProRS alone. Some proteins, e.g., GDF15, exhibited important discriminative values for various diseases. We also showed that the good discriminative performance observed could be largely translated into practical clinical utility. Taken together, proteomic profiles may serve as a replacement for complex laboratory tests or clinical measures to refine the comprehensive risk assessments of multiple diseases and mortalities simultaneously. Our models were internally validated in the UK Biobank; thus, further independent external validations are necessary to confirm our findings before application in clinical settings.
Collapse
Affiliation(s)
- Jia You
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yu Guo
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yi Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Ju-Jiao Kang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Lin-Bo Wang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Jian-Feng Feng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China.
- Zhangjiang Fudan International Innovation Center, Shanghai, China.
- School of Data Science, Fudan University, Shanghai, China.
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence, Zhejiang Normal University, Zhejiang, China.
| | - Wei Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China.
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence, Zhejiang Normal University, Zhejiang, China.
- Shanghai Medical College and Zhongshan Hospital Immunotherapy Technology Transfer Center, Shanghai, China.
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| |
Collapse
|
21
|
Wang W, Zhong Y, Zhou Y, Yu Y, Li J, Kang S, Ma Z, Fan X, Sun L, Tang L. Low-intensity pulsed ultrasound mitigates cognitive impairment by inhibiting muscle atrophy in hindlimb unloaded mice. THE JOURNAL OF THE ACOUSTICAL SOCIETY OF AMERICA 2023; 154:1427-1438. [PMID: 37672304 DOI: 10.1121/10.0020835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/14/2023] [Indexed: 09/07/2023]
Abstract
Microgravity leads to muscle loss, usually accompanied by cognitive impairment. Muscle reduction was associated with the decline of cognitive ability. Our previous studies showed that low-intensity pulsed ultrasound (LIPUS) promoted muscle hypertrophy and prevented muscle atrophy. This study aims to verify whether LIPUS can improve cognitive impairment by preventing muscle atrophy in hindlimb unloaded mice. In this study, mice were randomly divided into normal control (NC), hindlimb unloading (HU), hindlimb unloading + LIPUS (HU+LIPUS) groups. The mice in the HU+LIPUS group received a 30 mW/cm2 LIPUS irradiation on gastrocnemius for 20 min/d. After 21 days, LIPUS significantly prevented the decrease in muscle mass and strength caused by tail suspension. The HU+LIPUS mice showed an enhanced desire to explore unfamiliar environments and their spatial learning and memory abilities, enabling them to quickly identify differences between different objects, as well as their social discrimination abilities. MSTN is a negative regulator of muscle growth and also plays a role in regulating cognition. LIPUS significantly inhibited MSTN expression in skeletal muscle and serum and its receptor ActRIIB expression in brain, upregulated AKT and BDNF expression in brain. Taken together, LIPUS may improve the cognitive dysfunction in hindlimb unloaded rats by inhibiting muscle atrophy through MSTN/AKT/BDNF pathway.
Collapse
Affiliation(s)
- Wanzhao Wang
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Yi Zhong
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Yaling Zhou
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Yanan Yu
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Jiaxiang Li
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Sufang Kang
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Zhanke Ma
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Xiushan Fan
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Lijun Sun
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Liang Tang
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| |
Collapse
|
22
|
Duperron MG, Knol MJ, Le Grand Q, Evans TE, Mishra A, Tsuchida A, Roshchupkin G, Konuma T, Trégouët DA, Romero JR, Frenzel S, Luciano M, Hofer E, Bourgey M, Dueker ND, Delgado P, Hilal S, Tankard RM, Dubost F, Shin J, Saba Y, Armstrong NJ, Bordes C, Bastin ME, Beiser A, Brodaty H, Bülow R, Carrera C, Chen C, Cheng CY, Deary IJ, Gampawar PG, Himali JJ, Jiang J, Kawaguchi T, Li S, Macalli M, Marquis P, Morris Z, Muñoz Maniega S, Miyamoto S, Okawa M, Paradise M, Parva P, Rundek T, Sargurupremraj M, Schilling S, Setoh K, Soukarieh O, Tabara Y, Teumer A, Thalamuthu A, Trollor JN, Valdés Hernández MC, Vernooij MW, Völker U, Wittfeld K, Wong TY, Wright MJ, Zhang J, Zhao W, Zhu YC, Schmidt H, Sachdev PS, Wen W, Yoshida K, Joutel A, Satizabal CL, Sacco RL, Bourque G, Lathrop M, Paus T, Fernandez-Cadenas I, Yang Q, Mazoyer B, Boutinaud P, Okada Y, Grabe HJ, Mather KA, Schmidt R, Joliot M, Ikram MA, Matsuda F, Tzourio C, Wardlaw JM, Seshadri S, Adams HHH, Debette S. Genomics of perivascular space burden unravels early mechanisms of cerebral small vessel disease. Nat Med 2023; 29:950-962. [PMID: 37069360 PMCID: PMC10115645 DOI: 10.1038/s41591-023-02268-w] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 02/15/2023] [Indexed: 04/19/2023]
Abstract
Perivascular space (PVS) burden is an emerging, poorly understood, magnetic resonance imaging marker of cerebral small vessel disease, a leading cause of stroke and dementia. Genome-wide association studies in up to 40,095 participants (18 population-based cohorts, 66.3 ± 8.6 yr, 96.9% European ancestry) revealed 24 genome-wide significant PVS risk loci, mainly in the white matter. These were associated with white matter PVS already in young adults (N = 1,748; 22.1 ± 2.3 yr) and were enriched in early-onset leukodystrophy genes and genes expressed in fetal brain endothelial cells, suggesting early-life mechanisms. In total, 53% of white matter PVS risk loci showed nominally significant associations (27% after multiple-testing correction) in a Japanese population-based cohort (N = 2,862; 68.3 ± 5.3 yr). Mendelian randomization supported causal associations of high blood pressure with basal ganglia and hippocampal PVS, and of basal ganglia PVS and hippocampal PVS with stroke, accounting for blood pressure. Our findings provide insight into the biology of PVS and cerebral small vessel disease, pointing to pathways involving extracellular matrix, membrane transport and developmental processes, and the potential for genetically informed prioritization of drug targets.
Collapse
Affiliation(s)
- Marie-Gabrielle Duperron
- Bordeaux Population Health Research Center, UMR 1219, University of Bordeaux, Inserm, Bordeaux, France
- Department of Neurology, Institute of Neurodegenerative Diseases, Bordeaux University Hospital, Bordeaux, France
| | - Maria J Knol
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Quentin Le Grand
- Bordeaux Population Health Research Center, UMR 1219, University of Bordeaux, Inserm, Bordeaux, France
| | - Tavia E Evans
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Aniket Mishra
- Bordeaux Population Health Research Center, UMR 1219, University of Bordeaux, Inserm, Bordeaux, France
| | - Ami Tsuchida
- Bordeaux Population Health Research Center, UMR 1219, University of Bordeaux, Inserm, Bordeaux, France
- Groupe d'Imagerie Neurofonctionelle - Institut des maladies neurodégénératives (GIN-IMN), UMR 5293, University of Bordeaux, CNRS, CEA, Bordeaux, France
| | - Gennady Roshchupkin
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Takahiro Konuma
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
| | - David-Alexandre Trégouët
- Bordeaux Population Health Research Center, UMR 1219, University of Bordeaux, Inserm, Bordeaux, France
| | - Jose Rafael Romero
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- The Framingham Heart Study, Framingham, MA, USA
| | - Stefan Frenzel
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | | | - Edith Hofer
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, Graz, Austria
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, Graz, Austria
| | - Mathieu Bourgey
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine at McGill University, Montreal, Quebec, Canada
- Canadian Centre for Computational Genomics, McGill University, Montreal, Quebec, Canada
| | - Nicole D Dueker
- John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, USA
| | - Pilar Delgado
- Institut de Recerca Vall d'hebron, Neurovascular Research Lab, Universitat Autònoma de Barcelona, Barcelona, Spain
- Hospital Universitari Vall d'Hebron, Neurology Department, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Saima Hilal
- Memory Aging and Cognition Center, National University Health System, Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
| | - Rick M Tankard
- Department of Mathematics and Statistics, Curtin University, Perth, Western Australia, Australia
| | - Florian Dubost
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
- Department of Medical Informatics, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Jean Shin
- The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
- Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Yasaman Saba
- Bordeaux Population Health Research Center, UMR 1219, University of Bordeaux, Inserm, Bordeaux, France
- Institute for Molecular Biology & Biochemistry, Gottfried Schatz Research Center (for Cell Signaling, Metabolism and Aging), Medical University of Graz, Graz, Austria
| | - Nicola J Armstrong
- Department of Mathematics and Statistics, Curtin University, Perth, Western Australia, Australia
| | - Constance Bordes
- Bordeaux Population Health Research Center, UMR 1219, University of Bordeaux, Inserm, Bordeaux, France
| | - Mark E Bastin
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Alexa Beiser
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- The Framingham Heart Study, Framingham, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Henry Brodaty
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry & Mental Health, University of New South Wales, Sydney, New South Wales, Australia
- Dementia Collaborative Research Centre Assessment and Better Care, UNSW, Sydney, New South Wales, Australia
| | - Robin Bülow
- Institute for Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Caty Carrera
- Stroke Pharmacogenomics and Genetics Group, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Christopher Chen
- Memory Aging and Cognition Center, National University Health System, Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ching-Yu Cheng
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Center for Innovation and Precision Eye Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
| | - Ian J Deary
- School of Psychology, University of Edinburgh, Edinburgh, UK
| | - Piyush G Gampawar
- Institute for Molecular Biology & Biochemistry, Gottfried Schatz Research Center (for Cell Signaling, Metabolism and Aging), Medical University of Graz, Graz, Austria
| | - Jayandra J Himali
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- The Framingham Heart Study, Framingham, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA
- Department of Population Health Sciences, UT Health San Antonio, San Antonio, TX, USA
| | - Jiyang Jiang
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry & Mental Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Takahisa Kawaguchi
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shuo Li
- The Framingham Heart Study, Framingham, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Melissa Macalli
- Bordeaux Population Health Research Center, UMR 1219, University of Bordeaux, Inserm, Bordeaux, France
| | - Pascale Marquis
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine at McGill University, Montreal, Quebec, Canada
- Canadian Centre for Computational Genomics, McGill University, Montreal, Quebec, Canada
| | - Zoe Morris
- Neuroimaging, Department of Clinical Neurosciences, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Susana Muñoz Maniega
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute Centre at the University of Edinburgh, Edinburgh, UK
| | | | - Masakazu Okawa
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Matthew Paradise
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry & Mental Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Pedram Parva
- The Framingham Heart Study, Framingham, MA, USA
- Radiology Department, Boston University School of Medicine, Boston, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Tatjana Rundek
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, USA
- Evelyn F. McKnight Brain Institute, Department of Neurology, University of Miami, Miami, FL, USA
| | | | - Sabrina Schilling
- Bordeaux Population Health Research Center, UMR 1219, University of Bordeaux, Inserm, Bordeaux, France
| | - Kazuya Setoh
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Graduate School of Public Health, Shizuoka Graduate University of Public Health, Shizuoka, Japan
| | - Omar Soukarieh
- Bordeaux Population Health Research Center, UMR 1219, University of Bordeaux, Inserm, Bordeaux, France
| | - Yasuharu Tabara
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Graduate School of Public Health, Shizuoka Graduate University of Public Health, Shizuoka, Japan
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Anbupalam Thalamuthu
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry & Mental Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Julian N Trollor
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry & Mental Health, University of New South Wales, Sydney, New South Wales, Australia
- Department of Developmental Disability Neuropsychiatry, UNSW, Sydney, New South Wales, Australia
| | - Maria C Valdés Hernández
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Row Fogo Centre for Research into Ageing and the Brain, University of Edinburgh, Edinburgh, UK
| | - Meike W Vernooij
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Katharina Wittfeld
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Greifswald, Germany
| | - Tien Yin Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Tsinghua Medicine, Tsinghua University, Beijing, China
| | - Margaret J Wright
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
- Centre for Advanced Imaging, University of Queensland, Brisbane, Queensland, Australia
| | - Junyi Zhang
- Department of Neurology, Peking Union Medical College Hospital, Beijing, China
| | - Wanting Zhao
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- The Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore, Singapore
| | - Yi-Cheng Zhu
- Department of Neurology, Peking Union Medical College Hospital, Beijing, China
| | - Helena Schmidt
- Institute for Molecular Biology & Biochemistry, Gottfried Schatz Research Center (for Cell Signaling, Metabolism and Aging), Medical University of Graz, Graz, Austria
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry & Mental Health, University of New South Wales, Sydney, New South Wales, Australia
- Neuropsychiatric Institute, the Prince of Wales Hospital, Sydney, New South Wales, Australia
| | - Wei Wen
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry & Mental Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Kazumichi Yoshida
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Anne Joutel
- Institut de Psychiatrie et Neurosciences de Paris, Université Paris Cité, Inserm, France
| | - Claudia L Satizabal
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- The Framingham Heart Study, Framingham, MA, USA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA
- Department of Population Health Sciences, UT Health San Antonio, San Antonio, TX, USA
| | - Ralph L Sacco
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, USA
- Evelyn F. McKnight Brain Institute, Department of Neurology, University of Miami, Miami, FL, USA
- Department of Public Health Sciences, Miller School of Medicine, University of Miami, Miami, FL, USA
- Department of Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, USA
- Department of Neurosurgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Guillaume Bourque
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine at McGill University, Montreal, Quebec, Canada
- Canadian Centre for Computational Genomics, McGill University, Montreal, Quebec, Canada
| | - Mark Lathrop
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine at McGill University, Montreal, Quebec, Canada
| | - Tomas Paus
- University of Montreal, Faculty of Medicine, Departments of Psychiatry and Neuroscience, Montreal, Quebec, Canada
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
- Centre Hospitalier Universitaire Sainte Justine, Montreal, Quebec, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Israel Fernandez-Cadenas
- Stroke Pharmacogenomics and Genetics Group, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
- Stroke Pharmacogenomics and Genetics Group, Fundació per la Docència i la Recerca Mutua Terrassa, Terrassa, Spain
| | - Qiong Yang
- The Framingham Heart Study, Framingham, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Bernard Mazoyer
- Groupe d'Imagerie Neurofonctionelle - Institut des maladies neurodégénératives (GIN-IMN), UMR 5293, University of Bordeaux, CNRS, CEA, Bordeaux, France
- Bordeaux University Hospital, Bordeaux, France
| | | | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Japan
- Department of Genome Informatics, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Greifswald, Germany
| | - Karen A Mather
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry & Mental Health, University of New South Wales, Sydney, New South Wales, Australia
- Neuroscience Research Australia, Sydney, New South Wales, Australia
| | - Reinhold Schmidt
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, Graz, Austria
| | - Marc Joliot
- Groupe d'Imagerie Neurofonctionelle - Institut des maladies neurodégénératives (GIN-IMN), UMR 5293, University of Bordeaux, CNRS, CEA, Bordeaux, France
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Christophe Tzourio
- Bordeaux Population Health Research Center, UMR 1219, University of Bordeaux, Inserm, Bordeaux, France
- Department of Medical Informatics, Bordeaux University Hospital, Bordeaux, France
| | - Joanna M Wardlaw
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute Centre at the University of Edinburgh, Edinburgh, UK
- Row Fogo Centre for Research into Ageing and the Brain, University of Edinburgh, Edinburgh, UK
| | - Sudha Seshadri
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- The Framingham Heart Study, Framingham, MA, USA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA
- Department of Population Health Sciences, UT Health San Antonio, San Antonio, TX, USA
| | - Hieab H H Adams
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, the Netherlands.
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands.
- Latin American Brain Health (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile.
| | - Stéphanie Debette
- Bordeaux Population Health Research Center, UMR 1219, University of Bordeaux, Inserm, Bordeaux, France.
- Department of Neurology, Institute of Neurodegenerative Diseases, Bordeaux University Hospital, Bordeaux, France.
| |
Collapse
|
23
|
Short MI, Fohner AE, Skjellegrind HK, Beiser A, Gonzales MM, Satizabal CL, Austin TR, Longstreth W, Bis JC, Lopez O, Hveem K, Selbæk G, Larson MG, Yang Q, Aparicio HJ, McGrath ER, Gerszten RE, DeCarli CS, Psaty BM, Vasan RS, Zare H, Seshadri S. Proteome Network Analysis Identifies Potential Biomarkers for Brain Aging. J Alzheimers Dis 2023; 96:1767-1780. [PMID: 38007645 PMCID: PMC10741337 DOI: 10.3233/jad-230145] [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] [Accepted: 10/04/2023] [Indexed: 11/27/2023]
Abstract
BACKGROUND Alzheimer's disease and related dementias (ADRD) involve biological processes that begin years to decades before onset of clinical symptoms. The plasma proteome can offer insight into brain aging and risk of incident dementia among cognitively healthy adults. OBJECTIVE To identify biomarkers and biological pathways associated with neuroimaging measures and incident dementia in two large community-based cohorts by applying a correlation-based network analysis to the plasma proteome. METHODS Weighted co-expression network analysis of 1,305 plasma proteins identified four modules of co-expressed proteins, which were related to MRI brain volumes and risk of incident dementia over a median 20-year follow-up in Framingham Heart Study (FHS) Offspring cohort participants (n = 1,861). Analyses were replicated in the Cardiovascular Health Study (CHS) (n = 2,117, mean 6-year follow-up). RESULTS Two proteomic modules, one related to protein clearance and synaptic maintenance (M2) and a second to inflammation (M4), were associated with total brain volume in FHS (M2: p = 0.014; M4: p = 4.2×10-5). These modules were not significantly associated with hippocampal volume, white matter hyperintensities, or incident all-cause or AD dementia. Associations with TCBV did not replicate in CHS, an older cohort with a greater burden of comorbidities. CONCLUSIONS Proteome networks implicate an early role for biological pathways involving inflammation and synaptic function in preclinical brain atrophy, with implications for clinical dementia.
Collapse
Affiliation(s)
- Meghan I. Short
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center, San Antonio, TX, USA
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA, USA
- Department of Medicine, Tufts University School of Medicine, Boston, MA, USA
| | - Alison E. Fohner
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
| | - Håvard K. Skjellegrind
- HUNT Research Centre, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, NTNU, Levanger, Norway
- Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Alexa Beiser
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Framingham Heart Study, Framingham, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Mitzi M. Gonzales
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center, San Antonio, TX, USA
- Department of Neurology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Claudia L. Satizabal
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center, San Antonio, TX, USA
- Framingham Heart Study, Framingham, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Department of Population Health Sciences, University of Texas Health Science Center, San Antonio, TX, USA
| | - Thomas R. Austin
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
| | - W.T. Longstreth
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Neurology, University of Washington, Seattle, WA, USA
| | - Joshua C. Bis
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
| | - Oscar Lopez
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kristian Hveem
- HUNT Research Centre, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, NTNU, Levanger, Norway
- K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Geir Selbæk
- Norwegian National Centre for Ageing and Health, Vestfold Hospital Trust, Tønsberg, Norway
- Department of Geriatric Medicine, Oslo University Hospital, Oslo, Norway
| | - Martin G. Larson
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Framingham Heart Study, Framingham, MA, USA
| | - Qiong Yang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Framingham Heart Study, Framingham, MA, USA
| | - Hugo J. Aparicio
- Framingham Heart Study, Framingham, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Emer R. McGrath
- Framingham Heart Study, Framingham, MA, USA
- School of Medicine, National University of Ireland Galway, Galway, Ireland
- HRB Clinical Research Facility, National University of Ireland Galway, Galway, Ireland
| | - Robert E. Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Charles S. DeCarli
- Department of Neurology, School of Medicine and Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California, Davis, Sacramento, CA, USA
| | - Bruce M. Psaty
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Health Systems and Population Health, University of Washington, Seattle, WA, USA
| | - Ramachandran S. Vasan
- Framingham Heart Study, Framingham, MA, USA
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
- Boston University Center for Computing and Data Science, Boston, MA, USA
| | - Habil Zare
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, University of Texas Health Science Center, San Antonio, TX, USA
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center, San Antonio, TX, USA
- Framingham Heart Study, Framingham, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
24
|
Hillary RF, Gadd DA, McCartney DL, Shi L, Campbell A, Walker RM, Ritchie CW, Deary IJ, Evans KL, Nevado‐Holgado AJ, Hayward C, Porteous DJ, McIntosh AM, Lovestone S, Robinson MR, Marioni RE. Genome- and epigenome-wide studies of plasma protein biomarkers for Alzheimer's disease implicate TBCA and TREM2 in disease risk. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2022; 14:e12280. [PMID: 35475137 PMCID: PMC9019629 DOI: 10.1002/dad2.12280] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 12/03/2021] [Accepted: 12/07/2021] [Indexed: 11/24/2022]
Abstract
Introduction The levels of many blood proteins are associated with Alzheimer's disease (AD) or its pathological hallmarks. Elucidating the molecular factors that control circulating levels of these proteins may help to identify proteins associated with disease risk mechanisms. Methods Genome-wide and epigenome-wide studies (nindividuals ≤1064) were performed on plasma levels of 282 AD-associated proteins, identified by a structured literature review. Bayesian penalized regression estimated contributions of genetic and epigenetic variation toward inter-individual differences in plasma protein levels. Mendelian randomization (MR) and co-localization tested associations between proteins and disease-related phenotypes. Results Sixty-four independent genetic and 26 epigenetic loci were associated with 45 proteins. Novel findings included an association between plasma triggering receptor expressed on myeloid cells 2 (TREM2) levels and a polymorphism and cytosine-phosphate-guanine (CpG) site within the MS4A4A locus. Higher plasma tubulin-specific chaperone A (TBCA) and TREM2 levels were significantly associated with lower AD risk. Discussion Our data inform the regulation of biomarker levels and their relationships with AD.
Collapse
Affiliation(s)
- Robert F. Hillary
- Centre for Genomic and Experimental MedicineInstitute of Genetics and Cancer, University of EdinburghEdinburghUK
| | - Danni A. Gadd
- Centre for Genomic and Experimental MedicineInstitute of Genetics and Cancer, University of EdinburghEdinburghUK
| | - Daniel L. McCartney
- Centre for Genomic and Experimental MedicineInstitute of Genetics and Cancer, University of EdinburghEdinburghUK
| | - Liu Shi
- Department of PsychiatryUniversity of OxfordOxfordUK
| | - Archie Campbell
- Centre for Genomic and Experimental MedicineInstitute of Genetics and Cancer, University of EdinburghEdinburghUK
| | - Rosie M. Walker
- Centre for Genomic and Experimental MedicineInstitute of Genetics and Cancer, University of EdinburghEdinburghUK
- Centre for Clinical Brain Sciences, Chancellor's Building, 49 Little France CrescentUniversity of EdinburghEdinburghUK
| | - Craig W. Ritchie
- Edinburgh Dementia Prevention, Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUK
| | - Ian J. Deary
- Lothian Birth Cohorts, Department of PsychologyUniversity of EdinburghEdinburghUK
| | - Kathryn L. Evans
- Centre for Genomic and Experimental MedicineInstitute of Genetics and Cancer, University of EdinburghEdinburghUK
| | | | - Caroline Hayward
- MRC Human Genetics UnitInstitute of Genetics and CancerUniversity of EdinburghEdinburghUK
| | - David J. Porteous
- Centre for Genomic and Experimental MedicineInstitute of Genetics and Cancer, University of EdinburghEdinburghUK
| | - Andrew M. McIntosh
- Centre for Genomic and Experimental MedicineInstitute of Genetics and Cancer, University of EdinburghEdinburghUK
- Division of Psychiatry, Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUK
| | - Simon Lovestone
- Department of PsychiatryUniversity of OxfordOxfordUK
- NeurodegenerationJohnson and Johnson Medical LtdWokinghamUK
| | - Matthew R. Robinson
- Medical Genomics GroupInstitute of Science and Technology AustriaKlosterneuburgAustria
| | - Riccardo E. Marioni
- Centre for Genomic and Experimental MedicineInstitute of Genetics and Cancer, University of EdinburghEdinburghUK
| |
Collapse
|
25
|
Dansson HV, Stempfle L, Egilsdóttir H, Schliep A, Portelius E, Blennow K, Zetterberg H, Johansson FD. Predicting progression and cognitive decline in amyloid-positive patients with Alzheimer's disease. ALZHEIMERS RESEARCH & THERAPY 2021; 13:151. [PMID: 34488882 PMCID: PMC8422748 DOI: 10.1186/s13195-021-00886-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 08/08/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND In Alzheimer's disease, amyloid- β (A β) peptides aggregate in the lowering CSF amyloid levels - a key pathological hallmark of the disease. However, lowered CSF amyloid levels may also be present in cognitively unimpaired elderly individuals. Therefore, it is of great value to explain the variance in disease progression among patients with A β pathology. METHODS A cohort of n=2293 participants, of whom n=749 were A β positive, was selected from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database to study heterogeneity in disease progression for individuals with A β pathology. The analysis used baseline clinical variables including demographics, genetic markers, and neuropsychological data to predict how the cognitive ability and AD diagnosis of subjects progressed using statistical models and machine learning. Due to the relatively low prevalence of A β pathology, models fit only to A β-positive subjects were compared to models fit to an extended cohort including subjects without established A β pathology, adjusting for covariate differences between the cohorts. RESULTS A β pathology status was determined based on the A β42/A β40 ratio. The best predictive model of change in cognitive test scores for A β-positive subjects at the 2-year follow-up achieved an R2 score of 0.388 while the best model predicting adverse changes in diagnosis achieved a weighted F1 score of 0.791. A β-positive subjects declined faster on average than those without A β pathology, but the specific level of CSF A β was not predictive of progression rate. When predicting cognitive score change 4 years after baseline, the best model achieved an R2 score of 0.325 and it was found that fitting models to the extended cohort improved performance. Moreover, using all clinical variables outperformed the best model based only on a suite of cognitive test scores which achieved an R2 score of 0.228. CONCLUSION Our analysis shows that CSF levels of A β are not strong predictors of the rate of cognitive decline in A β-positive subjects when adjusting for other variables. Baseline assessments of cognitive function accounts for the majority of variance explained in the prediction of 2-year decline but is insufficient for achieving optimal results in longer-term predictions. Predicting changes both in cognitive test scores and in diagnosis provides multiple perspectives of the progression of potential AD subjects.
Collapse
Affiliation(s)
- Hákon Valur Dansson
- Department of Computer Science and Engineering, Chalmers University of Technology and University of Gothenburg, Gothenburg, Sweden
| | - Lena Stempfle
- Department of Computer Science and Engineering, Chalmers University of Technology and University of Gothenburg, Gothenburg, Sweden.
| | - Hildur Egilsdóttir
- Department of Computer Science and Engineering, Chalmers University of Technology and University of Gothenburg, Gothenburg, Sweden
| | - Alexander Schliep
- Department of Computer Science and Engineering, Chalmers University of Technology and University of Gothenburg, Gothenburg, Sweden
| | - Erik Portelius
- 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
| | - 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
| | - Henrik Zetterberg
- 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.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.,UK Dementia Research Institute, UCL, London, UK
| | - Fredrik D Johansson
- Department of Computer Science and Engineering, Chalmers University of Technology and University of Gothenburg, Gothenburg, Sweden
| | | |
Collapse
|
26
|
Schwab K, Melis V, Harrington CR, Wischik CM, Magbagbeolu M, Theuring F, Riedel G. Proteomic Analysis of Hydromethylthionine in the Line 66 Model of Frontotemporal Dementia Demonstrates Actions on Tau-Dependent and Tau-Independent Networks. Cells 2021; 10:2162. [PMID: 34440931 PMCID: PMC8391171 DOI: 10.3390/cells10082162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/16/2021] [Accepted: 08/19/2021] [Indexed: 12/21/2022] Open
Abstract
Abnormal aggregation of tau is the pathological hallmark of tauopathies including frontotemporal dementia (FTD). We have generated tau-transgenic mice that express the aggregation-prone P301S human tau (line 66). These mice present with early-onset, high tau load in brain and FTD-like behavioural deficiencies. Several of these behavioural phenotypes and tau pathology are reversed by treatment with hydromethylthionine but key pathways underlying these corrections remain elusive. In two proteomic experiments, line 66 mice were compared with wild-type mice and then vehicle and hydromethylthionine treatments of line 66 mice were compared. The brain proteome was investigated using two-dimensional electrophoresis and mass spectrometry to identify protein networks and pathways that were altered due to tau overexpression or modified by hydromethylthionine treatment. Overexpression of mutant tau induced metabolic/mitochondrial dysfunction, changes in synaptic transmission and in stress responses, and these functions were recovered by hydromethylthionine. Other pathways, such as NRF2, oxidative phosphorylation and protein ubiquitination were activated by hydromethylthionine, presumably independent of its function as a tau aggregation inhibitor. Our results suggest that hydromethylthionine recovers cellular activity in both a tau-dependent and a tau-independent fashion that could lead to a wide-spread improvement of homeostatic function in the FTD brain.
Collapse
Affiliation(s)
- Karima Schwab
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK; (K.S.); (V.M.); (C.R.H.); (C.M.W.)
- Charité—Universitätsmedizin Berlin, Hessische Str. 3-4, 10115 Berlin, Germany; (M.M.); (F.T.)
| | - Valeria Melis
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK; (K.S.); (V.M.); (C.R.H.); (C.M.W.)
| | - Charles R. Harrington
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK; (K.S.); (V.M.); (C.R.H.); (C.M.W.)
- TauRx Therapeutics Ltd., 395 King Street, Aberdeen AB24 5RP, UK
| | - Claude M. Wischik
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK; (K.S.); (V.M.); (C.R.H.); (C.M.W.)
- TauRx Therapeutics Ltd., 395 King Street, Aberdeen AB24 5RP, UK
| | - Mandy Magbagbeolu
- Charité—Universitätsmedizin Berlin, Hessische Str. 3-4, 10115 Berlin, Germany; (M.M.); (F.T.)
| | - Franz Theuring
- Charité—Universitätsmedizin Berlin, Hessische Str. 3-4, 10115 Berlin, Germany; (M.M.); (F.T.)
| | - Gernot Riedel
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK; (K.S.); (V.M.); (C.R.H.); (C.M.W.)
| |
Collapse
|
27
|
Higgins-Chen AT, Thrush KL, Levine ME. Aging biomarkers and the brain. Semin Cell Dev Biol 2021; 116:180-193. [PMID: 33509689 PMCID: PMC8292153 DOI: 10.1016/j.semcdb.2021.01.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 12/15/2022]
Abstract
Quantifying biological aging is critical for understanding why aging is the primary driver of morbidity and mortality and for assessing novel therapies to counter pathological aging. In the past decade, many biomarkers relevant to brain aging have been developed using various data types and modeling techniques. Aging involves numerous interconnected processes, and thus many complementary biomarkers are needed, each capturing a different slice of aging biology. Here we present a hierarchical framework highlighting how these biomarkers are related to each other and the underlying biological processes. We review those measures most studied in the context of brain aging: epigenetic clocks, proteomic clocks, and neuroimaging age predictors. Many studies have linked these biomarkers to cognition, mental health, brain structure, and pathology during aging. We also delve into the challenges and complexities in interpreting these biomarkers and suggest areas for further innovation. Ultimately, a robust mechanistic understanding of these biomarkers will be needed to effectively intervene in the aging process to prevent and treat age-related disease.
Collapse
Affiliation(s)
- Albert T Higgins-Chen
- Department of Psychiatry, Yale University School of Medicine, 300 George St, Suite 901, New Haven, CT 06511, USA.
| | - Kyra L Thrush
- Program in Computational Biology and Bioinformatics, Yale University, 300 George St, Suite 501, New Haven, CT 06511, USA.
| | - Morgan E Levine
- Department of Pathology, Yale University School of Medicine, 310 Cedar Street, Suite LH 315A, New Haven, CT 06520, USA.
| |
Collapse
|
28
|
Ubaida-Mohien C, Moaddel R, Moore AZ, Kuo PL, Faghri F, Tharakan R, Tanaka T, Nalls MA, Ferrucci L. Proteomics and Epidemiological Models of Human Aging. Front Physiol 2021; 12:674013. [PMID: 34135771 PMCID: PMC8202502 DOI: 10.3389/fphys.2021.674013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 05/03/2021] [Indexed: 12/21/2022] Open
Abstract
Human aging is associated with a decline of physical and cognitive function and high susceptibility to chronic diseases, which is influenced by genetics, epigenetics, environmental, and socio-economic status. In order to identify the factors that modulate the aging process, established measures of aging mechanisms are required, that are both robust and feasible in humans. It is also necessary to connect these measures to the phenotypes of aging and their functional consequences. In this review, we focus on how this has been addressed from an epidemiologic perspective using proteomics. The key aspects of epidemiological models of aging can be incorporated into proteomics and other omics which can provide critical detailed information on the molecular and biological processes that change with age, thus unveiling underlying mechanisms that drive multiple chronic conditions and frailty, and ideally facilitating the identification of new effective approaches for prevention and treatment.
Collapse
Affiliation(s)
- Ceereena Ubaida-Mohien
- Biomedical Research Center, National Institute on Aging, National Institute of Health, Baltimore, MD, United States
| | - Ruin Moaddel
- Biomedical Research Center, National Institute on Aging, National Institute of Health, Baltimore, MD, United States
| | - Ann Zenobia Moore
- Biomedical Research Center, National Institute on Aging, National Institute of Health, Baltimore, MD, United States
| | - Pei-Lun Kuo
- Biomedical Research Center, National Institute on Aging, National Institute of Health, Baltimore, MD, United States
| | - Faraz Faghri
- Center for Alzheimer’s and Related Dementias, National Institute on Aging, Bethesda, MD, United States
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, United States
- Data Tecnica International, Glen Echo, MD, United States
| | - Ravi Tharakan
- Biomedical Research Center, National Institute on Aging, National Institute of Health, Baltimore, MD, United States
| | - Toshiko Tanaka
- Biomedical Research Center, National Institute on Aging, National Institute of Health, Baltimore, MD, United States
| | - Mike A. Nalls
- Center for Alzheimer’s and Related Dementias, National Institute on Aging, Bethesda, MD, United States
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, United States
- Data Tecnica International, Glen Echo, MD, United States
| | - Luigi Ferrucci
- Biomedical Research Center, National Institute on Aging, National Institute of Health, Baltimore, MD, United States
| |
Collapse
|
29
|
Abstract
DNA methylation is an important biological process that involves the reversible addition of chemical tags called methyl groups to DNA and affects whether genes are active or inactive. Individual methylation profiles are determined by both genetic and environmental influences. Inter-individual variation in DNA methylation profiles can be exploited to estimate or predict a wide variety of human characteristics and disease risk profiles. Indeed, a number of methylation-based predictors of human traits have been developed and linked to important health outcomes. However, there is an unmet need to communicate the applicability and limitations of state-of-the-art methylation-based predictors to the wider community. To address this need, we have created a secure, web-based interactive platform called 'MethylDetectR' which automates the calculation of estimated values or scores for a variety of human traits using blood methylation data. These traits include age, lifestyle traits and high-density lipoprotein cholesterol. Methylation-based predictors often return scores on arbitrary scales. To provide meaning to these scores, users can interactively view how estimated trait scores for a given individual compare against other individuals in the sample. Users can optionally upload binary phenotypes and investigate how estimated traits vary according to case vs. control status for these phenotypes. Users can also view how different methylation-based predictors correlate with one another, and with phenotypic values for corresponding traits in a large reference sample (n = 4,450; Generation Scotland). The 'MethylDetectR' platform allows for the fast and secure calculation of DNA methylation-derived estimates for several human traits. This platform also helps to show the correlations between methylation-based scores and corresponding traits at the level of a sample, report estimated health profiles at an individual level, demonstrate how scores relate to important binary outcomes of interest and highlight the current limitations of molecular health predictors.
Collapse
Affiliation(s)
- Robert F. Hillary
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Midlothian, EH4 2XU, UK
| | - Riccardo E. Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Midlothian, EH4 2XU, UK
| |
Collapse
|
30
|
Hillary RF, Marioni RE. MethylDetectR: a software for methylation-based health profiling. Wellcome Open Res 2020; 5:283. [PMID: 33969230 PMCID: PMC8080939 DOI: 10.12688/wellcomeopenres.16458.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2020] [Indexed: 04/02/2024] Open
Abstract
DNA methylation is an important biological process which involves the reversible addition of chemical tags called methyl groups to DNA and affects whether genes are active or inactive. Individual methylation profiles are determined by both genetic and environmental influences. Inter-individual variation in DNA methylation profiles can be exploited to estimate or predict a wide variety of human characteristics and disease risk profiles. Indeed, a number of methylation-based predictors of human traits have been developed and linked to important health outcomes. However, there is an unmet need to communicate the applicability and limitations of state-of-the-art methylation-based predictors to the wider community. To address this, we created a secure, web-based interactive platform called 'MethylDetectR' which calculates estimated values or scores for a variety of human traits using blood methylation data. These traits include age, lifestyle traits, high-density lipoprotein cholesterol and the levels of 27 blood proteins related to inflammatory and neurological processes and disease. Methylation-based predictors often return scores on arbitrary scales. To provide meaning to these scores, users can interactively view how estimated trait scores for a given individual compare against other individuals in the sample. Users can optionally upload binary phenotypes and investigate how estimated traits vary according to case vs. control status for these phenotypes. Users can also view how different methylation-based predictors correlate with one another, and with phenotypic values for corresponding traits in a large reference sample (n = 4,450; Generation Scotland). The 'MethylDetectR' platform allows for the fast and secure calculation of DNA methylation-derived estimates for many human traits. This platform also helps to show the correlations between methylation-based scores and corresponding traits at the level of a sample, report estimated health profiles at an individual level, demonstrate how scores relate to important binary outcomes of interest and highlight the current limitations of molecular health predictors.
Collapse
Affiliation(s)
- Robert F. Hillary
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Midlothian, EH4 2XU, UK
| | - Riccardo E. Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Midlothian, EH4 2XU, UK
| |
Collapse
|