1
|
Kaur Sardarni U, Ambikan AT, Acharya A, Johnson SD, Avedissian SN, Végvári Á, Neogi U, Byrareddy SN. SARS-CoV-2 variants mediated tissue-specific metabolic reprogramming determines the disease pathophysiology in a hamster model. Brain Behav Immun 2025; 123:914-927. [PMID: 39481495 DOI: 10.1016/j.bbi.2024.10.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 09/28/2024] [Accepted: 10/26/2024] [Indexed: 11/02/2024] Open
Abstract
Despite significant effort, a clear understanding of host tissue-specific responses and their implications for immunopathogenicity against the severe acute respiratory syndrome coronavirus2 (SARS-CoV-2) variant infection has remained poorly defined. To shed light on the interaction between tissues and SARS-CoV-2 variants, we sought to characterize the complex relationship among acute multisystem manifestations, dysbiosis of the gut microbiota, and the resulting implications for SARS-CoV-2 variant-specific immunopathogenesis in the Golden Syrian Hamster (GSH) model using multi-omics approaches. Our investigation revealed the presence of increased SARS-CoV-2 genomic RNA in diverse tissues of delta-infected GSH compared to the omicron variant. Multi-omics analyses uncovered distinctive metabolic responses between the delta and omicron variants, with the former demonstrating dysregulation in synaptic transmission proteins associated with neurocognitive disorders. Additionally, delta-infected GSH exhibited an altered fecal microbiota composition, marked by increased inflammation-associated taxa and reduced commensal bacteria compared to the omicron variant. These findings underscore the SARS-CoV-2-mediated tissue insult, characterized by modified host metabolites, neurological protein dysregulation, and gut dysbiosis, highlighting the compromised gut-lung-brain axis during acute infection.
Collapse
Affiliation(s)
- Urvinder Kaur Sardarni
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Anoop T Ambikan
- The Systems Virology Laboratory, Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, ANA Futura, Campus Flemingsberg, Stockholm, Sweden
| | - Arpan Acharya
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Samuel D Johnson
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sean N Avedissian
- Antiviral Pharmacology Laboratory, Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ákos Végvári
- Division of Chemistry I, Department of Medical Biochemistry & Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ujjwal Neogi
- The Systems Virology Laboratory, Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, ANA Futura, Campus Flemingsberg, Stockholm, Sweden.
| | - Siddappa N Byrareddy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
2
|
Small SL. Precision neurology. Ageing Res Rev 2024; 104:102632. [PMID: 39657848 DOI: 10.1016/j.arr.2024.102632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 11/23/2024] [Accepted: 12/05/2024] [Indexed: 12/12/2024]
Abstract
Over the past several decades, high-resolution brain imaging, blood and cerebrospinal fluid analyses, and other advanced technologies have changed diagnosis from an exercise depending primarily on the history and physical examination to a computer- and online resource-aided process that relies on larger and larger quantities of data. In addition, randomized controlled trials (RCT) at a population level have led to many new drugs and devices to treat neurological disease, including disease-modifying therapies. We are now at a crossroads. Combinatorially profound increases in data about individuals has led to an alternative to population-based RCTs. Genotyping and comprehensive "deep" phenotyping can sort individuals into smaller groups, enabling precise medical decisions at a personal level. In neurology, precision medicine that includes prediction, prevention and personalization requires that genomic and phenomic information further incorporate imaging and behavioral data. In this article, we review the genomic, phenomic, and computational aspects of precision medicine for neurology. After defining biological markers, we discuss some applications of these "-omic" and neuroimaging measures, and then outline the role of computation and ultimately brain simulation. We conclude the article with a discussion of the relation between precision medicine and value-based care.
Collapse
Affiliation(s)
- Steven L Small
- Department of Neuroscience, University of Texas at Dallas, Dallas, TX, USA; Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Neurology, The University of Chicago, Chicago, IL, USA; Department of Neurology, University of California, Irvine, Orange, CA, USA.
| |
Collapse
|
3
|
Błażewicz A, Wojnicka J, Grabrucker AM, Sosnowski P, Trzpil A, Kozub-Pędrak A, Szałaj K, Szmagara A, Grywalska E, Skórzyńska-Dziduszko K. Preliminary investigations of plasma lipidome and selenium levels in adults with treated hypothyroidism and in healthy individuals without selenium deficiency. Sci Rep 2024; 14:29140. [PMID: 39587337 PMCID: PMC11589578 DOI: 10.1038/s41598-024-80862-9] [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: 02/15/2024] [Accepted: 11/21/2024] [Indexed: 11/27/2024] Open
Abstract
The present preliminary study aimed to provide a targeted lipidomic analysis of Hashimoto (HT) and non-HT patients with well-controlled hypothyroidism as well as in healthy adults, and is the first to demonstrate the association of several components of the human lipidome with hypothyroidism in relation to the total plasma selenium content. All the patients and age-, sex-, and BMI-matched healthy controls met the very strict qualification criteria. Se levels were analyzed by ICP-MS, and lipidome studies were conducted using TQ-LC/MS. The 40 acylcarnitines, 90 glycerophospholipids, and 15 sphingomyelins were identified and quantified. PCaaC26:0 and PCaaC40:1 were negatively correlated with Se concentrations. Other lipids that were negatively correlated with Se concentrations but did not present significant differences between the three groups in the Kruskal-Wallis ANOVA test were PCaaC32:0, PCaeC30:0, PCaeC36:5, SMC18:0, and SM C18:1. In the multiple linear regression analyses, Se levels showed negative relationship, whereas different phosphatidylcholines: PCaaC24:0, PCaaC26:0, PCaeC30:1, PCaeC34:0, PCaeC36:4, PCaeC42:0 were positively associated with the presence of (H). Different lipidome components were identified in healthy and hypothyroid patients regardless of the cause of that condition. Studies on larger populations are needed to determine cause-and-effect relations and the potential mechanisms underlying these associations.
Collapse
Affiliation(s)
- Anna Błażewicz
- Department of Pathobiochemistry and Interdisciplinary Applications of Ion Chromatography, Chair of Biomedical Sciences, Medical University of Lublin, 1 Chodźki Street, 20-093, Lublin, Poland.
- Department of Biological Sciences, University of Limerick, Limerick, V94 T9PX, Ireland.
| | - Julia Wojnicka
- Department of Pathobiochemistry and Interdisciplinary Applications of Ion Chromatography, Chair of Biomedical Sciences, Medical University of Lublin, 1 Chodźki Street, 20-093, Lublin, Poland
| | - Andreas M Grabrucker
- Department of Biological Sciences, University of Limerick, Limerick, V94 T9PX, Ireland
- Bernal Institute, University of Limerick, Limerick, V94 T9PX, Ireland
- Health Research Institute (HRI), University of Limerick, Limerick, V94 T9PX, Ireland
| | - Piotr Sosnowski
- Department of Bioanalytics, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090, Lublin, Poland
| | - Alicja Trzpil
- Department of Bioanalytics, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090, Lublin, Poland
| | - Anna Kozub-Pędrak
- Department of Bioanalytics, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090, Lublin, Poland
| | - Klaudia Szałaj
- Department of Bioanalytics, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090, Lublin, Poland
| | - Agnieszka Szmagara
- Faculty of Medicine, Institute of Biological Sciences, Department of Chemistry, The John Paul II Catholic University of Lublin, Konstantynow 1J, 20-708, Lublin, Poland
| | - Ewelina Grywalska
- Department of Experimental Immunology, Medical University of Lublin, 4a Chodźki Street, 20-093, Lublin, Poland
| | | |
Collapse
|
4
|
Ellis D, Watanabe K, Wilmanski T, Lustgarten MS, Korat AVA, Glusman G, Hadlock JJ, Fiehn O, Sebastiani P, Price ND, Hood L, Magis AT, Evans SJ, Pflieger L, Lovejoy JC, Gibbons SM, Funk CC, Baloni P, Rappaport N. APOE Genotype and Biological Age Impact Inter-Omic Associations Related to Bioenergetics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.17.618322. [PMID: 39605362 PMCID: PMC11601402 DOI: 10.1101/2024.10.17.618322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Apolipoprotein E ( APOE ) modifies human aging; specifically, the ε2 and ε4 alleles are among the strongest genetic predictors of longevity and Alzheimer's disease (AD) risk, respectively. However, detailed mechanisms for their influence on aging remain unclear. Herein, we analyzed inter-omic, context-dependent association patterns across APOE genotypes, sex, and health axes in 2,229 community-dwelling individuals to test APOE genotypes for variation in metabolites and metabolite-associations tied to a previously-validated metric of biological aging (BA) based on blood biomarkers. Our analysis, supported by validation in an independent cohort, identified top APOE -associated plasma metabolites as diacylglycerols, which were increased in ε2-carriers and trended higher in ε4-carriers compared to ε3-homozygotes, despite the known opposing aging effects of the allele variants. 'Omics association patterns of ε2-carriers and increased biological age were also counter-intuitively similar, displaying increased associations between insulin resistance markers and energy-generating pathway metabolites. These results provide an atlas of APOE -related 'omic associations and support the involvement of bioenergetic pathways in mediating the impact of APOE on aging.
Collapse
|
5
|
Liang H, Luo H, Sang Z, Jia M, Jiang X, Wang Z, Cong S, Yao X. GREMI: An Explainable Multi-Omics Integration Framework for Enhanced Disease Prediction and Module Identification. IEEE J Biomed Health Inform 2024; 28:6983-6996. [PMID: 39110558 DOI: 10.1109/jbhi.2024.3439713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Multi-omics integration has demonstrated promising performance in complex disease prediction. However, existing research typically focuses on maximizing prediction accuracy, while often neglecting the essential task of discovering meaningful biomarkers. This issue is particularly important in biomedicine, as molecules often interact rather than function individually to influence disease outcomes. To this end, we propose a two-phase framework named GREMI to assist multi-omics classification and explanation. In the prediction phase, we propose to improve prediction performance by employing a graph attention architecture on sample-wise co-functional networks to incorporate biomolecular interaction information for enhanced feature representation, followed by the integration of a joint-late mixed strategy and the true-class-probability block to adaptively evaluate classification confidence at both feature and omics levels. In the interpretation phase, we propose a multi-view approach to explain disease outcomes from the interaction module perspective, providing a more intuitive understanding and biomedical rationale. We incorporate Monte Carlo tree search (MCTS) to explore local-view subgraphs and pinpoint modules that highly contribute to disease characterization from the global-view. Extensive experiments demonstrate that the proposed framework outperforms state-of-the-art methods in seven different classification tasks, and our model effectively addresses data mutual interference when the number of omics types increases. We further illustrate the functional- and disease-relevance of the identified modules, as well as validate the classification performance of discovered modules using an independent cohort.
Collapse
|
6
|
Zonneveld MH, Al Kuhaili N, Mooijaart SP, Slagboom PE, Jukema JW, Noordam R, Trompet S. Increased 1H-NMR metabolomics-based health score associates with declined cognitive performance and functional independence in older adults at risk of cardiovascular disease. GeroScience 2024:10.1007/s11357-024-01391-x. [PMID: 39436550 DOI: 10.1007/s11357-024-01391-x] [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: 06/05/2024] [Accepted: 10/11/2024] [Indexed: 10/23/2024] Open
Abstract
The 1-HMR metabolomics-based MetaboHealth score, comprised of 14 serum metabolic markers, associates with disease-specific mortality, but it is unclear whether the score also reflects cognitive changes and functional impairment. We aimed to assess the associations between the MetaboHealth score with cognitive function and functional decline in older adults at increased cardiovascular risk. A total of 5292 older adults free of dementia at baseline with mean age 75.3 years (SD = 3.4) from the Prospective Study of Pravastatin in the Elderly (PROSPER). MetaboHealth score were measured at baseline, and cognitive function and functional independence were measured at baseline and every 3 months during up to 2.5 years follow-up. Cognitive function was assessed using the Stroop test (selective attention), the Letter Digit Coding test (LDCT) (processing speed), and the two versions of the Picture Learning test (delayed and immediate; memory). Two tests of functional independence were used: Barthel Index (BI) and instrumental activities at daily living (IADL). A higher MetaboHealth score was associated with worse cognitive function (in all domains) and with worse functional independence. For example, after full adjustments, a 1-SD higher MetaboHealth score was associated with 9.02 s (95%CI 7.29, 10.75) slower performance on the Stroop test and 2.79 (2.21, 3.26) less digits coded on the LDCT. During follow-up, 1-SD higher MetaboHealth score was associated with an additional decline of 0.53 s (0.23, 0.83) on the Stroop test and - 0.08 (- 0.11, - 0.06) points on the IADL. Metabolic disturbance, as reflected by an increased metabolomics-based health score, may mark future cognitive and functional decline.
Collapse
Affiliation(s)
- Michelle H Zonneveld
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Nour Al Kuhaili
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Simon P Mooijaart
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
- LUMC Center for Medicine for Older People, Leiden University Medical Center, Leiden, The Netherlands
| | - P Eline Slagboom
- Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Netherlands Heart Institute, Utrecht, The Netherlands
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands.
| | - Stella Trompet
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| |
Collapse
|
7
|
Shen H, Yu Y, Wang J, Nie Y, Tang Y, Qu M. Plasma lipidomic signatures of dementia with Lewy bodies revealed by machine learning, and compared to alzheimer's disease. Alzheimers Res Ther 2024; 16:226. [PMID: 39407312 PMCID: PMC11476188 DOI: 10.1186/s13195-024-01585-7] [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: 04/29/2024] [Accepted: 09/29/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Dementia with Lewy Bodies (DLB) is a complex neurodegenerative disorder that often overlaps clinically with Alzheimer's disease (AD), presenting challenges in accurate diagnosis and underscoring the need for novel biomarkers. Lipidomic emerges as a promising avenue for uncovering disease-specific metabolic alterations and potential biomarkers, particularly as the lipidomics landscape of DLB has not been previously explored. We aim to identify potential diagnostic biomarkers and elucidate the disease's pathophysiological mechanisms. METHODS This study conducted a lipidomic analysis of plasma samples from patients with DLB, AD, and healthy controls (HCs) at Xuanwu Hospital. Untargeted plasma lipidomic profiling was conducted via liquid chromatography coupled with mass spectrometry. Machine learning methods were employed to discern lipidomic signatures specific to DLB and to differentiate it from AD. RESULTS The study enrolled 159 participants, including 57 with AD, 48 with DLB, and 54 HCs. Significant differences in lipid profiles were observed between the DLB and HC groups, particularly in the classes of sphingolipids and phospholipids. A total of 55 differentially expressed lipid species were identified between DLB and HCs, and 17 between DLB and AD. Correlations were observed linking these lipidomic profiles to clinical parameters like Unified Parkinson's Disease Rating Scale III (UPDRS III) and cognitive scores. Machine learning models demonstrated to be highly effective in distinguishing DLB from both HCs and AD, achieving substantial accuracy through the utilization of specific lipidomic signatures. These include PC(15:0_18:2), PC(15:0_20:5), and SPH(d16:0) for differentiation between DLB and HCs; and a panel includes 13 lipid molecules: four PCs, two PEs, three SPHs, two Cers, and two Hex1Cers for distinguishing DLB from AD. CONCLUSIONS This study presents a novel and comprehensive lipidomic profile of DLB, distinguishing it from AD and HCs. Predominantly, sphingolipids (e.g., ceramides and SPHs) and phospholipids (e.g., PE and PC) were the most dysregulated lipids in relation to DLB patients. The lipidomics panels identified through machine learning may serve as effective plasma biomarkers for diagnosing DLB and differentiating it from AD dementia.
Collapse
Affiliation(s)
- Huixin Shen
- Departments of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Yueyi Yu
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jing Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yuting Nie
- Departments of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Yi Tang
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
| | - Miao Qu
- Departments of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- Departments of Chinese Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
8
|
Tian Q, Yao S, Marron MM, Greig EE, Shore S, Ferrucci L, Shah R, Murthy VL, Newman AB. Shared plasma metabolomic profiles of cognitive and mobility decline predict future dementia. GeroScience 2024; 46:4883-4894. [PMID: 38829458 PMCID: PMC11336156 DOI: 10.1007/s11357-024-01228-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 05/27/2024] [Indexed: 06/05/2024] Open
Abstract
Experiencing decline in both cognition and mobility is associated with a substantially higher dementia risk than cognitive decline only. Metabolites associated with both cognitive and mobility declines may be early predictors of dementia and reveal specific pathways to dementia. We analyzed data from 2450 participants initially free of dementia who had 613 metabolites measured in plasma in 1998-1999 (mean age = 75.2 ± 2.9 years old, 37.8% Black, 50% women) from the Health, Aging and Body Composition study. Dementia diagnosis was determined by race-specific decline in 3MS scores, medication use, and hospital records through 2014. Cognition and mobility were repeatedly measured using 3MS and a 20-m walking test up to 10 years, respectively. We examined metabolite associations with changes in 3MS (n = 2046) and gait speed (n = 2019) using multivariable linear regression adjusted for age, sex, race, and baseline performance and examined metabolite associations with dementia risk using Cox regression. During a mean follow-up of 9.3 years, 534 (21.8%) participants developed dementia. On average, 3MS declined 0.47/year and gait declined 0.04 m/sec/year. After covariate adjustment, 75 metabolites were associated with cognitive decline, and 111 metabolites were associated with gait decline (FDR-adjusted p < 0.05). Twenty-six metabolites were associated with both cognitive and gait declines. Eighteen of 26 metabolites were associated with dementia risk (p < 0.05), notably amino acids, glycerophospholipids (lysoPCs, PCs, PEs), and sphingolipids. Results remained similar after adjusting for cardiovascular disease or apolipoprotein E ɛ4 carrier status. During aging, metabolomic profiles of cognitive decline and mobility decline show distinct and shared signatures. Shared metabolomic profiles suggest that inflammation and deficits in mitochondria and the urea cycle in addition to the central nervous system may play key roles in both cognitive and mobility declines and predict dementia. Future studies are warranted to investigate longitudinal metabolite changes and metabolomic markers with dementia pathologies.
Collapse
Affiliation(s)
- Qu Tian
- Longitudinal Studies Section, National Institute on Aging, 251 Bayview Blvd M04B332, Baltimore, MD, 21224, USA.
| | - Shanshan Yao
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Megan M Marron
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Erin E Greig
- Longitudinal Studies Section, National Institute on Aging, 251 Bayview Blvd M04B332, Baltimore, MD, 21224, USA
| | | | - Luigi Ferrucci
- Longitudinal Studies Section, National Institute on Aging, 251 Bayview Blvd M04B332, Baltimore, MD, 21224, USA
| | - Ravi Shah
- University of Michigan, Ann Arbor, MI, USA
| | | | - Anne B Newman
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
9
|
Zhao T, Zeng J, Zhang R, Wang H, Pu L, Yang H, Liang J, Dai X, Fan W, Han L. Identification of Blood Biomarkers in Ischemic Stroke by Integrated Analysis of Metabolomics and Proteomics. J Proteome Res 2024; 23:4082-4094. [PMID: 39167481 DOI: 10.1021/acs.jproteome.4c00394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
We aimed to uncover the pathological mechanism of ischemic stroke (IS) using a combined analysis of untargeted metabolomics and proteomics. The serum samples from a discovery set of 44 IS patients and 44 matched controls were analyzed using a specific detection method. The same method was then used to validate metabolites and proteins in the two validation sets: one with 30 IS patients and 30 matched controls, and the other with 50 IS patients and 50 matched controls. A total of 105 and 221 differentially expressed metabolites or proteins were identified, and the association between the two omics was determined in the discovery set. Enrichment analysis of the top 25 metabolites and 25 proteins in the two-way orthogonal partial least-squares with discriminant analysis, which was employed to identify highly correlated biomarkers, highlighted 15 pathways relevant to the pathological process. One metabolite and seven proteins exhibited differences between groups in the validation set. The binary logistic regression model, which included metabolite 2-hydroxyhippuric acid and proteins APOM_O95445, MASP2_O00187, and PRTN3_D6CHE9, achieved an area under the curve of 0.985 (95% CI: 0.966-1) in the discovery set. This study elucidated alterations and potential coregulatory influences of metabolites and proteins in the blood of IS patients.
Collapse
Affiliation(s)
- Tian Zhao
- Department of Clinical Epidemiology, Ningbo No. 2 Hospital, Ningbo, Zhejiang 315000, China
- Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Guoke Ningbo Life Science and Health Industry Research Institute, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315000, China
| | - Jingjing Zeng
- Department of Clinical Epidemiology, Ningbo No. 2 Hospital, Ningbo, Zhejiang 315000, China
- Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Guoke Ningbo Life Science and Health Industry Research Institute, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315000, China
| | - Ruijie Zhang
- Department of Clinical Epidemiology, Ningbo No. 2 Hospital, Ningbo, Zhejiang 315000, China
- Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Guoke Ningbo Life Science and Health Industry Research Institute, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315000, China
| | - Han Wang
- Department of Clinical Epidemiology, Ningbo No. 2 Hospital, Ningbo, Zhejiang 315000, China
- Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Guoke Ningbo Life Science and Health Industry Research Institute, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315000, China
| | - Liyuan Pu
- Department of Clinical Epidemiology, Ningbo No. 2 Hospital, Ningbo, Zhejiang 315000, China
- Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Guoke Ningbo Life Science and Health Industry Research Institute, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315000, China
| | - Huiqun Yang
- Department of Clinical Epidemiology, Ningbo No. 2 Hospital, Ningbo, Zhejiang 315000, China
- Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Guoke Ningbo Life Science and Health Industry Research Institute, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315000, China
| | - Jie Liang
- Department of Clinical Epidemiology, Ningbo No. 2 Hospital, Ningbo, Zhejiang 315000, China
- Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Guoke Ningbo Life Science and Health Industry Research Institute, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315000, China
| | - Xiaoyu Dai
- Department of Anus & Intestine Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang 315000, China
| | - Weinv Fan
- Department of Neurology, Ningbo No. 2 Hospital, Ningbo, Zhejiang 315000, China
| | - Liyuan Han
- Department of Clinical Epidemiology, Ningbo No. 2 Hospital, Ningbo, Zhejiang 315000, China
- Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Guoke Ningbo Life Science and Health Industry Research Institute, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315000, China
| |
Collapse
|
10
|
Lüleci HB, Jones A, Çakır T. Multi-omics analyses highlight molecular differences between clinical and neuropathological diagnoses in Alzheimer's disease. Eur J Neurosci 2024; 60:4922-4936. [PMID: 39072881 DOI: 10.1111/ejn.16482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 05/14/2024] [Accepted: 07/13/2024] [Indexed: 07/30/2024]
Abstract
Both clinical diagnosis and neuropathological diagnosis are commonly used in literature to categorize individuals as Alzheimer's disease (AD) or non-AD in omics analyses. Whether these diagnostic strategies result in distinct profiles of molecular abnormalities is poorly understood. Here, we analysed one of the most commonly used AD omics datasets in the literature from the Religious Orders Study and Memory and Aging Project (ROSMAP) cohort and compared the two diagnosis strategies using brain transcriptome and metabolome by grouping individuals as non-AD and AD according to clinical or neuropathological diagnosis separately. Differentially expressed genes, associated pathways related with AD hallmarks and AD-related genes showed that the categorization based on neuropathological diagnosis more accurately reflects the disease state at the molecular level than the categorization based on clinical diagnosis. We further identified consensus biomarker candidates between the two diagnosis strategies such as 5-hydroxylysine, sphingomyelin and 1-myristoyl-2-palmitoyl-GPC as metabolite biomarkers and sphingolipid metabolism as a pathway biomarker, which could be robust AD biomarkers since they are independent of diagnosis strategies. We also used consensus AD and consensus non-AD individuals between the two diagnostic strategies to train a machine-learning based model, which we used to classify the individuals who were cognitively normal but diagnosed as AD based on neuropathological diagnosis (asymptomatic AD individuals). The majority of these individuals were classified as consensus AD patients for both omics data types. Our study provides a detailed characterization of both diagnostic strategies in terms of the association of the corresponding multi-omics profiles with AD.
Collapse
Affiliation(s)
| | - Attila Jones
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Tunahan Çakır
- Department of Bioengineering, Gebze Technical University, Kocaeli, Turkey
| |
Collapse
|
11
|
Zhang A, Pan C, Wu M, Lin Y, Chen J, Zhong N, Zhang R, Pu L, Han L, Pan H. Causal association between plasma metabolites and neurodegenerative diseases. Prog Neuropsychopharmacol Biol Psychiatry 2024; 134:111067. [PMID: 38908505 DOI: 10.1016/j.pnpbp.2024.111067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 06/06/2024] [Accepted: 06/19/2024] [Indexed: 06/24/2024]
Abstract
BACKGROUND Establishing causal relationships between metabolic biomarkers and neurodegenerative diseases such as Alzheimer's disease (AD) and Parkinson's disease (PD) is a challenge faced by observational studies. In this study, our aim was to investigate the causal associations between plasma metabolites and neurodegenerative diseases using Mendelian Randomization (MR) methods. METHODS We utilized genetic associations with 1400 plasma metabolic traits as exposures. We used large-scale genome-wide association study (GWAS) summary statistics for AD and PD as our discovery datasets. For validation, we performed repeated analyses using different GWAS datasets. The main statistical method employed was inverse variance-weighted (IVW). We also conducted enrichment pathway analysis for IVW-identified metabolites. RESULTS In the discovered dataset, there are a total of 69 metabolites (36 negatively, 33 positively) potentially associated with AD, and 47 metabolites (24 negatively, 23 positively) potentially associated with PD. Among these, 4 significant metabolites overlap with significant metabolites (PIVW < 0.05)in the validation dataset for AD, and 1 metabolite overlaps with significant metabolites in the validation dataset for PD. Three metabolites serve as common potential metabolic markers for both AD and PD, including Tryptophan betaine, Palmitoleoylcarnitine (C16:1), and X-23655 levels. Further pathway enrichment analysis suggests that the SLC-mediated transmembrane transport pathway, involving tryptophan betaine and carnitine metabolites, may represent potential intervention targets for treating AD and PD. CONCLUSION This study offers novel insights into the causal effects of plasma metabolites on degenerative diseases through the integration of genomics and metabolomics. The identification of metabolites and metabolic pathways linked to AD and PD enhances our comprehension of the underlying biological mechanisms and presents promising targets for future therapeutic interventions in AD and PD.
Collapse
Affiliation(s)
- Ao Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Guangdong Medical University, Dongguan City, Guangdong Province, China
| | - Congcong Pan
- Department of Epidemiology and Health Statistics, School of Public Health, Guangdong Medical University, Dongguan City, Guangdong Province, China
| | - Meifen Wu
- Department of Endocrinology, The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan City, Guangdong Province, China
| | - Yue Lin
- Department of Epidemiology and Health Statistics, School of Public Health, Guangdong Medical University, Dongguan City, Guangdong Province, China
| | - Jiashen Chen
- Department of Epidemiology and Health Statistics, School of Public Health, Guangdong Medical University, Dongguan City, Guangdong Province, China
| | - Ni Zhong
- Department of Epidemiology and Health Statistics, School of Public Health, Guangdong Medical University, Dongguan City, Guangdong Province, China
| | - Ruijie Zhang
- Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Ningbo Institute of Life Sciences and Health Industry Research Institute, Chinese Academy of Sciences, Ningbo, Zhejiang Province, China
| | - Liyuan Pu
- Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Ningbo Institute of Life Sciences and Health Industry Research Institute, Chinese Academy of Sciences, Ningbo, Zhejiang Province, China
| | - Liyuan Han
- Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Ningbo Institute of Life Sciences and Health Industry Research Institute, Chinese Academy of Sciences, Ningbo, Zhejiang Province, China.
| | - Haiyan Pan
- Department of Epidemiology and Health Statistics, School of Public Health, Guangdong Medical University, Dongguan City, Guangdong Province, China.
| |
Collapse
|
12
|
Li Y, Wang Z, Kong M, Yong Y, Yang X, Liu C. The role of GZMA as a target of cysteine and biomarker in Alzheimer's disease, pelvic organ prolapse, and tumor progression. Front Pharmacol 2024; 15:1447605. [PMID: 39228516 PMCID: PMC11368878 DOI: 10.3389/fphar.2024.1447605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 07/30/2024] [Indexed: 09/05/2024] Open
Abstract
Objective: This study aims to investigate how changes in peripheral blood metabolites in Alzheimer's Disease (AD) patients affect the development of Pelvic Organ Prolapse (POP) using a multi-omics approach. We specifically explore the interactions of signaling pathways, gene expression, and protein-metabolite interactions, with a focus on GZMA and cysteine in age-related diseases. Methods: This study utilized multi-omics analysis, including metabolomics and transcriptomics, to evaluate the perturbations in peripheral blood metabolites and their effect on POP in AD patients. Additionally, a comprehensive pan-cancer and immune infiltration analysis was performed on the core targets of AD combined with POP, exploring their potential roles in tumor progression and elucidating their pharmacological relevance to solid tumors. Results: We identified 47 differential metabolites linked to 9 significant signaling pathways, such as unsaturated fatty acid biosynthesis and amino acid metabolism. A thorough gene expression analysis revealed numerous differentially expressed genes (DEGs), with Gene Set Enrichment Analysis (GSEA) showing significant changes in gene profiles of AD and POP. Network topology analysis highlighted central nodes in the AD-POP co-expressed genes network. Functional analyses indicated involvement in critical biological processes and pathways. Molecular docking studies showed strong interactions between cysteine and proteins PTGS2 and GZMA, and molecular dynamics simulations confirmed the stability of these complexes. In vitro validation demonstrated that cysteine reduced ROS levels and protected cell viability. GZMA was widely expressed in various cancers, associated with immune cells, and correlated with patient survival prognosis. Conclusion: Multi-omics analysis revealed the role of peripheral blood metabolites in the molecular dynamics of AD and their interactions with POP. This study identified potential biomarkers and therapeutic targets, emphasizing the effectiveness of integrative approaches in treating AD and POP concurrently. The findings highlight the need for in-depth research on novel targets and biomarkers to advance therapeutic strategies.
Collapse
Affiliation(s)
- Yan Li
- Department of Gynecology and Obstetrics, Affiliated Beijing Chaoyang Hospital of Capital Medical University, Beijing, China
- Department of Gynecology and Obstetrics, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Zhuo Wang
- Department of Gynecology and Obstetrics, Ningxia Medical University, Yinchuan, China
| | - Min Kong
- Department of Gynecology and Obstetrics, Ningxia Medical University, Yinchuan, China
| | - Yuanyuan Yong
- Department of Gynecology and Obstetrics, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Xin Yang
- Department of Gynecology and Obstetrics, Ningxia Medical University, Yinchuan, China
| | - Chongdong Liu
- Department of Gynecology and Obstetrics, Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| |
Collapse
|
13
|
Milaneschi Y, Montanari S, Jansen R, Schranner D, Kastenmüller G, Arnold M, Janiri D, Sani G, Bhattacharyya S, Dehkordi SM, Dunlop B, Rush A, Penninx B, Kaddurah-Daouk R. Acylcarnitines metabolism in depression: association with diagnostic status, depression severity and symptom profile in the NESDA cohort. RESEARCH SQUARE 2024:rs.3.rs-4638158. [PMID: 39149483 PMCID: PMC11326352 DOI: 10.21203/rs.3.rs-4638158/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Acylcarnitines (ACs) are involved in bioenergetics processes that may play a role in the pathophysiology of depression. Previous genomic evidence identified four ACs potentially linked to depression risk. We carried forward these ACs and tested the association of their circulating levels with Major Depressive Disorder (MDD) diagnosis, overall depression severity and specific symptom profiles. The sample from the Netherlands Study of Depression and Anxiety included participants with current (n = 1035) or remitted (n = 739) MDD and healthy controls (n = 800). Plasma levels of four ACs (short-chain: acetylcarnitine C2 and propionylcarnitine C3; medium-chain: octanoylcarnitine C8 and decanoylcarnitine C10) were measured. Overall depression severity as well as atypical/energy-related (AES), anhedonic and melancholic symptom profiles were derived from the Inventory of Depressive Symptomatology. As compared to healthy controls, subjects with current or remitted MDD presented similarly lower mean C2 levels (Cohen's d = 0.2, p ≤ 1e-4). Higher overall depression severity was significantly associated with higher C3 levels (ß=0.06, SE = 0.02, p = 1.21e-3). No associations were found for C8 and C10. Focusing on symptom profiles, only higher AES scores were linked to lower C2 (ß=-0.05, SE = 0.02, p = 1.85e-2) and higher C3 (ß=0.08, SE = 0.02, p = 3.41e-5) levels. Results were confirmed in analyses pooling data with an additional internal replication sample from the same subjects measured at 6-year follow-up (totaling 4141 observations). Small alterations in levels of short-chain acylcarnitine levels were related to the presence and severity of depression, especially for symptoms reflecting altered energy homeostasis. Cellular metabolic dysfunctions may represent a key pathway in depression pathophysiology potentially accessible through AC metabolism.
Collapse
Affiliation(s)
| | | | - Rick Jansen
- Amsterdam UMC location Vrije Universiteit Amsterdam
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Yoon JH, Lee H, Kwon D, Lee D, Lee S, Cho E, Kim J, Kim D. Integrative approach of omics and imaging data to discover new insights for understanding brain diseases. Brain Commun 2024; 6:fcae265. [PMID: 39165479 PMCID: PMC11334939 DOI: 10.1093/braincomms/fcae265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 06/03/2024] [Accepted: 08/07/2024] [Indexed: 08/22/2024] Open
Abstract
Treatments that can completely resolve brain diseases have yet to be discovered. Omics is a novel technology that allows researchers to understand the molecular pathways underlying brain diseases. Multiple omics, including genomics, transcriptomics and proteomics, and brain imaging technologies, such as MRI, PET and EEG, have contributed to brain disease-related therapeutic target detection. However, new treatment discovery remains challenging. We focused on establishing brain multi-molecular maps using an integrative approach of omics and imaging to provide insights into brain disease diagnosis and treatment. This approach requires precise data collection using omics and imaging technologies, data processing and normalization. Incorporating a brain molecular map with the advanced technologies through artificial intelligence will help establish a system for brain disease diagnosis and treatment through regulation at the molecular level.
Collapse
Affiliation(s)
- Jong Hyuk Yoon
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Hagyeong Lee
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Dayoung Kwon
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Dongha Lee
- Cognitive Science Research Group, Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Seulah Lee
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Eunji Cho
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Jaehoon Kim
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Dayea Kim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI hub), Daegu 41061, Republic of Korea
| |
Collapse
|
15
|
Song Z, He J, Yu W, He C, Yang M, Li P, Li Z, Jian G, Cheng S. Exploring the multifaceted therapeutic mechanism of Schisanlactone E (XTS) in APP/PS1 mouse model of Alzheimer's disease through multi-omics analysis. Front Microbiol 2024; 15:1440564. [PMID: 39044957 PMCID: PMC11263214 DOI: 10.3389/fmicb.2024.1440564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 06/28/2024] [Indexed: 07/25/2024] Open
Abstract
Background Schisanlactone E, also known as XueTongSu (XTS), is an active compound extracted from the traditional Tujia medicine Kadsura heteroclita ("XueTong"). Recent studies highlight its anti-inflammatory and antioxidant properties, yet the mechanisms of XTS's therapeutic effects on Alzheimer's disease (AD) are unclear. This study aims to elucidate the therapeutic efficacy and mechanisms of XTS in AD. Methods Ten C57BL/6 mice were assigned to the control group (NC), and twenty APP/PS1 transgenic mice were randomly divided into the model group (M) (10 mice) and the XTS treatment group (Tre) (10 mice). After an acclimatization period of 7 days, intraperitoneal injections were administered over a 60-day treatment period. The NC and M groups received saline, while the Tre group received XTS at 2 mg/kg. Learning and memory abilities were assessed using the Morris Water Maze (MWM) test. Histopathological changes were evaluated using hematoxylin and eosin (HE) and Nissl staining, and immunofluorescence was used to assess pathological products and glial cell activation. Cytokine levels (IL-1β, IL-6, TNF-α) in the hippocampus were quantified by qPCR. 16S rDNA sequencing analyzed gut microbiota metabolic alterations, and metabolomic analysis was performed on cortical samples. The KEGG database was used to analyze the regulatory mechanisms of XTS in AD treatment. Results XTS significantly improved learning and spatial memory in APP/PS1 mice and ameliorated histopathological changes, reducing Aβ plaque aggregation and glial cell activation. XTS decreased the expression of inflammatory cytokines IL-1β, IL-6, and TNF-α. It also enhanced gut microbiota diversity, notably increasing Akkermansia species, and modulated levels of metabolites such as isosakuranetin, 5-KETE, 4-methylcatechol, and sphinganine. Pathway analysis indicated that XTS regulated carbohydrate metabolism, neuroactive ligand-receptor interactions, and alanine, aspartate, and glutamate metabolism, mitigating gut microbiota dysbiosis and metabolic disturbances. Conclusion XTS ameliorates cognitive deficits, pathological changes, and inflammatory responses in APP/PS1 mice. It significantly modulates the gut microbiota, particularly increasing Akkermansia abundance, and influences levels of key metabolites in both the gut and brain. These findings suggest that XTS exerts anti-AD effects through the microbial-gut-brain axis (MGBA).
Collapse
Affiliation(s)
- Zhenyan Song
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jiawei He
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Wenjing Yu
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Chunxiang He
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Miao Yang
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ping Li
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ze Li
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Gonghui Jian
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Shaowu Cheng
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan University of Chinese Medicine, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
16
|
Barzegar Behrooz A, Latifi‐Navid H, Lotfi J, Khodagholi F, Shojaei S, Ghavami S, Fahanik Babaei J. CSF amino acid profiles in ICV-streptozotocin-induced sporadic Alzheimer's disease in male Wistar rat: a metabolomics and systems biology perspective. FEBS Open Bio 2024; 14:1116-1132. [PMID: 38769074 PMCID: PMC11216934 DOI: 10.1002/2211-5463.13814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 03/19/2024] [Accepted: 04/24/2024] [Indexed: 05/22/2024] Open
Abstract
Alzheimer's disease (AD) is an increasingly important public health concern due to the increasing proportion of older individuals within the general population. The impairment of processes responsible for adequate brain energy supply primarily determines the early features of the aging process. Restricting brain energy supply results in brain hypometabolism prior to clinical symptoms and is anatomically and functionally associated with cognitive impairment. The present study investigated changes in metabolic profiles induced by intracerebroventricular-streptozotocin (ICV-STZ) in an AD-like animal model. To this end, male Wistar rats received a single injection of STZ (3 mg·kg-1) by ICV (2.5 μL into each ventricle for 5 min on each side). In the second week after receiving ICV-STZ, rats were tested for cognitive performance using the Morris Water Maze test and subsequently prepared for positron emission tomography (PET) to confirm AD-like symptoms. Tandem Mass Spectrometry (MS/MS) analysis was used to detect amino acid changes in cerebrospinal fluid (CFS) samples. Our metabolomics study revealed a reduction in the concentrations of various amino acids (alanine, arginine, aspartic acid, glutamic acid, glycine, isoleucine, methionine, phenylalanine, proline, serine, threonine, tryptophane, tyrosine, and valine) in CSF of ICV-STZ-treated animals as compared to controls rats. The results of the current study indicate amino acid levels could potentially be considered targets of nutritional and/or pharmacological interventions to interfere with AD progression.
Collapse
Affiliation(s)
- Amir Barzegar Behrooz
- Electrophysiology Research Center, Neuroscience InstituteTehran University of Medical SciencesIran
- Department of Human Anatomy and Cell Science, College of MedicineUniversity of ManitobaWinnipegCanada
| | - Hamid Latifi‐Navid
- Electrophysiology Research Center, Neuroscience InstituteTehran University of Medical SciencesIran
- Department of Molecular MedicineNational Institute of Genetic Engineering and BiotechnologyTehranIran
- School of Biological SciencesInstitute for Research in Fundamental Sciences (IPM)TehranIran
| | - Jabar Lotfi
- Growth and Development Research CenterTehran University of Medical SciencesIran
| | - Fariba Khodagholi
- Neuroscience Research CenterShahid Beheshti University of Medical SciencesTehranIran
| | - Shahla Shojaei
- Department of Human Anatomy and Cell Science, College of MedicineUniversity of ManitobaWinnipegCanada
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, College of MedicineUniversity of ManitobaWinnipegCanada
- Faculty of Medicine in ZabrzeUniversity of Technology in KatowiceZabrzePoland
- Research Institute of Oncology and HematologyCancer Care Manitoba‐University of ManitobaWinnipegCanada
- Children Hospital Research Institute of ManitobaUniversity of ManitobaWinnipegCanada
| | - Javad Fahanik Babaei
- Electrophysiology Research Center, Neuroscience InstituteTehran University of Medical SciencesIran
| |
Collapse
|
17
|
Teng CY, Kao NJ, Nguyen NTK, Lin CI, Cross TWL, Lin SH. Effects of xylo-oligosaccharide on gut microbiota, brain protein expression, and lipid profile induced by high-fat diet. J Nutr Biochem 2024; 129:109640. [PMID: 38583497 DOI: 10.1016/j.jnutbio.2024.109640] [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: 11/03/2023] [Revised: 03/31/2024] [Accepted: 04/03/2024] [Indexed: 04/09/2024]
Abstract
Midlife overweight and obesity are risk factors of cognitive decline and Alzheimer' s disease (AD) in late life. In addition to increasing risk of obesity and cognitive dysfunction, diets rich in fats also contributes to an imbalance of gut microbiota. Xylo-oligosaccharides (XOS) are a kind of prebiotic with several biological advantages, and can selectively promote the growth of beneficial microorganisms in the gut. To explore whether XOS can alleviate cognitive decline induced by high-fat diet (HFD) through improving gut microbiota composition, mice were fed with normal control or 60% HFD for 9 weeks to induce obesity. After that, mice were supplemented with XOS (30 g or 60 g/kg-diet) or without, respectively, for 12 weeks. The results showed that XOS inhibited weight gain, decreased epidydimal fat weight, and improved fasting blood sugar and blood lipids in mice. Additionally, XOS elevated spatial learning and memory function, decreased amyloid plaques accumulation, increased brain-derived neurotrophic factor levels, and improved neuroinflammation status in hippocampus. Changes in glycerolipids metabolism-associated lipid compounds caused by HFD in hippocampus were reversed after XOS intervention. On the other hand, after XOS intervention, increase in immune-mediated bacteria, Faecalibacterium was observed. In conclusion, XOS improved gut dysbiosis and ameliorated spatial learning and memory dysfunction caused by HFD by decreasing cognitive decline-associated biomarkers and changing lipid composition in hippocampus.
Collapse
Affiliation(s)
- Chu-Yun Teng
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei, Taiwan
| | - Ning-Jo Kao
- Department of Nutrition and Health Sciences, Kainan University, Taoyuan, Taiwan
| | - Ngan Thi Kim Nguyen
- Program of Nutrition Science, National Taiwan Normal University, Taipei, Taiwan
| | - Ching-I Lin
- Department of Nutrition and Health Sciences, Chang-Gung University of Science and Technology, Taoyuan, Taiwan
| | - Tzu-Wen L Cross
- Departmen of Nutrition Science, Purdue University, West Lafayette, Indiana, USA
| | - Shyh-Hsiang Lin
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei, Taiwan; School of Food Safety, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
18
|
Yu X, Tao J, Xiao T, Duan X. 4,4'-methylenediphenol reduces Aβ-induced toxicity in a Caenorhabditis elegans model of Alzheimer's disease. Front Aging Neurosci 2024; 16:1393721. [PMID: 38872629 PMCID: PMC11171718 DOI: 10.3389/fnagi.2024.1393721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/22/2024] [Indexed: 06/15/2024] Open
Abstract
Introduction Gastrodia elata Blume is a widely used medicinal and edible herb with a rich chemical composition. Moreover, prescriptions containing Gastrodia elata are commonly used for the prevention and treatment of cardiovascular, cerebrovascular, and aging-related diseases. Recent pharmacological studies have confirmed the antioxidant and neuroprotective effects of Gastrodia elata, and, in recent years, this herb has also been used in the treatment of Alzheimer's disease (AD) and other neurodegenerative disorders. We have previously shown that 4,4'-methylenediphenol, a key active ingredient of Gastrodia elata, can mitigate amyloid-β (Aβ)-induced paralysis in AD model worms as well as prolong the lifespan of the animals, thus displaying potential as a treatment of AD. Methods We investigated the effects of 4,4'-methylenediphenol on AD and aging through paralysis, lifespan, and behavioral assays. In addition, we determined the anti-AD effects of 4,4'-methylenediphenol by reactive oxygen species (ROS) assay, lipofuscin analysis, thioflavin S staining, metabolomics analysis, GFP reporter gene worm assay, and RNA interference assay and conducted in-depth studies on its mechanism of action. Results 4,4'-Methylenediphenol not only delayed paralysis onset and senescence in the AD model worms but also enhanced their motility and stress tolerance. Meanwhile, 4,4'-methylenediphenol treatment also reduced the contents of reactive oxygen species (ROS) and lipofuscin, and decreased Aβ protein deposition in the worms. Broad-spectrum targeted metabolomic analysis showed that 4,4'-methylenediphenol administration had a positive effect on the metabolite profile of the worms. In addition, 4,4'-methylenediphenol promoted the nuclear translocation of DAF-16 and upregulated the expression of SKN-1, SOD-3, and GST-4 in the respective GFP reporter lines, accompanied by an enhancement of antioxidant activity and a reduction in Aβ toxicity; importantly, our results suggested that these effects of 4,4'-methylenediphenol were mediated, at least partly, via the activation of DAF-16. Conclusion We have demonstrated that 4,4'-methylenediphenol can reduce Aβ-induced toxicity in AD model worms, suggesting that it has potential for development as an anti-AD drug. Our findings provide ideas and references for further research into the anti-AD effects of Gastrodia elata and its active ingredients.
Collapse
Affiliation(s)
| | | | | | - Xiaohua Duan
- Yunnan Key Laboratory of Dai and Yi Medicines, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| |
Collapse
|
19
|
Wang F, Wang A, Huang Y, Gao W, Xu Y, Zhang W, Guo G, Song W, Kong Y, Wang Q, Wang S, Shi F. Lipoproteins and metabolites in diagnosing and predicting Alzheimer's disease using machine learning. Lipids Health Dis 2024; 23:152. [PMID: 38773573 PMCID: PMC11107010 DOI: 10.1186/s12944-024-02141-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 05/09/2024] [Indexed: 05/24/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a chronic neurodegenerative disorder that poses a substantial economic burden. The Random forest algorithm is effective in predicting AD; however, the key factors influencing AD onset remain unclear. This study aimed to analyze the key lipoprotein and metabolite factors influencing AD onset using machine-learning methods. It provides new insights for researchers and medical personnel to understand AD and provides a reference for the early diagnosis, treatment, and early prevention of AD. METHODS A total of 603 participants, including controls and patients with AD with complete lipoprotein and metabolite data from the Alzheimer's disease Neuroimaging Initiative (ADNI) database between 2005 and 2016, were enrolled. Random forest, Lasso regression, and CatBoost algorithms were employed to rank and filter 213 lipoprotein and metabolite variables. Variables with consistently high importance rankings from any two methods were incorporated into the models. Finally, the variables selected from the three methods, with the participants' age, sex, and marital status, were used to construct a random forest predictive model. RESULTS Fourteen lipoprotein and metabolite variables were screened using the three methods, and 17 variables were included in the AD prediction model based on age, sex, and marital status of the participants. The optimal random forest modeling was constructed with "mtry" set to 3 and "ntree" set to 300. The model exhibited an accuracy of 71.01%, a sensitivity of 79.59%, a specificity of 65.28%, and an AUC (95%CI) of 0.724 (0.645-0.804). When Mean Decrease Accuracy and Gini were used to rank the proteins, age, phospholipids to total lipids ratio in intermediate-density lipoproteins (IDL_PL_PCT), and creatinine were among the top five variables. CONCLUSIONS Age, IDL_PL_PCT, and creatinine levels play crucial roles in AD onset. Regular monitoring of lipoproteins and their metabolites in older individuals is significant for early AD diagnosis and prevention.
Collapse
Affiliation(s)
- Fenglin Wang
- Department of Health Statistics, School of Public Health, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Aimin Wang
- Department of Health Statistics, School of Public Health, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Yiming Huang
- Department of Health Statistics, School of Public Health, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Wenfeng Gao
- Department of Rheumatology and Immunology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, 261031, China
| | - Yaqi Xu
- Department of Health Statistics, School of Public Health, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Wenjing Zhang
- Department of Health Statistics, School of Public Health, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Guiya Guo
- Department of Health Statistics, School of Public Health, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Wangchen Song
- Department of Health Statistics, School of Public Health, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Yujia Kong
- Department of Health Statistics, School of Public Health, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Qinghua Wang
- Department of Health Statistics, School of Public Health, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Suzhen Wang
- Department of Health Statistics, School of Public Health, Shandong Second Medical University, Weifang, Shandong, 261053, China.
| | - Fuyan Shi
- Department of Health Statistics, School of Public Health, Shandong Second Medical University, Weifang, Shandong, 261053, China.
| |
Collapse
|
20
|
Lin C, Tian Q, Guo S, Xie D, Cai Y, Wang Z, Chu H, Qiu S, Tang S, Zhang A. Metabolomics for Clinical Biomarker Discovery and Therapeutic Target Identification. Molecules 2024; 29:2198. [PMID: 38792060 PMCID: PMC11124072 DOI: 10.3390/molecules29102198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/10/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
As links between genotype and phenotype, small-molecule metabolites are attractive biomarkers for disease diagnosis, prognosis, classification, drug screening and treatment, insight into understanding disease pathology and identifying potential targets. Metabolomics technology is crucial for discovering targets of small-molecule metabolites involved in disease phenotype. Mass spectrometry-based metabolomics has implemented in applications in various fields including target discovery, explanation of disease mechanisms and compound screening. It is used to analyze the physiological or pathological states of the organism by investigating the changes in endogenous small-molecule metabolites and associated metabolism from complex metabolic pathways in biological samples. The present review provides a critical update of high-throughput functional metabolomics techniques and diverse applications, and recommends the use of mass spectrometry-based metabolomics for discovering small-molecule metabolite signatures that provide valuable insights into metabolic targets. We also recommend using mass spectrometry-based metabolomics as a powerful tool for identifying and understanding metabolic patterns, metabolic targets and for efficacy evaluation of herbal medicine.
Collapse
Affiliation(s)
- Chunsheng Lin
- Graduate School and Second Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (C.L.); (S.G.); (Y.C.); (Z.W.)
| | - Qianqian Tian
- Faculty of Social Sciences, The University of Hong Kong, Hong Kong 999077, China;
| | - Sifan Guo
- Graduate School and Second Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (C.L.); (S.G.); (Y.C.); (Z.W.)
- International Advanced Functional Omics Platform, Scientific Experiment Center, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Engineering Research Center for Biological Sample Resources of Major Diseases (First Affiliated Hospital of Hainan Medical University), Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Hainan Medical University, Xueyuan Road 3, Haikou 571199, China; (D.X.); (S.Q.); (S.T.)
| | - Dandan Xie
- International Advanced Functional Omics Platform, Scientific Experiment Center, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Engineering Research Center for Biological Sample Resources of Major Diseases (First Affiliated Hospital of Hainan Medical University), Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Hainan Medical University, Xueyuan Road 3, Haikou 571199, China; (D.X.); (S.Q.); (S.T.)
| | - Ying Cai
- Graduate School and Second Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (C.L.); (S.G.); (Y.C.); (Z.W.)
- International Advanced Functional Omics Platform, Scientific Experiment Center, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Engineering Research Center for Biological Sample Resources of Major Diseases (First Affiliated Hospital of Hainan Medical University), Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Hainan Medical University, Xueyuan Road 3, Haikou 571199, China; (D.X.); (S.Q.); (S.T.)
| | - Zhibo Wang
- Graduate School and Second Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (C.L.); (S.G.); (Y.C.); (Z.W.)
- International Advanced Functional Omics Platform, Scientific Experiment Center, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Engineering Research Center for Biological Sample Resources of Major Diseases (First Affiliated Hospital of Hainan Medical University), Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Hainan Medical University, Xueyuan Road 3, Haikou 571199, China; (D.X.); (S.Q.); (S.T.)
| | - Hang Chu
- Department of Biomedical Sciences, Beijing City University, Beijing 100193, China;
| | - Shi Qiu
- International Advanced Functional Omics Platform, Scientific Experiment Center, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Engineering Research Center for Biological Sample Resources of Major Diseases (First Affiliated Hospital of Hainan Medical University), Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Hainan Medical University, Xueyuan Road 3, Haikou 571199, China; (D.X.); (S.Q.); (S.T.)
| | - Songqi Tang
- International Advanced Functional Omics Platform, Scientific Experiment Center, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Engineering Research Center for Biological Sample Resources of Major Diseases (First Affiliated Hospital of Hainan Medical University), Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Hainan Medical University, Xueyuan Road 3, Haikou 571199, China; (D.X.); (S.Q.); (S.T.)
| | - Aihua Zhang
- Graduate School and Second Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (C.L.); (S.G.); (Y.C.); (Z.W.)
- International Advanced Functional Omics Platform, Scientific Experiment Center, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Engineering Research Center for Biological Sample Resources of Major Diseases (First Affiliated Hospital of Hainan Medical University), Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Hainan Medical University, Xueyuan Road 3, Haikou 571199, China; (D.X.); (S.Q.); (S.T.)
| |
Collapse
|
21
|
Ge Q, Lu H, Geng X, Chen X, Liu X, Sun H, Guo Z, Sun J, Qi F, Niu X, Wang A, He J, Sun W, Xu L. Serum metabolism alteration behind different etiology, diagnosis, and prognosis of disorders of consciousness. Chin Neurosurg J 2024; 10:12. [PMID: 38594757 PMCID: PMC11003070 DOI: 10.1186/s41016-024-00365-4] [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: 07/20/2023] [Accepted: 03/26/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND Patients with disorders of consciousness (DoC) exhibit varied revival outcomes based on different etiologies and diagnoses, the mechanisms of which remain largely unknown. The fluctuating clinical presentations in DoC pose challenges in accurately assessing consciousness levels and prognoses, often leading to misdiagnoses. There is an urgent need for a deeper understanding of the physiological changes in DoC and the development of objective diagnostic and prognostic biomarkers to improve treatment guidance. METHODS To explore biomarkers and understand the biological processes, we conducted a comprehensive untargeted metabolomic analysis on serum samples from 48 patients with DoC. Patients were categorized based on etiology (TBI vs. non-TBI), CRS-R scores, and prognosis. Advanced analytical techniques, including PCA and OPLS-DA models, were employed to identify differential metabolites. RESULTS Our analysis revealed a distinct separation in metabolomic profiles among the different groups. The primary differential metabolites distinguishing patients with varying etiologies were predominantly phospholipids, with a notable decrease in glycerophospholipids observed in the TBI group. Patients with higher CRS-R scores exhibited a pattern of impaired carbohydrate metabolism coupled with enhanced lipid metabolism. Notably, serum concentrations of both LysoPE and PE were reduced in patients with improved outcomes, suggesting their potential as prognostic biomarkers. CONCLUSIONS Our study underscores the critical role of phospholipid metabolism in the brain's metabolic alterations in patients with DoC. It identifies key biomarkers for diagnosis and prognosis, offering insights that could lead to novel therapeutic targets. These findings highlight the value of metabolomic profiling in understanding and potentially treating DoC.
Collapse
Affiliation(s)
- Qianqian Ge
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hezhen Lu
- China-Japan Union Hospital of Jilin University, Changchun, China
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Xiaoli Geng
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xueling Chen
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiaoyan Liu
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Haidan Sun
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Zhengguang Guo
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Jiameng Sun
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Feng Qi
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Xia Niu
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Aiwei Wang
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Jianghong He
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases (NCRC-ND), Beijing, China
| | - Wei Sun
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China.
| | - Long Xu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- China National Clinical Research Center for Neurological Diseases (NCRC-ND), Beijing, China.
| |
Collapse
|
22
|
Liu S, Zhong H, Zhu J, Wu L. Identification of blood metabolites associated with risk of Alzheimer's disease by integrating genomics and metabolomics data. Mol Psychiatry 2024; 29:1153-1162. [PMID: 38216726 PMCID: PMC11176029 DOI: 10.1038/s41380-023-02400-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 12/17/2023] [Accepted: 12/22/2023] [Indexed: 01/14/2024]
Abstract
Specific metabolites have been reported to be potentially associated with Alzheimer's disease (AD) risk. However, the comprehensive understanding of roles of metabolite biomarkers in AD etiology remains elusive. We performed a large AD metabolome-wide association study (MWAS) by developing blood metabolite genetic prediction models. We evaluated associations between genetically predicted levels of metabolites and AD risk in 39,106 clinically diagnosed AD cases, 46,828 proxy AD and related dementia (proxy-ADD) cases, and 401,577 controls. We further conducted analyses to determine microbiome features associated with the detected metabolites and characterize associations between predicted microbiome feature levels and AD risk. We identified fourteen metabolites showing an association with AD risk. Five microbiome features were further identified to be potentially related to associations of five of the metabolites. Our study provides new insights into the etiology of AD that involves blood metabolites and gut microbiome, which warrants further investigation.
Collapse
Affiliation(s)
- Shuai Liu
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Hua Zhong
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Jingjing Zhu
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Lang Wu
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, HI, USA.
| |
Collapse
|
23
|
Liu J, Tan Y, Zhang F, Wang Y, Chen S, Zhang N, Dai W, Zhou L, Li J. Metabolomic analysis of plasma biomarkers in children with autism spectrum disorders. MedComm (Beijing) 2024; 5:e488. [PMID: 38420161 PMCID: PMC10901282 DOI: 10.1002/mco2.488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 01/10/2024] [Accepted: 01/22/2024] [Indexed: 03/02/2024] Open
Abstract
Autism spectrum disorder (ASD) presents a significant risk to human well-being and has emerged as a worldwide public health concern. Twenty-eight children with ASD and 33 healthy children (HC) were selected for the quantitative determination of their plasma metabolites using an ultraperformance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) platform. A total of 1997 metabolites were detected in the study cohort, from which 116 metabolites were found to be differentially expressed between the ASD and HC groups. Through analytical algorithms such as least absolute shrinkage selection operator (LASSO), support vector machine (SVM), and random forest (RF), three potential metabolic markers were identified as FAHFA (18:1(9Z)/9-O-18:0), DL-2-hydroxystearic acid, and 7(S),17(S)-dihydroxy-8(E),10(Z),13(Z),15(E),19(Z)-docosapentaenoic acid. These metabolites demonstrated superior performance in distinguishing the ASD group from the HC group, as indicated by the area under curves (AUCs) of 0.935, 0.897, and 0.963 for the three candidate biomarkers, respectively. The samples were divided into training and validation sets according to 7:3. Diagnostic models were constructed using logistic regression (LR), SVM, and RF. The constructed three-biomarker diagnostic model also exhibited strong discriminatory efficacy. These findings contribute to advancing our understanding of the underlying mechanisms involved in the occurrence of ASD and provide a valuable reference for clinical diagnosis.
Collapse
Affiliation(s)
- Jun Liu
- Medical Research CenterYue Bei People's Hospital, Shantou University Medical CollegeShaoguanChina
| | - Yuhua Tan
- Shaoguan Maternal and Child Health HospitalShaoguanChina
| | - Fan Zhang
- Medical Research CenterYue Bei People's Hospital, Shantou University Medical CollegeShaoguanChina
| | - Yan Wang
- Shaoguan Maternal and Child Health HospitalShaoguanChina
| | - Shu Chen
- Shaoguan Maternal and Child Health HospitalShaoguanChina
| | - Na Zhang
- Shaoguan Maternal and Child Health HospitalShaoguanChina
| | - Wenjie Dai
- Medical Research CenterYue Bei People's Hospital, Shantou University Medical CollegeShaoguanChina
| | - Liqing Zhou
- Medical Research CenterYue Bei People's Hospital, Shantou University Medical CollegeShaoguanChina
| | - Ji‐Cheng Li
- Medical Research CenterYue Bei People's Hospital, Shantou University Medical CollegeShaoguanChina
- Institute of Cell BiologyZhejiang UniversityHangzhouChina
- Major Disease Biomarkers Research LaboratorySchool of Basic Medical Science, Henan UniversityKaifengChina
| |
Collapse
|
24
|
Flores AC, Zhang X, Kris-Etherton PM, Sliwinski MJ, Shearer GC, Gao X, Na M. Metabolomics and Risk of Dementia: A Systematic Review of Prospective Studies. J Nutr 2024; 154:826-845. [PMID: 38219861 DOI: 10.1016/j.tjnut.2024.01.012] [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: 10/31/2023] [Revised: 01/05/2024] [Accepted: 01/10/2024] [Indexed: 01/16/2024] Open
Abstract
BACKGROUND The projected increase in the prevalence of dementia has sparked interest in understanding the pathophysiology and underlying causal factors in its development and progression. Identifying novel biomarkers in the preclinical or prodromal phase of dementia may be important for predicting early disease risk. Applying metabolomic techniques to prediagnostic samples in prospective studies provides the opportunity to identify potential disease biomarkers. OBJECTIVE The objective of this systematic review was to summarize the evidence on the associations between metabolite markers and risk of dementia and related dementia subtypes in human studies with a prospective design. DESIGN We searched PubMed, PsycINFO, and Web of Science databases from inception through December 8, 2023. Thirteen studies (mean/median follow-up years: 2.1-21.0 y) were included in the review. RESULTS Several metabolites detected in biological samples, including amino acids, fatty acids, acylcarnitines, lipid and lipoprotein variations, hormones, and other related metabolites, were associated with risk of developing dementia. Our systematic review summarized the adjusted associations between metabolites and dementia risk; however, our findings should be interpreted with caution because of the heterogeneity across the included studies and potential sources of bias. Further studies are warranted with well-designed prospective cohort studies that have defined study populations, longer follow-up durations, the inclusion of additional diverse biological samples, standardization of techniques in metabolomics and ascertainment methods for diagnosing dementia, and inclusion of other related dementia subtypes. CONCLUSIONS This study contributes to the limited systematic reviews on metabolomics and dementia by summarizing the prospective associations between metabolites in prediagnostic biological samples with dementia risk. Our review discovered additional metabolite markers associated with the onset of developing dementia and may help aid in the understanding of dementia etiology. The protocol is registered in the International Prospective Register of Systematic Reviews (PROSPERO) database (https://www.crd.york.ac.uk/prospero/; registration ID: CRD42022357521).
Collapse
Affiliation(s)
- Ashley C Flores
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, United States
| | - Xinyuan Zhang
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Penny M Kris-Etherton
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, United States
| | - Martin J Sliwinski
- Center for Healthy Aging, The Pennsylvania State University, University Park, PA, United States; Department of Human Development and Family Studies, The Pennsylvania State University, University Park, PA, United States
| | - Greg C Shearer
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, United States
| | - Xiang Gao
- School of Public Health, Institute of Nutrition, Fudan University, Shanghai, China.
| | - Muzi Na
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, United States.
| |
Collapse
|
25
|
Compton H, Smith ML, Bull C, Korologou-Linden R, Ben-Shlomo Y, Bell JA, Williams DM, Anderson EL. Life course plasma metabolomic signatures of genetic liability to Alzheimer's disease. Sci Rep 2024; 14:3896. [PMID: 38365930 PMCID: PMC10873397 DOI: 10.1038/s41598-024-54569-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 02/14/2024] [Indexed: 02/18/2024] Open
Abstract
Mechanisms through which most known Alzheimer's disease (AD) loci operate to increase AD risk remain unclear. Although Apolipoprotein E (APOE) is known to regulate lipid homeostasis, the effects of broader AD genetic liability on non-lipid metabolites remain unknown, and the earliest ages at which metabolic perturbations occur and how these change over time are yet to be elucidated. We examined the effects of AD genetic liability on the plasma metabolome across the life course. Using a reverse Mendelian randomization framework in two population-based cohorts [Avon Longitudinal Study of Parents and Children (ALSPAC, n = 5648) and UK Biobank (n ≤ 118,466)], we estimated the effects of genetic liability to AD on 229 plasma metabolites, at seven different life stages, spanning 8 to 73 years. We also compared the specific effects of APOE ε4 and APOE ε2 carriage on metabolites. In ALSPAC, AD genetic liability demonstrated the strongest positive associations with cholesterol-related traits, with similar magnitudes of association observed across all age groups including in childhood. In UK Biobank, the effect of AD liability on several lipid traits decreased with age. Fatty acid metabolites demonstrated positive associations with AD liability in both cohorts, though with smaller magnitudes than lipid traits. Sensitivity analyses indicated that observed effects are largely driven by the strongest AD instrument, APOE, with many contrasting effects observed on lipids and fatty acids for both ε4 and ε2 carriage. Our findings indicate pronounced effects of the ε4 and ε2 genetic variants on both pro- and anti-atherogenic lipid traits and sphingomyelins, which begin in childhood and either persist into later life or appear to change dynamically.
Collapse
Affiliation(s)
- Hannah Compton
- Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, UK
| | - Madeleine L Smith
- Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, UK
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Caroline Bull
- Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, UK
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- School of Translational Health Sciences, University of Bristol, Bristol, UK
| | - Roxanna Korologou-Linden
- Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, UK
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Yoav Ben-Shlomo
- Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, UK
| | - Joshua A Bell
- Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, UK
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Dylan M Williams
- MRC Unit for Lifelong Health & Ageing at UCL, University College London, London, UK
| | - Emma L Anderson
- Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, UK.
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK.
- Division of Psychiatry, University College London, 149 Tottenham Court Road, London, W1T 7NF, UK.
| |
Collapse
|
26
|
Zhuang H, Cao X, Tang X, Zou Y, Yang H, Liang Z, Yan X, Chen X, Feng X, Shen L. Investigating metabolic dysregulation in serum of triple transgenic Alzheimer's disease male mice: implications for pathogenesis and potential biomarkers. Amino Acids 2024; 56:10. [PMID: 38315232 PMCID: PMC10844422 DOI: 10.1007/s00726-023-03375-1] [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: 05/15/2023] [Accepted: 11/11/2023] [Indexed: 02/07/2024]
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disease that lacks convenient and accessible peripheral blood diagnostic markers and effective drugs. Metabolic dysfunction is one of AD risk factors, which leaded to alterations of various metabolites in the body. Pathological changes of the brain can be reflected in blood metabolites that are expected to explain the disease mechanisms or be candidate biomarkers. The aim of this study was to investigate the changes of targeted metabolites within peripheral blood of AD mouse model, with the purpose of exploring the disease mechanism and potential biomarkers. Targeted metabolomics was used to quantify 256 metabolites in serum of triple transgenic AD (3 × Tg-AD) male mice. Compared with controls, 49 differential metabolites represented dysregulation in purine, pyrimidine, tryptophan, cysteine and methionine and glycerophospholipid metabolism. Among them, adenosine, serotonin, N-acetyl-5-hydroxytryptamine, and acetylcholine play a key role in regulating neural transmitter network. The alteration of S-adenosine-L-homocysteine, S-adenosine-L-methionine, and trimethylamine-N-oxide in AD mice serum can served as indicator of AD risk. The results revealed the changes of metabolites in serum, suggesting that metabolic dysregulation in periphery in AD mice may be related to the disturbances in neuroinhibition, the serotonergic system, sleep function, the cholinergic system, and the gut microbiota. This study provides novel insights into the dysregulation of several key metabolites and metabolic pathways in AD, presenting potential avenues for future research and the development of peripheral biomarkers.
Collapse
Affiliation(s)
- Hongbin Zhuang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Xueshan Cao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Xiaoxiao Tang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Yongdong Zou
- Center for Instrumental Analysis, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Hongbo Yang
- Center for Instrumental Analysis, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Zhiyuan Liang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Xi Yan
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, People's Republic of China
| | - Xiaolu Chen
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, People's Republic of China
| | - Xingui Feng
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Liming Shen
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China.
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, People's Republic of China.
| |
Collapse
|
27
|
Bhalala OG, Watson R, Yassi N. Multi-Omic Blood Biomarkers as Dynamic Risk Predictors in Late-Onset Alzheimer's Disease. Int J Mol Sci 2024; 25:1231. [PMID: 38279230 PMCID: PMC10816901 DOI: 10.3390/ijms25021231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024] Open
Abstract
Late-onset Alzheimer's disease is the leading cause of dementia worldwide, accounting for a growing burden of morbidity and mortality. Diagnosing Alzheimer's disease before symptoms are established is clinically challenging, but would provide therapeutic windows for disease-modifying interventions. Blood biomarkers, including genetics, proteins and metabolites, are emerging as powerful predictors of Alzheimer's disease at various timepoints within the disease course, including at the preclinical stage. In this review, we discuss recent advances in such blood biomarkers for determining disease risk. We highlight how leveraging polygenic risk scores, based on genome-wide association studies, can help stratify individuals along their risk profile. We summarize studies analyzing protein biomarkers, as well as report on recent proteomic- and metabolomic-based prediction models. Finally, we discuss how a combination of multi-omic blood biomarkers can potentially be used in memory clinics for diagnosis and to assess the dynamic risk an individual has for developing Alzheimer's disease dementia.
Collapse
Affiliation(s)
- Oneil G. Bhalala
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; (R.W.); (N.Y.)
- Department of Neurology, Melbourne Brain Centre at The Royal Melbourne Hospital, University of Melbourne, Parkville 3050, Australia
| | - Rosie Watson
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; (R.W.); (N.Y.)
- Department of Medicine, The Royal Melbourne Hospital, University of Melbourne, Parkville 3050, Australia
| | - Nawaf Yassi
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; (R.W.); (N.Y.)
- Department of Neurology, Melbourne Brain Centre at The Royal Melbourne Hospital, University of Melbourne, Parkville 3050, Australia
- Department of Medicine, The Royal Melbourne Hospital, University of Melbourne, Parkville 3050, Australia
| |
Collapse
|
28
|
He J, Jin Y, He C, Li Z, Yu W, Zhou J, Luo R, Chen Q, Wu Y, Wang S, Song Z, Cheng S. Danggui Shaoyao San: comprehensive modulation of the microbiota-gut-brain axis for attenuating Alzheimer's disease-related pathology. Front Pharmacol 2024; 14:1338804. [PMID: 38283834 PMCID: PMC10811133 DOI: 10.3389/fphar.2023.1338804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 12/26/2023] [Indexed: 01/30/2024] Open
Abstract
Background: Alzheimer's disease (AD), an age-associated neurodegenerative disorder, currently lacks effective clinical therapeutics. Traditional Chinese Medicine (TCM) holds promising potential in AD treatment, exemplified by Danggui Shaoyao San (DSS), a TCM formulation. The precise therapeutic mechanisms of DSS in AD remain to be fully elucidated. This study aims to uncover the therapeutic efficacy and underlying mechanisms of DSS in AD, employing an integrative approach encompassing gut microbiota and metabolomic analyses. Methods: Thirty Sprague-Dawley (SD) rats were allocated into three groups: Blank Control (Con), AD Model (M), and Danggui Shaoyao San (DSS). AD models were established via bilateral intracerebroventricular injections of streptozotocin (STZ). DSS was orally administered at 24 g·kg-1·d-1 (weight of raw herbal materials) for 14 days. Cognitive functions were evaluated using the Morris Water Maze (MWM) test. Pathological alterations were assessed through hematoxylin and eosin (HE) staining. Bloodstream metabolites were characterized, gut microbiota profiled through 16S rDNA sequencing, and cortical metabolomics analyzed. Hippocampal proinflammatory cytokines (IL-1β, IL-6, TNF-α) were quantified using RT-qPCR, and oxidative stress markers (SOD, CAT, GSH-PX, MDA) in brain tissues were measured with biochemical assays. Results: DSS identified a total of 1,625 bloodstream metabolites, predominantly Benzene derivatives, Carboxylic acids, and Fatty Acyls. DSS significantly improved learning and spatial memory in AD rats and ameliorated cerebral tissue pathology. The formulation enriched the probiotic Ligilactobacillus, modulating metabolites like Ophthalmic acid (OA), Phosphocreatine (PCr), Azacridone A, Inosine, and NAD. DSS regulated Purine and Nicotinate-nicotinamide metabolism, restoring balance in the Candidatus Saccharibacteria-OA interplay and stabilizing gut microbiota-metabolite homeostasis. Additionally, DSS reduced hippocampal IL-1β, IL-6, TNF-α expression, attenuating the inflammatory state. It elevated antioxidative enzymes (SOD, CAT, GSH-PX) while reducing MDA levels, indicating diminished oxidative stress in AD rat brains. Conclusion: DSS addresses AD pathology through multifaceted mechanisms, encompassing gut microbiome regulation, specific metabolite modulation, and the mitigation of inflammation and oxidative stress within the brain. This holistic intervention through the Microbial-Gut-Brain Axis (MGBA) underscores DSS's potential as an integrative therapeutic agent in combatting AD.
Collapse
Affiliation(s)
- Jiawei He
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yijie Jin
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Chunxiang He
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ze Li
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Wenjing Yu
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jinyong Zhou
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Rongsiqing Luo
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Qi Chen
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yixiao Wu
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Shiwei Wang
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Zhenyan Song
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Shaowu Cheng
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Office of Science and Technology, Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
29
|
Vardarajan B, Kalia V, Reyes-Dumeyer D, Dubey S, Nandakumar R, Lee A, Lantigua R, Medrano M, Rivera D, Honig L, Mayeux R, Miller G. Lysophosphatidylcholines are associated with P-tau181 levels in early stages of Alzheimer's Disease. RESEARCH SQUARE 2024:rs.3.rs-3346076. [PMID: 38260644 PMCID: PMC10802729 DOI: 10.21203/rs.3.rs-3346076/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Background We profiled circulating plasma metabolites to identify systemic biochemical changes in clinical and biomarker-assisted diagnosis of Alzheimer's disease (AD). Methods We used an untargeted approach with liquid chromatography coupled to high-resolution mass spectrometry to measure small molecule plasma metabolites from 150 clinically diagnosed AD patients and 567 age-matched healthy elderly of Caribbean Hispanic ancestry. Plasma biomarkers of AD were measured including P-tau181, Aβ40, Aβ42, total-tau, neurofilament light chain (NfL) and glial fibrillary acidic protein (GFAP). Association of individual and co-abundant modules of metabolites were tested with clinical diagnosis of AD, as well as biologically-defined AD pathological process based on P-tau181 and other biomarker levels. Results Over 6000 metabolomic features were measured with high accuracy. First principal component (PC) of lysophosphatidylcholines (lysoPC) that bind to or interact with docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA) and arachidonic acid (AHA) was associated with decreased risk of AD (OR = 0.91 [0.89-0.96], p = 2e-04). Association was restricted to individuals without an APOE ε4 allele (OR = 0.89 [0.84-0.94], p = 8.7e-05). Among individuals carrying at least one APOE ε4 allele, PC4 of lysoPCs moderately increased risk of AD (OR = 1.37 [1.16-1.6], p = 1e-04). Essential amino acids including tyrosine metabolism pathways were enriched among metabolites associated with P-tau181 levels and heparan and keratan sulfate degradation pathways were associated with Aβ42/Aβ40 ratio. Conclusions Unbiased metabolic profiling can identify critical metabolites and pathways associated with β-amyloid and phosphotau pathology. We also observed an APOE-ε4 dependent association of lysoPCs with AD and biologically based diagnostic criteria may aid in the identification of unique pathogenic mechanisms.
Collapse
Affiliation(s)
| | - Vrinda Kalia
- Columbia University Mailman School of Public Health
| | | | | | | | - Annie Lee
- Center for Translational & Computational Neuroimmunology
| | | | | | | | | | | | | |
Collapse
|
30
|
Pausova Z, Sliz E. Large-Scale Population-Based Studies of Blood Metabolome and Brain Health. Curr Top Behav Neurosci 2024; 68:177-219. [PMID: 38509405 DOI: 10.1007/7854_2024_463] [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] [Indexed: 03/22/2024]
Abstract
Metabolomics technologies enable the quantification of multiple metabolomic measures simultaneously, which provides novel insights into molecular aspects of human health and disease. In large-scale, population-based studies, blood is often the preferred biospecimen. Circulating metabolome may relate to brain health either by affecting or reflecting brain metabolism. Peripheral metabolites may act at or cross the blood-brain barrier and, subsequently, influence brain metabolism, or they may reflect brain metabolism if similar pathways are engaged. Peripheral metabolites may also include those penetrating the circulation from the brain, indicating, for example, brain damage. Most brain health-related metabolomics studies have been conducted in the context of neurodegenerative disorders and cognition, but some studies have also focused on neuroimaging markers of these disorders. Moreover, several metabolomics studies of neurodevelopmental disorders have been performed. Here, we provide a brief background on the types of blood metabolites commonly assessed, and we review the literature describing the relationships between human blood metabolome (n > 50 metabolites) and brain health reported in large-scale studies (n > 500 individuals).
Collapse
Affiliation(s)
- Zdenka Pausova
- Centre hospitalier universitaire Sainte-Justine and Department of Pediatrics, University of Montreal, Montreal, QC, Canada
| | - Eeva Sliz
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland.
| |
Collapse
|
31
|
Miao G, Yu L, Yang J, Bennett DA, Zhao J, Wu SS. Learning from vertically distributed data across multiple sites: An efficient privacy-preserving algorithm for Cox proportional hazards model with variable selection. J Biomed Inform 2024; 149:104581. [PMID: 38142903 PMCID: PMC10996392 DOI: 10.1016/j.jbi.2023.104581] [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: 05/21/2023] [Revised: 08/24/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023]
Abstract
OBJECTIVE To develop a lossless distributed algorithm for regularized Cox proportional hazards model with variable selection to support federated learning for vertically distributed data. METHODS We propose a novel distributed algorithm for fitting regularized Cox proportional hazards model when data sharing among different data providers is restricted. Based on cyclical coordinate descent, the proposed algorithm computes intermediary statistics by each site and then exchanges them to update the model parameters in other sites without accessing individual patient-level data. We evaluate the performance of the proposed algorithm with (1) a simulation study and (2) a real-world data analysis predicting the risk of Alzheimer's dementia from the Religious Orders Study and Rush Memory and Aging Project (ROSMAP). Moreover, we compared the performance of our method with existing privacy-preserving models. RESULTS Our algorithm achieves privacy-preserving variable selection for time-to-event data in the vertically distributed setting, without degradation of accuracy compared with a centralized approach. Simulation demonstrates that our algorithm is highly efficient in analyzing high-dimensional datasets. Real-world data analysis reveals that our distributed Cox model yields higher accuracy in predicting the risk of Alzheimer's dementia than the conventional Cox model built by each data provider without data sharing. Moreover, our algorithm is computationally more efficient compared with existing privacy-preserving Cox models with or without regularization term. CONCLUSION The proposed algorithm is lossless, privacy-preserving and highly efficient to fit regularized Cox model for vertically distributed data. It provides a suitable and convenient approach for modeling time-to-event data in a distributed manner.
Collapse
Affiliation(s)
- Guanhong Miao
- Department of Epidemiology, College of Public Health & Health Professions and College of Medicine, University of Florida, Gainesville, FL, USA; Center for Genetic Epidemiology and Bioinformatics, University of Florida, Gainesville, FL, USA; Department of Biostatistics, College of Public Health & Health Professions and College of Medicine, University of Florida, Gainesville, FL, USA
| | - Lei Yu
- Rush Alzheimer's Disease Center & Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Jingyun Yang
- Rush Alzheimer's Disease Center & Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center & Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Jinying Zhao
- Department of Epidemiology, College of Public Health & Health Professions and College of Medicine, University of Florida, Gainesville, FL, USA; Center for Genetic Epidemiology and Bioinformatics, University of Florida, Gainesville, FL, USA
| | - Samuel S Wu
- Department of Biostatistics, College of Public Health & Health Professions and College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
32
|
Tang Y, Chen S, Wang S, Xu K, Zhang K, Wang D, Feng N. Decanoylcarnitine Inhibits Triple-Negative Breast Cancer Progression via Mmp9 in an Intermittent Fasting Obesity Mouse. Technol Cancer Res Treat 2024; 23:15330338241233443. [PMID: 38409962 PMCID: PMC10898300 DOI: 10.1177/15330338241233443] [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] [Indexed: 02/28/2024] Open
Abstract
Purpose: Treatment of triple-negative breast cancer (TNBC) remains challenging. Intermittent fasting (IF) has emerged as a promising approach to improve metabolic health of various metabolic disorders. Clinical studies indicate IF is essential for TNBC progression. However, the molecular mechanisms underlying metabolic remodeling in regulating IF and TNBC progression are still unclear. Methods: In this study, we utilized a robust mouse model of TNBC and exposed subjects to a high-fat diet (HFD) with IF to explore its impact on the metabolic reprogramming linked to cancer progression. To identify crucial serum metabolites and signaling events, we utilized targeted metabolomics and RNA sequencing (RNA-seq). Furthermore, we conducted immunoblotting, real-time quantitative polymerase chain reaction (RT-qPCR), cell migration assays, lentivirus-mediated Mmp9 overexpression, and Mmp9 inhibitor experiments to elucidate the role of decanoylcarnitine/Mmp9 in TNBC cell migration. Results: Our observations indicate that IF exerts notable inhibitory effects on both the proliferation and cancer metastasis. Utilizing targeted metabolomics and RNA-seq, we initially identified pivotal serum metabolites and signaling events in the progression of TNBC. Among the 349 serum metabolites identified, decanoylcarnitine was picked out to inhibit TNBC cell proliferation and migration. RNA-seq analysis of TNBC cells treated with decanoylcarnitine revealed its suppressive effects on extracellular matrix-related protein components, with a notable reduction observed in Mmp9. Further investigations confirmed that decanoylcarnitine could inhibit Mmp9 expression in TNBC cells, primary tumors, lung, and liver metastasis tissues. Mmp9 overexpression abolished the inhibitory effect of decanoylcarnitine on cell migration. Conclusion: This study pioneers the exploration of IF intervention and the role of decanoylcarnitine/Mmp9 in the progression of TNBC in obese mice, enhancing our comprehension of the potential roles of various dietary patterns in the process of cancer treatment.
Collapse
Affiliation(s)
- Yifan Tang
- Department of Urology, Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - Shuai Chen
- Department of Gastrointestinal Surgery, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Saijun Wang
- Department of Gastrointestinal Surgery, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Ke Xu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, National Health Commission Key Laboratory of Antibody Techniques, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Kun Zhang
- Department of Radiotherapy, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Dongmei Wang
- Department of Gastrointestinal Surgery, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Ninghan Feng
- Department of Urology, Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| |
Collapse
|
33
|
Snytnikova O, Telegina D, Savina E, Tsentalovich Y, Kolosova N. Quantitative Metabolomic Analysis of the Rat Hippocampus: Effects of Age and of the Development of Alzheimer's Disease-Like Pathology. J Alzheimers Dis 2024; 99:S327-S344. [PMID: 37980669 DOI: 10.3233/jad-230706] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2023]
Abstract
Background Alzheimer's disease (AD) is the most common type of dementia in the elderly. Incomplete knowledge about the pathogenesis of this disease determines the absence of medications for the treatment of AD today. Animal models can provide the necessary knowledge to understand the mechanisms of biochemical processes occurring in the body in health and disease. Objective To identify the most promising metabolomic predictors and biomarkers reflecting metabolic disorders in the development of AD signs. Methods High resolution 1H NMR spectroscopy was used for quantitative metabolomic profiling of the hippocampus of OXYS rats, an animal model of sporadic AD, which demonstrates key characteristics of this disease. Animals were examined during several key periods: 20 days group corresponds to the "preclinical" period preceding the development of AD signs, during their manifestation (3 months), and active progression (18 months). Wistar rats of the same age were used as control. Results Ranges of variation and mean concentrations were established for 59 brain metabolites. The main metabolic patterns during aging, which are involved in energy metabolism pathways and metabolic shifts of neurotransmitters, have been established. Of particular note is the significant increase of scyllo-inositol and decrease of hypotaurine in the hippocampus of OXYS rats as compared to Wistars for all studied age groups. Conclusions We suggest that the accumulation of scyllo-inositol and the reduction of hypotaurine in the brain, even at an early age, can be considered as predictors and potential biomarkers of the development of AD signs in OXYS rats and, probably, in humans.
Collapse
Affiliation(s)
- Olga Snytnikova
- International Tomography Center, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Darya Telegina
- The Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Ekaterina Savina
- International Tomography Center, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Yuri Tsentalovich
- International Tomography Center, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Nataliya Kolosova
- The Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
34
|
Chen Y, Li Y, Fan Y, Chen S, Chen L, Chen Y, Chen Y. Gut microbiota-driven metabolic alterations reveal gut-brain communication in Alzheimer's disease model mice. Gut Microbes 2024; 16:2302310. [PMID: 38261437 PMCID: PMC10807476 DOI: 10.1080/19490976.2024.2302310] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 01/03/2024] [Indexed: 01/25/2024] Open
Abstract
The gut microbiota (GM) and its metabolites affect the host nervous system and are involved in the pathogeneses of various neurological diseases. However, the specific GM alterations under pathogenetic pressure and their contributions to the "microbiota - metabolite - brain axis" in Alzheimer's disease (AD) remain unclear. Here, we investigated the GM and the fecal, serum, cortical metabolomes in APP/PS1 and wild-type (WT) mice, revealing distinct hub bacteria in AD mice within scale-free GM networks shared by both groups. Moreover, we identified diverse peripheral - central metabolic landscapes between AD and WT mice that featured bile acids (e.g. deoxycholic and isodeoxycholic acid) and unsaturated fatty acids (e.g. 11Z-eicosenoic and palmitoleic acid). Machine-learning models revealed the relationships between the differential/hub bacteria and these metabolic signatures from the periphery to the brain. Notably, AD-enriched Dubosiella affected AD occurrence via cortical palmitoleic acid and vice versa. Considering the transgenic background of the AD mice, we propose that Dubosiella enrichment impedes AD progression via the synthesis of palmitoleic acid, which has protective properties against inflammation and metabolic disorders. We identified another association involving fecal deoxycholic acid-mediated interactions between the AD hub bacteria Erysipelatoclostridium and AD occurrence, which was corroborated by the correlation between deoxycholate levels and cognitive scores in humans. Overall, this study elucidated the GM network alterations, contributions of the GM to peripheral - central metabolic landscapes, and mediatory roles of metabolites between the GM and AD occurrence, thus revealing the critical roles of bacteria in AD pathogenesis and gut - brain communications under pathogenetic pressure.
Collapse
Affiliation(s)
- Yijing Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen–Hong Kong Institute of Brain Science–Shenzhen Fundamental Research Institutions, Shenzhen, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, China
| | - Yinhu Li
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen–Hong Kong Institute of Brain Science–Shenzhen Fundamental Research Institutions, Shenzhen, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, China
| | - Yingying Fan
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen–Hong Kong Institute of Brain Science–Shenzhen Fundamental Research Institutions, Shenzhen, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, China
| | - Shuai Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen–Hong Kong Institute of Brain Science–Shenzhen Fundamental Research Institutions, Shenzhen, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, China
| | - Li Chen
- Department of Neurology, Shenzhen Children’s Hospital, Shenzhen, China
| | - Yuewen Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen–Hong Kong Institute of Brain Science–Shenzhen Fundamental Research Institutions, Shenzhen, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, China
| | - Yu Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen–Hong Kong Institute of Brain Science–Shenzhen Fundamental Research Institutions, Shenzhen, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, China
| |
Collapse
|
35
|
Goodman MJ, Li XR, Livschitz J, Huang CC, Bendlin BB, Granadillo ED. Comparing Symmetric Dimethylarginine and Amyloid-β42 as Predictors of Alzheimer's Disease Development. J Alzheimers Dis Rep 2023; 7:1427-1444. [PMID: 38225970 PMCID: PMC10789286 DOI: 10.3233/adr-230054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 11/15/2023] [Indexed: 01/17/2024] Open
Abstract
Background Physicians may soon be able to diagnose Alzheimer's disease (AD) in its early stages using fluid biomarkers like amyloid. However, it is acknowledged that additional biomarkers need to be characterized which would facilitate earlier monitoring of AD pathogenesis. Objective To determine if a potential novel inflammation biomarker for AD, symmetric dimethylarginine, has utility as a baseline serum biomarker for discriminating prodromal AD from cognitively unimpaired controls in comparison to cerebrospinal fluid amyloid-β42 (Aβ42). Methods Data including demographics, magnetic resonance imaging and fluorodeoxyglucose-positron emission tomography scans, Mini-Mental State Examination and Functional Activities Questionnaire scores, and biomarker concentrations were obtained from the Alzheimer's Disease Neuroimaging Initiative for a total of 146 prodromal AD participants and 108 cognitively unimpaired controls. Results Aβ42 (p = 0.65) and symmetric dimethylarginine (p = 0.45) were unable to predict age-matched cognitively unimpaired controls and prodromal AD participants. Aβ42 was negatively associated with regional brain atrophy and hypometabolism as well as cognitive and functional decline in cognitively unimpaired control participants (p < 0.05) that generally decreased in time. There were no significant associations between Aβ42 and symmetric dimethylarginine with imaging or neurocognitive biomarkers in prodromal AD patients. Conclusions Correlations were smaller between Aβ42 and neuropathological biomarkers over time and were absent in prodromal AD participants, suggesting a plateau effect dependent on age and disease stage. Evidence supporting symmetric dimethylarginine as a novel biomarker for AD as a single measurement was not found.
Collapse
Affiliation(s)
| | - Xin Ran Li
- Medical College of Wisconsin, Wauwatosa, WI, USA
| | | | | | | | - Elias D. Granadillo
- Medical College of Wisconsin, Wauwatosa, WI, USA
- University of Wisconsin, Madison, WI, USA
| | | |
Collapse
|
36
|
Milos T, Rojo D, Nedic Erjavec G, Konjevod M, Tudor L, Vuic B, Svob Strac D, Uzun S, Mimica N, Kozumplik O, Barbas C, Zarkovic N, Pivac N, Nikolac Perkovic M. Metabolic profiling of Alzheimer's disease: Untargeted metabolomics analysis of plasma samples. Prog Neuropsychopharmacol Biol Psychiatry 2023; 127:110830. [PMID: 37454721 DOI: 10.1016/j.pnpbp.2023.110830] [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: 04/23/2023] [Revised: 06/07/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Alzheimer's disease (AD) is often not recognized or is diagnosed very late, which significantly reduces the effectiveness of available pharmacological treatments. Metabolomic analyzes have great potential for improving existing knowledge about the pathogenesis and etiology of AD and represent a novel approach towards discovering biomarkers that could be used for diagnosis, prognosis, and therapy monitoring. In this study, we applied the untargeted metabolomic approach to investigate the changes in biochemical pathways related to AD pathology. We used gas chromatography and liquid chromatography coupled to mass spectrometry (GC-MS and LC-MS, respectively) to identify metabolites whose levels have changed in subjects with AD diagnosis (N = 40) compared to healthy controls (N = 40) and individuals with mild cognitive impairment (MCI, N = 40). The GC-MS identified significant differences between groups in levels of metabolites belonging to the classes of benzene and substituted derivatives, carboxylic acids and derivatives, fatty acyls, hydroxy acids and derivatives, keto acids and derivatives, and organooxygen compounds. Most of the compounds identified by the LC-MS were various fatty acyls, glycerolipids and glycerophospholipids. All of these compounds were decreased in AD patients and in subjects with MCI compared to healthy controls. The results of the study indicate disturbed metabolism of lipids and amino acids and an imbalance of metabolites involved in energy metabolism in individuals diagnosed with AD, compared to healthy controls and MCI subjects.
Collapse
Affiliation(s)
- Tina Milos
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia.
| | - David Rojo
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo CEU, CEU Universities Madrid, Spain.
| | | | - Marcela Konjevod
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia.
| | - Lucija Tudor
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia.
| | - Barbara Vuic
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia.
| | | | - Suzana Uzun
- School of Medicine, University of Zagreb, Zagreb, Croatia; Department for Biological Psychiatry and Psychogeriatrics, University Psychiatric Hospital Vrapče, Zagreb, Croatia.
| | - Ninoslav Mimica
- Department for Biological Psychiatry and Psychogeriatrics, University Psychiatric Hospital Vrapče, Zagreb, Croatia.
| | - Oliver Kozumplik
- Department for Biological Psychiatry and Psychogeriatrics, University Psychiatric Hospital Vrapče, Zagreb, Croatia.
| | - Coral Barbas
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo CEU, CEU Universities Madrid, Spain.
| | - Neven Zarkovic
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia.
| | - Nela Pivac
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia; University of Applied Sciences Hrvatsko Zagorje Krapina, Krapina, Croatia.
| | | |
Collapse
|
37
|
Huang SY, Zhang YR, Yang L, Li YZ, Wu BS, Chen SD, Feng JF, Dong Q, Cheng W, Yu JT. Circulating metabolites and risk of incident dementia: A prospective cohort study. J Neurochem 2023; 167:668-679. [PMID: 37908051 DOI: 10.1111/jnc.15997] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/06/2023] [Accepted: 10/10/2023] [Indexed: 11/02/2023]
Abstract
Identifying circulating metabolites associated with dementia, cognition, and brain volume may improve the understanding of dementia pathogenesis and provide novel insights for preventive and therapeutic interventions. This cohort study included a total of 87 885 participants (median follow-up of 9.1 years, 54% female) without dementia at baseline from the UK Biobank. A total of 249 plasma metabolites were measured using nuclear magnetic resonance spectroscopy at baseline. Cox proportional regression was used to examine the associations of each metabolite with incident dementia (cases = 1134), Alzheimer's disease (AD; cases = 488), and vascular dementia (VD; cases = 257) during follow-up. Dementia-associated metabolites were further analyzed for association with cognitive deficits (N = 87 885) and brain volume (N = 7756) using logistic regression and linear regression. We identified 26 metabolites associated with incident dementia, of which 6 were associated with incident AD and 5 were associated with incident VD. These 26 dementia-related metabolites were subfractions of intermediate-density lipoprotein, large low-density lipoprotein (L-LDL), small high-density lipoprotein (S-HDL), very-low-density lipoprotein, fatty acids, ketone bodies, citrate, glucose, and valine. Among them, the cholesterol percentage in L-LDL (L-LDL-C%) was associated with lower risk of AD (HR [95% CI] = 0.92 [0.87-0.97], p = 0.002), higher brain cortical (β = 0.047, p = 3.91 × 10-6 ), and hippocampal (β = 0.043, p = 1.93 × 10-4 ) volume. Cholesteryl ester-to-total lipid ratio in L-LDL (L-LDL-CE%) was associated with lower risk of AD (HR [95% CI] = 0.93 [0.90-0.96], p = 1.48 × 10-4 ), cognitive deficits (odds ratio = 0.98, p = 0.009), and higher hippocampal volume (β = 0.027, p = 0.009). Cholesteryl esters in S-HDL (S-HDL-CE) were associated with lower risk of VD (HR [95% CI] = 0.81 [0.71-0.93], p = 0.002), but not AD. Taken together, circulating levels of L-LDL-CE% and L-LDL-C% were robustly associated with risk of AD and AD phenotypes, but not with VD. S-HDL-CE was associated with lower risk of VD, but not with AD or AD phenotypes. These metabolites may play a role in the advancement of future intervention trials. Additional research is necessary to gain a complete comprehension of the molecular mechanisms behind these associations.
Collapse
Affiliation(s)
- Shu-Yi 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
| | - 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
| | - Yu-Zhu Li
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Bang-Sheng Wu
- 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
| | - Shi-Dong 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
| | - 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
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-Inspired Intelligence, Zhejiang Normal University, Jinhua, 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
| | - 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
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-Inspired Intelligence, Zhejiang Normal University, Jinhua, 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, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
38
|
Shi Y, Katdare KA, Kim H, Rosch JC, Neal EH, Vafaie-Partin S, Bauer JA, Lippmann ES. An arrayed CRISPR knockout screen identifies genetic regulators of GLUT1 expression. Sci Rep 2023; 13:21038. [PMID: 38030680 PMCID: PMC10687026 DOI: 10.1038/s41598-023-48361-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/25/2023] [Indexed: 12/01/2023] Open
Abstract
Glucose, a primary fuel source under homeostatic conditions, is transported into cells by membrane transporters such as glucose transporter 1 (GLUT1). Due to its essential role in maintaining energy homeostasis, dysregulation of GLUT1 expression and function can adversely affect many physiological processes in the body. This has implications in a wide range of disorders such as Alzheimer's disease (AD) and several types of cancers. However, the regulatory pathways that govern GLUT1 expression, which may be altered in these diseases, are poorly characterized. To gain insight into GLUT1 regulation, we performed an arrayed CRISPR knockout screen using Caco-2 cells as a model cell line. Using an automated high content immunostaining approach to quantify GLUT1 expression, we identified more than 300 genes whose removal led to GLUT1 downregulation. Many of these genes were enriched along signaling pathways associated with G-protein coupled receptors, particularly the rhodopsin-like family. Secondary hit validation confirmed that removal of select genes, or modulation of the activity of a corresponding protein, yielded changes in GLUT1 expression. Overall, this work provides a resource and framework for understanding GLUT1 regulation in health and disease.
Collapse
Affiliation(s)
- Yajuan Shi
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Ketaki A Katdare
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Hyosung Kim
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Jonah C Rosch
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Emma H Neal
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Sidney Vafaie-Partin
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Joshua A Bauer
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, USA
- Department of Biochemistry, Vanderbilt University, Nashville, TN, USA
| | - Ethan S Lippmann
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA.
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA.
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA.
- Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, USA.
- Interdisciplinary Materials Science Program, Vanderbilt University, Nashville, TN, USA.
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
39
|
Saeed A, Lopez O, Cohen A, Reis SE. Cardiovascular Disease and Alzheimer's Disease: The Heart-Brain Axis. J Am Heart Assoc 2023; 12:e030780. [PMID: 37929715 PMCID: PMC10727398 DOI: 10.1161/jaha.123.030780] [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] [Indexed: 11/07/2023]
Abstract
Cardiovascular disease (CVD) remains one of the leading causes of morbidity and mortality in aging adults across the United States. Prior studies indicate that the presence of atherosclerosis, the pathogenic basis of CVD, is linked with dementias. Alzheimer's disease (AD) and AD-related dementias are a major public health challenge in the United States. Recent studies indicate that ≈3.7 million Americans ≥65 years of age had clinical AD in 2017, with projected increases to 9.3 million by 2060. Treatment options for AD remain limited. Development of disease-modifying therapies are challenging due, in part, to the long preclinical window of AD. The preclinical incubation period of AD starts in midlife, providing a critical window for identification and optimization of AD risk factors. Studies link AD with CVD risk factors such as hypertension, inflammation, and dyslipidemia. Both AD and CVD are progressive diseases with decades-long development periods. CVD can clinically manifest several years earlier than AD, making CVD and its risk factors a potential predictor of future AD. The current review focuses on the state of literature on molecular and metabolic pathways modulating the heart-brain axis underlying the potential association of midlife CVD risk factors and their effect on AD and related dementias. Further, we explore potential CVD/dementia preventive strategies during the window of opportunity in midlife and the future of research in the field in the multiomics and novel biomarker use era.
Collapse
Affiliation(s)
- Anum Saeed
- University of Pittsburgh School of MedicinePittsburghPAUSA
- Heart and Vascular InstituteUniversity of Pittsburgh Medical CenterPAPittsburghUSA
| | - Oscar Lopez
- University of Pittsburgh School of MedicinePittsburghPAUSA
- Cognitive and Behavioral and Neurology DivisionUniversity of Pittsburgh Medical CenterPAPittsburghUSA
| | - Ann Cohen
- University of Pittsburgh School of MedicinePittsburghPAUSA
- Division of PsychiatryUniversity of Pittsburgh Medical CenterPAPittsburghUSA
| | - Steven E. Reis
- University of Pittsburgh School of MedicinePittsburghPAUSA
- Heart and Vascular InstituteUniversity of Pittsburgh Medical CenterPAPittsburghUSA
| |
Collapse
|
40
|
Sheng C, Du W, Liang Y, Xu P, Ding Q, Chen X, Jia S, Wang X. An integrated neuroimaging-omics approach for the gut-brain communication pathways in Alzheimer's disease. Front Aging Neurosci 2023; 15:1211979. [PMID: 37869373 PMCID: PMC10587434 DOI: 10.3389/fnagi.2023.1211979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 09/22/2023] [Indexed: 10/24/2023] Open
Abstract
A key role of the gut microbiota in the pathogenesis of neurodegenerative diseases, such as Alzheimer's disease (AD), has been identified over the past decades. Increasing clinical and preclinical evidence implicates that there is bidirectional communication between the gut microbiota and the central nervous system (CNS), which is also known as the microbiota-gut-brain axis. Nevertheless, current knowledge on the interplay between gut microbiota and the brain remains largely unclear. One of the primary mediating factors by which the gut microbiota interacts with the host is peripheral metabolites, including blood or gut-derived metabolites. However, mechanistic knowledge about the effect of the microbiome and metabolome signaling on the brain is limited. Neuroimaging techniques, such as multi-modal magnetic resonance imaging (MRI), and fluorodeoxyglucose-positron emission tomography (FDG-PET), have the potential to directly elucidate brain structural and functional changes corresponding with alterations of the gut microbiota and peripheral metabolites in vivo. Employing a combination of gut microbiota, metabolome, and advanced neuroimaging techniques provides a future perspective in illustrating the microbiota-gut-brain pathway and further unveiling potential therapeutic targets for AD treatments.
Collapse
Affiliation(s)
- Can Sheng
- Department of Neurology, The Affiliated Hospital of Jining Medical University, Jining, China
| | - Wenying Du
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | - Yuan Liang
- Department of Neurology, The Affiliated Hospital of Jining Medical University, Jining, China
| | - Peng Xu
- Department of Neurology, The Affiliated Hospital of Jining Medical University, Jining, China
| | - Qingqing Ding
- Department of Neurology, The Affiliated Hospital of Jining Medical University, Jining, China
| | - Xue Chen
- Department of Neurology, The Affiliated Hospital of Jining Medical University, Jining, China
| | - Shulei Jia
- Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Xiaoni Wang
- Department of Neurology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
41
|
Ambeskovic M, Hopkins G, Hoover T, Joseph JT, Montina T, Metz GAS. Metabolomic Signatures of Alzheimer's Disease Indicate Brain Region-Specific Neurodegenerative Progression. Int J Mol Sci 2023; 24:14769. [PMID: 37834217 PMCID: PMC10573054 DOI: 10.3390/ijms241914769] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/25/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
Pathological mechanisms contributing to Alzheimer's disease (AD) are still elusive. Here, we identified the metabolic signatures of AD in human post-mortem brains. Using 1H NMR spectroscopy and an untargeted metabolomics approach, we identified (1) metabolomic profiles of AD and age-matched healthy subjects in post-mortem brain tissue, and (2) region-common and region-unique metabolome alterations and biochemical pathways across eight brain regions revealed that BA9 was the most affected. Phenylalanine and phosphorylcholine were mainly downregulated, suggesting altered neurotransmitter synthesis. N-acetylaspartate and GABA were upregulated in most regions, suggesting higher inhibitory activity in neural circuits. Other region-common metabolic pathways indicated impaired mitochondrial function and energy metabolism, while region-unique pathways indicated oxidative stress and altered immune responses. Importantly, AD caused metabolic changes in brain regions with less well-documented pathological alterations that suggest degenerative progression. The findings provide a new understanding of the biochemical mechanisms of AD and guide biomarker discovery for personalized risk prediction and diagnosis.
Collapse
Affiliation(s)
- Mirela Ambeskovic
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (M.A.); (G.H.); (T.H.)
| | - Giselle Hopkins
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (M.A.); (G.H.); (T.H.)
| | - Tanzi Hoover
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (M.A.); (G.H.); (T.H.)
| | - Jeffrey T. Joseph
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada;
| | - Tony Montina
- Department of Chemistry and Biochemistry, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
- Southern Alberta Genome Sciences Centre, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Gerlinde A. S. Metz
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (M.A.); (G.H.); (T.H.)
- Southern Alberta Genome Sciences Centre, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| |
Collapse
|
42
|
Kalia V, Reyes-Dumeyer D, Dubey S, Nandakumar R, Lee AJ, Lantigua R, Medrano M, Rivera D, Honig LS, Mayeux R, Miller GW, Vardarajan BN. Lysophosphatidylcholines are associated with P-tau181 levels in early stages of Alzheimer's Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.08.24.23294581. [PMID: 37662203 PMCID: PMC10473810 DOI: 10.1101/2023.08.24.23294581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Background We investigated systemic biochemical changes in Alzheimer's disease (AD) by investigating the relationship between circulating plasma metabolites and both clinical and biomarker-assisted diagnosis of AD. Methods We used an untargeted approach with liquid chromatography coupled to high-resolution mass spectrometry to measure exogenous and endogenous small molecule metabolites in plasma from 150 individuals clinically diagnosed with AD and 567 age-matched elderly without dementia of Caribbean Hispanic ancestry. Plasma biomarkers of AD were also measured including P-tau181, Aβ40, Aβ42, total tau, neurofilament light chain (NfL) and glial fibrillary acidic protein (GFAP). Association of individual and co-expressed modules of metabolites were tested with the clinical diagnosis of AD, as well as biologically-defined AD pathological process based on P-tau181 and other biomarker levels. Results Over 4000 metabolomic features were measured with high accuracy. First principal component (PC) of lysophosphatidylcholines (lysoPC) that bind to or interact with docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA) and arachidonic acid (AHA) was associated with decreased risk of AD (OR=0.91 [0.89-0.96], p=2e-04). Restricted to individuals without an APOE ε4 allele (OR=0.89 [0.84-0.94], p= 8.7e-05), the association remained. Among individuals carrying at least one APOE ε4 allele, PC4 of lysoPCs moderately increased risk of AD (OR=1.37 [1.16-1.6], p=1e-04). Essential amino acids including tyrosine metabolism pathways were enriched among metabolites associated with P-tau181 levels and heparan and keratan sulfate degradation pathways were associated with Aβ42/Aβ40 ratio reflecting different pathways enriched in early and middle stages of disease. Conclusions Our findings indicate that unbiased metabolic profiling can identify critical metabolites and pathways associated with β-amyloid and phosphotau pathology. We also observed an APOE ε4 dependent association of lysoPCs with AD and that biologically-based diagnostic criteria may aid in the identification of unique pathogenic mechanisms.
Collapse
Affiliation(s)
- Vrinda Kalia
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University. 722 West 168 Street, New York, NY 10032
| | - Dolly Reyes-Dumeyer
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University. 630 West 168 Street, New York, NY 10032
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University. 630 West 168 Street, New York, NY 10032
| | - Saurabh Dubey
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University. 722 West 168 Street, New York, NY 10032
| | - Renu Nandakumar
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University. 722 West 168 Street, New York, NY 10032
| | - Annie J. Lee
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University. 630 West 168 Street, New York, NY 10032
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University. 630 West 168 Street, New York, NY 10032
| | - Rafael Lantigua
- Department of Medicine, College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital. 630 West 168 Street, New York, NY 10032
| | - Martin Medrano
- School of Medicine, Pontificia Universidad Católica Madre y Maestra, Santiago, Dominican Republic
| | - Diones Rivera
- Department of Neurosurgery, CEDIMAT, Plaza de la Salud, Santo Domingo, Dominican Republic
| | - Lawrence S. Honig
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University. 630 West 168 Street, New York, NY 10032
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University. 630 West 168 Street, New York, NY 10032
- Department of Neurology, College of Physicians and Surgeons, Columbia University and the New York Presbyterian Hospital. 710 West 168 Street, New York, NY 10032
| | - Richard Mayeux
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University. 630 West 168 Street, New York, NY 10032
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University. 630 West 168 Street, New York, NY 10032
- Department of Neurology, College of Physicians and Surgeons, Columbia University and the New York Presbyterian Hospital. 710 West 168 Street, New York, NY 10032
- Department of Epidemiology, Mailman School of Public Health, Columbia University. 722 West 168 Street, New York, NY 10032
| | - Gary W. Miller
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University. 722 West 168 Street, New York, NY 10032
- Department of Epidemiology, Mailman School of Public Health, Columbia University. 722 West 168 Street, New York, NY 10032
| | - Badri N. Vardarajan
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University. 630 West 168 Street, New York, NY 10032
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University. 630 West 168 Street, New York, NY 10032
- Department of Neurology, College of Physicians and Surgeons, Columbia University and the New York Presbyterian Hospital. 710 West 168 Street, New York, NY 10032
| |
Collapse
|
43
|
Xu L, Ge Q, Lu H, Zhuang Y, Geng X, Chen X, Liu X, Sun H, Guo Z, Sun J, Qi F, Niu X, Wang A, Sun W, He J. Cerebrospinal fluid metabolite alterations in patients with different etiologies, diagnoses, and prognoses of disorders of consciousness. Brain Behav 2023; 13:e3070. [PMID: 37421239 PMCID: PMC10454269 DOI: 10.1002/brb3.3070] [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: 12/27/2022] [Revised: 04/29/2023] [Accepted: 03/05/2023] [Indexed: 07/10/2023] Open
Abstract
INTRODUCTION Medical management of disorders of consciousness (DoC) is a growing issue imposing a major burden on families and societies. Recovery rates vary widely among patients with DoC, and recovery predictions strongly influence decisions on medical care. However, the specific mechanisms underlying different etiologies, consciousness levels, and prognoses are still unclear. METHODS We analyzed the comprehensive cerebrospinal fluid (CSF) metabolome through liquid chromatography-mass spectrometry. Metabolomic analyses were used to identify the metabolic differences between patients with different etiologies, diagnoses, and prognoses. RESULTS We found that the CSF levels of multiple acylcarnitines were lower in patients with traumatic DoC, suggesting mitochondrial function preservation in the CNS, which might contribute to the better consciousness outcomes of these patients. Metabolites related to glutamate and GABA metabolism were altered and showed a good ability to distinguish the patients in the minimally conscious state and the vegetative state. Moreover, we identified 8 phospholipids as potential biomarkers to predict the recovery of consciousness. CONCLUSIONS Our findings shed light on the differences in physiological activities underlying DoC with different etiologies and identified some potential biomarkers used for DoC diagnosis and prognosis.
Collapse
Affiliation(s)
- Long Xu
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Department of NeurosurgeryChina National Clinical Research Center for Neurological Diseases (NCRC‐ND)BeijingChina
| | - Qianqian Ge
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Hezhen Lu
- Department of clinical laboratoryChina‐Japan Union Hospital of Jilin UniversityChangchunChina
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic MedicinePeking Union Medical CollegeBeijingChina
| | - Yutong Zhuang
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Xiaoli Geng
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Xueling Chen
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Xiaoyan Liu
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic MedicinePeking Union Medical CollegeBeijingChina
| | - Haidan Sun
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic MedicinePeking Union Medical CollegeBeijingChina
| | - Zhengguang Guo
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic MedicinePeking Union Medical CollegeBeijingChina
| | - Jiameng Sun
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic MedicinePeking Union Medical CollegeBeijingChina
| | - Feng Qi
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic MedicinePeking Union Medical CollegeBeijingChina
| | - Xia Niu
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic MedicinePeking Union Medical CollegeBeijingChina
| | - Aiwei Wang
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic MedicinePeking Union Medical CollegeBeijingChina
| | - Wei Sun
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic MedicinePeking Union Medical CollegeBeijingChina
| | - Jianghong He
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Department of NeurosurgeryChina National Clinical Research Center for Neurological Diseases (NCRC‐ND)BeijingChina
| |
Collapse
|
44
|
Lista S, González-Domínguez R, López-Ortiz S, González-Domínguez Á, Menéndez H, Martín-Hernández J, Lucia A, Emanuele E, Centonze D, Imbimbo BP, Triaca V, Lionetto L, Simmaco M, Cuperlovic-Culf M, Mill J, Li L, Mapstone M, Santos-Lozano A, Nisticò R. Integrative metabolomics science in Alzheimer's disease: Relevance and future perspectives. Ageing Res Rev 2023; 89:101987. [PMID: 37343679 DOI: 10.1016/j.arr.2023.101987] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/12/2023] [Accepted: 06/15/2023] [Indexed: 06/23/2023]
Abstract
Alzheimer's disease (AD) is determined by various pathophysiological mechanisms starting 10-25 years before the onset of clinical symptoms. As multiple functionally interconnected molecular/cellular pathways appear disrupted in AD, the exploitation of high-throughput unbiased omics sciences is critical to elucidating the precise pathogenesis of AD. Among different omics, metabolomics is a fast-growing discipline allowing for the simultaneous detection and quantification of hundreds/thousands of perturbed metabolites in tissues or biofluids, reproducing the fluctuations of multiple networks affected by a disease. Here, we seek to critically depict the main metabolomics methodologies with the aim of identifying new potential AD biomarkers and further elucidating AD pathophysiological mechanisms. From a systems biology perspective, as metabolic alterations can occur before the development of clinical signs, metabolomics - coupled with existing accessible biomarkers used for AD screening and diagnosis - can support early disease diagnosis and help develop individualized treatment plans. Presently, the majority of metabolomic analyses emphasized that lipid metabolism is the most consistently altered pathway in AD pathogenesis. The possibility that metabolomics may reveal crucial steps in AD pathogenesis is undermined by the difficulty in discriminating between the causal or epiphenomenal or compensatory nature of metabolic findings.
Collapse
Affiliation(s)
- Simone Lista
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid, Spain.
| | - Raúl González-Domínguez
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, Universidad de Cádiz, Cádiz, Spain
| | - Susana López-Ortiz
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid, Spain
| | - Álvaro González-Domínguez
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, Universidad de Cádiz, Cádiz, Spain
| | - Héctor Menéndez
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid, Spain
| | - Juan Martín-Hernández
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid, Spain
| | - Alejandro Lucia
- Research Institute of the Hospital 12 de Octubre ('imas12'), Madrid, Spain; Faculty of Sport Sciences, European University of Madrid, Villaviciosa de Odón, Madrid, Spain; CIBER of Frailty and Healthy Ageing (CIBERFES), Madrid, Spain
| | | | - Diego Centonze
- Department of Systems Medicine, Tor Vergata University, Rome, Italy; Unit of Neurology, IRCCS Neuromed, Pozzilli, IS, Italy
| | - Bruno P Imbimbo
- Department of Research and Development, Chiesi Farmaceutici, Parma, Italy
| | - Viviana Triaca
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Rome, Italy
| | - Luana Lionetto
- Clinical Biochemistry, Mass Spectrometry Section, Sant'Andrea University Hospital, Rome, Italy; Department of Neuroscience, Mental Health and Sensory Organs, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Maurizio Simmaco
- Clinical Biochemistry, Mass Spectrometry Section, Sant'Andrea University Hospital, Rome, Italy; Department of Neuroscience, Mental Health and Sensory Organs, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Miroslava Cuperlovic-Culf
- Digital Technologies Research Center, National Research Council, Ottawa, Canada; Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Jericha Mill
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Lingjun Li
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA; School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Mark Mapstone
- Department of Neurology, Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
| | - Alejandro Santos-Lozano
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid, Spain; Research Institute of the Hospital 12 de Octubre ('imas12'), Madrid, Spain
| | - Robert Nisticò
- School of Pharmacy, University of Rome "Tor Vergata", Rome, Italy; Laboratory of Pharmacology of Synaptic Plasticity, EBRI Rita Levi-Montalcini Foundation, Rome, Italy
| |
Collapse
|
45
|
Kainuma M, Kawakatsu S, Kim JD, Ouma S, Iritani O, Yamashita KI, Ohara T, Hirano S, Suda S, Hamano T, Hieda S, Yasui M, Yoshiiwa A, Shiota S, Hironishi M, Wada-Isoe K, Sasabayashi D, Yamasaki S, Murata M, Funakoshi K, Hayashi K, Shirafuji N, Sasaki H, Kajimoto Y, Mori Y, Suzuki M, Ito H, Ono K, Tsuboi Y. Metabolic changes in the plasma of mild Alzheimer's disease patients treated with Hachimijiogan. Front Pharmacol 2023; 14:1203349. [PMID: 37377927 PMCID: PMC10292017 DOI: 10.3389/fphar.2023.1203349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Background: Alzheimer's disease (AD), the most prevalent form of dementia, is a debilitating, progressive neurodegeneration. Amino acids play a wide variety of physiological and pathophysiological roles in the nervous system, and their levels and disorders related to their synthesis have been related to cognitive impairment, the core feature of AD. Our previous multicenter trial showed that hachimijiogan (HJG), a traditional Japanese herbal medicine (Kampo), has an adjuvant effect for Acetylcholine estelase inhibitors (AChEIs) and that it delays the deterioration of the cognitive dysfunction of female patients with mild AD. However, there are aspects of the molecular mechanism(s) by which HJG improves cognitive dysfunction that remain unclear. Objectives: To elucidate through metabolomic analysis the mechanism(s) of HJG for mild AD based on changes in plasma metabolites. Methods: Sixty-seven patients with mild AD were randomly assigned to either an HJG group taking HJG extract 7.5 g/day in addition to AChEI or to a control group treated only with AChEI (HJG:33, Control:34). Blood samples were collected before, 3 months, and 6 months after the first drug administration. Comprehensive metabolomic analyses of plasma samples were done by optimized LC-MS/MS and GC-MS/MS methods. The web-based software MetaboAnalyst 5.0 was used for partial least square-discriminant analysis (PLS-DA) to visualize and compare the dynamics of changes in the concentrations of the identified metabolites. Results: The VIP (Variable Importance in Projection) score of the PLS-DA analysis of female participants revealed a significantly higher increase in plasma metabolite levels after HJG administration for 6 months than was seen in the control group. In univariate analysis, the aspartic acid level of female participants showed a significantly higher increase from baseline after HJG administration for 6 months when compared with the control group. Conclusion: Aspartic acid was a major contributor to the difference between the female HJG and control group participants of this study. Several metabolites were shown to be related to the mechanism of HJG effectiveness for mild AD.
Collapse
Affiliation(s)
- Mosaburo Kainuma
- Department of Japanese Oriental Medicine Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Shinobu Kawakatsu
- Aizu Medical Center, Department of Neuropsychiatry, Fukushima Medical University, Aizuwakamatsu, Japan
| | - Jun-Dal Kim
- Department of Research and Development, Division of Complex Biosystem Research (CBR), Institute of National Medicine (INM), University of Toyama, Toyama, Japan
| | - Shinji Ouma
- Department of Neurology, School of Medicine, Fukuoka University, Fukuoka, Japan
| | - Osamu Iritani
- Department of Geriatric Medicine, Kanazawa Medical University, Ishikawa, Japan
| | - Ken-Ichiro Yamashita
- Translational Neuroscience Center, Graduate School of Medicine, International University of Health and Welfare, Tochigi, Japan
| | - Tomoyuki Ohara
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shigeki Hirano
- Department of Neurology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Shiro Suda
- Department of Psychiatry, Jichi Medical University, Tochigi, Japan
| | - Tadanori Hamano
- Second Department of Internal Medicine, Division of Neurology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Sotaro Hieda
- Department of Medicine, Division of Neurology, Showa University School of Medicine, Tokyo, Japan
| | - Masaaki Yasui
- Department of Neurology, Wakayama Medical University, Wakayama, Japan
| | - Aoi Yoshiiwa
- Department of General Medicine, Oita University Faculty of Medicine, Oita, Japan
| | - Seiji Shiota
- Department of General Medicine, Oita University Faculty of Medicine, Oita, Japan
| | - Masaya Hironishi
- Department of Internal Medicine, Wakayama Medical University Kihoku Hospital, Wakayama, Japan
| | - Kenji Wada-Isoe
- Department of Dementia Medicine, Kawasaki Medical School, Okayama, Japan
| | - Daiki Sasabayashi
- Department of Neuropsychiatry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Sho Yamasaki
- Department of General Internal Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Masayuki Murata
- Department of General Internal Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Kouta Funakoshi
- Department of Clinical Research Promotion, Kyushu University Hospital, Fukuoka, Japan
| | - Kouji Hayashi
- Department of Rehabilitation, Fukui Health Science University, Fukui, Japan
| | - Norimichi Shirafuji
- Second Department of Internal Medicine, Division of Neurology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Hirohito Sasaki
- Second Department of Internal Medicine, Division of Neurology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Yoshinori Kajimoto
- Department of Internal Medicine, Wakayama Medical University Kihoku Hospital, Wakayama, Japan
| | - Yukiko Mori
- Department of Medicine, Division of Neurology, Showa University School of Medicine, Tokyo, Japan
| | - Michio Suzuki
- Department of Neuropsychiatry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Hidefumi Ito
- Department of Neurology, Wakayama Medical University, Wakayama, Japan
| | - Kenjiro Ono
- Department of Neurology, Kanazawa University Graduate School of Medical Sciences, Ishikawa, Japan
| | - Yoshio Tsuboi
- Department of Neurology, School of Medicine, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
46
|
Chang CW, Hsu JY, Hsiao PZ, Chen YC, Liao PC. Identifying Hair Biomarker Candidates for Alzheimer's Disease Using Three High Resolution Mass Spectrometry-Based Untargeted Metabolomics Strategies. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2023; 34:550-561. [PMID: 36973238 DOI: 10.1021/jasms.2c00294] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
High-resolution mass spectrometry (HRMS)-based untargeted metabolomics strategies have emerged as an effective tool for discovering biomarkers of Alzheimer's disease (AD). There are various HRMS-based untargeted metabolomics strategies for biomarker discovery, including the data-dependent acquisition (DDA) method, the combination of full scan and target MS/MS, and the all ion fragmentation (AIF) method. Hair has emerged as a potential biospecimen for biomarker discovery in clinical research since it might reflect the circulating metabolic profiles over several months, while the analytical performances of the different data acquisition methods for hair biomarker discovery have been rarely investigated. Here, the analytical performances of three data acquisition methods in HRMS-based untargeted metabolomics for hair biomarker discovery were evaluated. The human hair samples from AD patients (N = 23) and cognitively normal individuals (N = 23) were used as an example. The most significant number of discriminatory features was acquired using the full scan (407), which is approximately 10-fold higher than that using the DDA strategy (41) and 11% higher than that using the AIF strategy (366). Only 66% of discriminatory chemicals discovered in the DDA strategy were discriminatory features in the full scan dataset. Moreover, compared to the deconvoluted MS/MS spectra with coeluted and background ions from the AIF method, the MS/MS spectrum obtained from the targeted MS/MS approach is cleaner and purer. Therefore, an untargeted metabolomics strategy combining the full scan with the targeted MS/MS method could obtain most discriminatory features along with a high quality MS/MS spectrum for discovering the AD biomarkers.
Collapse
Affiliation(s)
- Chih-Wei Chang
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan 704, Taiwan
| | - Jen-Yi Hsu
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan 704, Taiwan
| | - Ping-Zu Hsiao
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan 704, Taiwan
| | - Yuan-Chih Chen
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan 704, Taiwan
| | - Pao-Chi Liao
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan 704, Taiwan
- Department of Food Safety/Hygiene and Risk Management, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan 704, Taiwan
| |
Collapse
|
47
|
Yang M, Matan-Lithwick S, Wang Y, De Jager PL, Bennett DA, Felsky D. Multi-omic integration via similarity network fusion to detect molecular subtypes of ageing. Brain Commun 2023; 5:fcad110. [PMID: 37082508 PMCID: PMC10110975 DOI: 10.1093/braincomms/fcad110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/17/2023] [Accepted: 04/03/2023] [Indexed: 04/07/2023] Open
Abstract
Molecular subtyping of brain tissue provides insights into the heterogeneity of common neurodegenerative conditions, such as Alzheimer's disease. However, existing subtyping studies have mostly focused on single data modalities and only those individuals with severe cognitive impairment. To address these gaps, we applied similarity network fusion, a method capable of integrating multiple high-dimensional multi-omic data modalities simultaneously, to an elderly sample spanning the full spectrum of cognitive ageing trajectories. We analyzed human frontal cortex brain samples characterized by five omic modalities: bulk RNA sequencing (18 629 genes), DNA methylation (53 932 CpG sites), histone acetylation (26 384 peaks), proteomics (7737 proteins) and metabolomics (654 metabolites). Similarity network fusion followed by spectral clustering was used for subtype detection, and subtype numbers were determined by Eigen-gap and rotation cost statistics. Normalized mutual information determined the relative contribution of each modality to the fused network. Subtypes were characterized by associations with 13 age-related neuropathologies and cognitive decline. Fusion of all five data modalities (n = 111) yielded two subtypes (n S1 = 53, n S2 = 58), which were nominally associated with diffuse amyloid plaques; however, this effect was not significant after correction for multiple testing. Histone acetylation (normalized mutual information = 0.38), DNA methylation (normalized mutual information = 0.18) and RNA abundance (normalized mutual information = 0.15) contributed most strongly to this network. Secondary analysis integrating only these three modalities in a larger subsample (n = 513) indicated support for both three- and five-subtype solutions, which had significant overlap, but showed varying degrees of internal stability and external validity. One subtype showed marked cognitive decline, which remained significant even after correcting for tests across both three- and five-subtype solutions (p Bonf = 5.9 × 10-3). Comparison to single-modality subtypes demonstrated that the three-modal subtypes were able to uniquely capture cognitive variability. Comprehensive sensitivity analyses explored influences of sample size and cluster number parameters. We identified highly integrative molecular subtypes of ageing derived from multiple high dimensional, multi-omic data modalities simultaneously. Fusing RNA abundance, DNA methylation, and histone acetylation measures generated subtypes that were associated with cognitive decline. This work highlights the potential value and challenges of multi-omic integration in unsupervised subtyping of post-mortem brain.
Collapse
Affiliation(s)
- Mu Yang
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
- The Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
| | - Stuart Matan-Lithwick
- The Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
| | - Yanling Wang
- Rush Alzheimer’s Disease Center, Rush University, Chicago, IL 60612, USA
| | - Philip L De Jager
- The Center for Translational and Computational Neuroimmunology, Columbia University Medical Center, New York, NY 10033, USA
| | - David A Bennett
- Rush Alzheimer’s Disease Center, Rush University, Chicago, IL 60612, USA
| | - Daniel Felsky
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
- The Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
48
|
Zou Y, Wang Q, Cheng X. Causal Relationship Between Basal Metabolic Rate and Alzheimer's Disease: A Bidirectional Two-sample Mendelian Randomization Study. Neurol Ther 2023; 12:763-776. [PMID: 36894827 DOI: 10.1007/s40120-023-00458-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 02/23/2023] [Indexed: 03/11/2023] Open
Abstract
INTRODUCTION Objective observational studies have shown that basal metabolic rate (BMR) decreases in patients with Alzheimer's disease (AD), but the causal relationship between BMR and AD has not been established. We determined the causal relationship between BMR and AD by two-way Mendelian randomization (MR) and investigated the impact of factors associated with BMR on AD. METHODS We obtained BMR (n = 454,874) and AD from a large genome-wide association study (GWAS) database (21,982 patients with AD, 41,944 controls). The causal relationship between AD and BMR was investigated using two-way MR. Additionally, we identified the causal relationship between AD and factors related with BMR, hyperthyroidism (hy/thy) and type 2 diabetes (T2D), height and weight. RESULTS BMR had a causal relationship with AD [451 single nucleotide polymorphisms (SNPs), odds ratio (OR) 0.749, 95% confidence intervals (CIs) 0.663-0.858, P = 2.40E-03]. There was no causal relationship between hy/thy or T2D and AD (P > 0.05). The bidirectional MR showed that there was also a causal relationship between AD and BMR (OR 0.992, Cls 0.987-0.997, NSNPs18, P = 1.50E-03). BMR, height and weight have a protective effect on AD. Based on MVMR analysis, we found that genetically determined height and weight may be adjusted by BMR to have a causal effect on AD, not height and weight themselves. CONCLUSION Our study showed that higher BMR reduced the risk of AD, and patients with AD had a lower BMR. Because of a positive correlation with BMR, height and weight may have a protective effect on AD. The two metabolism-related diseases, hy/thy and T2D, had no causal relationship with AD.
Collapse
Affiliation(s)
- Yuexiao Zou
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Qingxian Wang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Xiaorui Cheng
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
49
|
Lin CH, Lin YN, Lane HY, Chen CJ. The identification of a potential plasma metabolite marker for Alzheimer’s disease by LC-MS untargeted metabolomics. J Chromatogr B Analyt Technol Biomed Life Sci 2023; 1222:123686. [PMID: 37068461 DOI: 10.1016/j.jchromb.2023.123686] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 03/17/2023] [Accepted: 03/19/2023] [Indexed: 04/03/2023]
Abstract
BACKGROUND AND AIMS Alzheimer's disease (AD), the most common type of dementia, is hard to recognize early, resulting in delayed treatment and poor outcome. At present, there is neither reliable, non-invasive methods to diagnose it accurately and nor effective drugs to recover it. Discovery and quantification of novel metabolite markers in plasma of AD patients and investigation of the correlation between the markers and AD assessment scores. MATERIALS AND METHODS Untargeted liquid chromatography-mass spectrometry (LC-MS)-based metabolomics with LC-quadrupole- time-of-flight (Q-TOF) was performed in plasma samples of age-matched AD patients and healthy controls. The potential markers were further quantified with targeted multiple reaction monitoring (MRM) approach. RESULTS Among the candidates, progesterone, and 3-indoleacetic acid (3-IAA) were successfully identified and then validated in 50 plasma samples from 25 AD patients and 25 matched normal controls with MRM approach. As a result, 3-IAA was significantly altered in AD patients and correlated with some AD assessment scores. CONCLUSION By using untargeted LC-MS metabolomic and LC-MRM approaches to analyze plasma metabolites of AD patients and normal subjects, 3-IAA was discovered and quantified to be significantly altered in AD patients and correlated with several AD assessment scores.
Collapse
Affiliation(s)
- Chieh-Hsin Lin
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan; School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Ning Lin
- Proteomics Core Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Hsien-Yuan Lane
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan; Department of Psychiatry and Brain Disease Research Center, China Medical University Hospital, Taichung, Taiwan; Department of Psychology, College of Medical and Health Sciences, Asia University, Taichung, Taiwan.
| | - Chao-Jung Chen
- Proteomics Core Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan; Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan.
| |
Collapse
|
50
|
Zhao F, Li P, Deng F, Wang J, Cheng Y, Pei K, Wang Y, Wang Y. Integrated Lipidomics and Network Pharmacology Reveal Mechanism of Memory Impairment Improvement by Yuanzhi San. Chem Biodivers 2023; 20:e202200920. [PMID: 36683009 DOI: 10.1002/cbdv.202200920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 01/05/2023] [Accepted: 01/20/2023] [Indexed: 01/24/2023]
Abstract
Memory impairment (MI) is caused by a variety of causes, endangering human health. Yuanzhi San (YZS) is a common prescription used for the treatment of MI, but its mechanism of action needs further exploration. The purpose of this study was to investigate this mechanism through lipidomics and network pharmacology. Sprague Dawley (SD) rats were divided randomly into the normal, model, and YZS groups. The rats were gavaged with aluminum chloride (200 mg/kg) and intraperitoneally injected with D-galactose (400 mg/kg) every day for 60 days, except for the normal group. From the 30th day, YZS (13.34 g/kg) was gavaged once a day to the rats in the YZS group. Post-YZS treatment, ultra-high-performance liquid chromatography-mass spectrometry (UHPLC/MS) analysis was implemented to conduct a lipidomics study in the hippocampus of rats with memory impairment induced by aluminum chloride and D-galactose. Eight differential metabolites were identified between the normal group and the model group, whereas between the model group and the YZS group, 20 differential metabolites were established. Metabolic pathway analysis was performed on the aforementioned lipid metabolites, all of which were involved in sphingolipid and glycerophospholipid metabolism. Furthermore, serum pharmacochemistry analysis of YZS was carried out at the early stage of our research, which discovered 62 YZS prototype components. The results of the network pharmacology analysis showed that they were related to 1030 genes, and 451 disease genes were related to MI. There were 73 intersections between the YZS and MI targets. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed that these targets were closely related to the sphingolipid metabolic, calcium signaling, and other pathways. The integrated approach of lipidomics and network pharmacology was then focused on four major targets, including PHK2, GBA, SPTLC1, and AChE, as well as their essential metabolites (glucosylceramide, N-acylsphingosine, phosphatidylserine, phosphatidylcholine, and phosphatidylcholine) and pathways (sphingolipid, glycerophospholipid, and arachidonic acid metabolism). The significant affinity of the primary target for YZS was confirmed by molecular docking. The obtained results revealed that the combination of lipidomics and network pharmacology could be used to determine the effect of YZS on the MI biological network and metabolic state, and evaluate the drug efficacy of YZS and its related mechanisms of action.
Collapse
Affiliation(s)
- Fuxia Zhao
- Department of Pharmacy, Shanxi University of Traditional Chinese Medicine, Jinzhong, 030619, China
| | - Pei Li
- Department of Pharmacy, Shanxi University of Traditional Chinese Medicine, Jinzhong, 030619, China
| | - Fanying Deng
- Department of Pharmacy, Shanxi University of Traditional Chinese Medicine, Jinzhong, 030619, China
| | - Jiamin Wang
- Department of Pharmacy, Shanxi University of Traditional Chinese Medicine, Jinzhong, 030619, China
| | - Yangang Cheng
- Department of Pharmacy, Shanxi University of Traditional Chinese Medicine, Jinzhong, 030619, China
| | - Ke Pei
- Department of Pharmacy, Shanxi University of Traditional Chinese Medicine, Jinzhong, 030619, China
| | - Yingli Wang
- Department of Pharmacy, Shanxi University of Traditional Chinese Medicine, Jinzhong, 030619, China
| | - Yan Wang
- Department of Pharmacy, Shanxi University of Traditional Chinese Medicine, Jinzhong, 030619, China
| |
Collapse
|