1
|
Franco R, Garrigós C, Lillo J, Rivas-Santisteban R. The Potential of Metabolomics to Find Proper Biomarkers for Addressing the Neuroprotective Efficacy of Drugs Aimed at Delaying Parkinson's and Alzheimer's Disease Progression. Cells 2024; 13:1288. [PMID: 39120318 PMCID: PMC11311351 DOI: 10.3390/cells13151288] [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: 06/18/2024] [Revised: 07/25/2024] [Accepted: 07/28/2024] [Indexed: 08/10/2024] Open
Abstract
The first objective is to highlight the lack of tools to measure whether a given intervention affords neuroprotection in patients with Alzheimer's or Parkinson's diseases. A second aim is to present the primary outcome measures used in clinical trials in cohorts of patients with neurodegenerative diseases. The final aim is to discuss whether metabolomics using body fluids may lead to the discovery of biomarkers of neuroprotection. Information on the primary outcome measures in clinical trials related to Alzheimer's and Parkinson's disease registered since 2018 was collected. We analysed the type of measures selected to assess efficacy, not in terms of neuroprotection since, as stated in the aims, there is not yet any marker of neuroprotection. Proteomic approaches using plasma or CSF have been proposed. PET could estimate the extent of lesions, but disease progression does not necessarily correlate with a change in tracer uptake. We propose some alternatives based on considering the metabolome. A new opportunity opens with metabolomics because there have been impressive technological advances that allow the detection, among others, of metabolites related to mitochondrial function and mitochondrial structure in serum and/or cerebrospinal fluid; some of the differentially concentrated metabolites can become reliable biomarkers of neuroprotection.
Collapse
Affiliation(s)
- Rafael Franco
- Molecular Neurobiology Laboratory, Departament de Bioquimica i Biomedicina Molecular, Universitat de Barcelona, Diagonal 643, 08028 Barcelona, Spain; (C.G.); (J.L.)
- Network Center Neurodegenerative Diseases, CiberNed, Spanish National Health Center Carlos iii, Monforte de Lemos 3, 28029 Madrid, Spain
- School of Chemistry, Universitat de Barcelona, Diagonal 645, 08028 Barcelona, Spain
| | - Claudia Garrigós
- Molecular Neurobiology Laboratory, Departament de Bioquimica i Biomedicina Molecular, Universitat de Barcelona, Diagonal 643, 08028 Barcelona, Spain; (C.G.); (J.L.)
| | - Jaume Lillo
- Molecular Neurobiology Laboratory, Departament de Bioquimica i Biomedicina Molecular, Universitat de Barcelona, Diagonal 643, 08028 Barcelona, Spain; (C.G.); (J.L.)
- Network Center Neurodegenerative Diseases, CiberNed, Spanish National Health Center Carlos iii, Monforte de Lemos 3, 28029 Madrid, Spain
| | - Rafael Rivas-Santisteban
- Network Center Neurodegenerative Diseases, CiberNed, Spanish National Health Center Carlos iii, Monforte de Lemos 3, 28029 Madrid, Spain
- Laboratory of Computational Medicine, Biostatistics Unit, Faculty of Medicine, Autonomous University of Barcelona, Campus Bellaterra, 08193 Barcelona, Spain
| |
Collapse
|
2
|
Dunham SJB, Avelar‐Barragan J, Rothman JA, Adams ED, Faraci G, Forner S, Kawauchi S, Tenner AJ, Green KN, LaFerla FM, MacGregor GR, Mapstone M, Whiteson KL. Sex-specific associations between AD genotype and the microbiome of human amyloid beta knock-in (hAβ-KI) mice. Alzheimers Dement 2024; 20:4935-4950. [PMID: 38572865 PMCID: PMC11247698 DOI: 10.1002/alz.13794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 04/05/2024]
Abstract
INTRODUCTION Emerging evidence links changes in the gut microbiome to late-onset Alzheimer's disease (LOAD), necessitating examination of AD mouse models with consideration of the microbiome. METHODS We used shotgun metagenomics and untargeted metabolomics to study the human amyloid beta knock-in (hAβ-KI) murine model for LOAD compared to both wild-type (WT) mice and a model for early-onset AD (3xTg-AD). RESULTS Eighteen-month female (but not male) hAβ-KI microbiomes were distinct from WT microbiomes, with AD genotype accounting for 18% of the variance by permutational multivariate analysis of variance (PERMANOVA). Metabolomic diversity differences were observed in females, however no individual metabolites were differentially abundant. hAβ-KI mice microbiomes were distinguishable from 3xTg-AD animals (81% accuracy by random forest modeling), with separation primarily driven by Romboutsia ilealis and Turicibacter species. Microbiomes were highly cage specific, with cage assignment accounting for more than 40% of the PERMANOVA variance between the groups. DISCUSSION These findings highlight a sex-dependent variation in the microbiomes of hAβ-KI mice and underscore the importance of considering the microbiome when designing studies that use murine models for AD. HIGHLIGHTS Microbial diversity and the abundance of several species differed in human amyloid beta knock-in (hAβ-KI) females but not males. Correlations to Alzheimer's disease (AD) genotype were stronger for the microbiome than the metabolome. Microbiomes from hAβ-KI mice were distinct from 3xTg-AD mice. Cage effects accounted for most of the variance in the microbiome and metabolome.
Collapse
Affiliation(s)
- Sage J. B. Dunham
- Department of Molecular Biology & BiochemistryUniversity of California IrvineIrvineCaliforniaUSA
| | - Julio Avelar‐Barragan
- Department of Molecular Biology & BiochemistryUniversity of California IrvineIrvineCaliforniaUSA
| | - Jason A. Rothman
- Department of Molecular Biology & BiochemistryUniversity of California IrvineIrvineCaliforniaUSA
| | - Eric D. Adams
- Department of Molecular Biology & BiochemistryUniversity of California IrvineIrvineCaliforniaUSA
| | - Gina Faraci
- Department of Molecular Biology & BiochemistryUniversity of California IrvineIrvineCaliforniaUSA
| | - Stefania Forner
- Institute for Memory Impairments and Neurological Disorders (UCI MIND)University of California IrvineIrvineCaliforniaUSA
| | - Shimako Kawauchi
- Department of Anatomy and NeurobiologyDevelopmental Biology CenterUniversity of California IrvineCollege of MedicineIrvineCaliforniaUSA
| | - Andrea J. Tenner
- Department of Molecular Biology & BiochemistryDepartment of Neurobiology and BehaviorDepartment of Pathology and Laboratory MedicineSchool of MedicineInstitute for Memory Impairments and Neurological Disorders (UCI MIND)University of California IrvineIrvineCaliforniaUSA
| | - Kim N. Green
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), Department of Neurobiology and BehaviorSchool of Biological SciencesCenter for Neural Circuit MappingUniversity of California IrvineIrvineCaliforniaUSA
| | - Frank M. LaFerla
- Institute for Memory Impairments and Neurological Disorders (UCI MIND)Department of Neurobiology and BehaviorUniversity of California IrvineIrvineCaliforniaUSA
| | - Grant R. MacGregor
- Department of Developmental and Cell BiologyUniversity of California IrvineIrvineCaliforniaUSA
| | - Mark Mapstone
- Department of NeurologyUniversity of California IrvineIrvineCaliforniaUSA
| | - Katrine L. Whiteson
- Department of Molecular Biology & BiochemistryUniversity of California IrvineIrvineCaliforniaUSA
| |
Collapse
|
3
|
Wardhani K, Yazzie S, Edeh O, Grimes M, Dixson C, Jacquez Q, Zychowski KE. Neuroinflammation is dependent on sex and ovarian hormone presence following acute woodsmoke exposure. Sci Rep 2024; 14:12995. [PMID: 38844478 PMCID: PMC11156661 DOI: 10.1038/s41598-024-63562-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/30/2024] [Indexed: 06/09/2024] Open
Abstract
Woodsmoke (WS) exposure is associated with significant health-related sequelae. Different populations can potentially exhibit varying susceptibility, based on endocrine phenotypes, to WS and investigating neurological impacts following inhaled WS is a growing area of research. In this study, a whole-body inhalation chamber was used to expose both male and female C57BL/6 mice (n = 8 per group) to either control filtered air (FA) or acute WS (0.861 ± 0.210 mg/m3) for 4 h/d for 2 days. Neuroinflammatory and lipid-based biological markers were then assessed. In a second set of studies, female mice were divided into two groups: one group was ovariectomized (OVX) to simulate an ovarian hormone-deficient state (surgical menopause), and the other underwent Sham surgery as controls, to mechanistically assess the impact of ovarian hormone presence on neuroinflammation following FA and acute WS exposure to simulate an acute wildfire episode. There was a statistically significant impact of sex (P ≤ 0.05) and statistically significant interactions between sex and treatment in IL-1β, CXCL-1, TGF-β, and IL-6 brain relative gene expression. Hippocampal and cortex genes also exhibited significant changes in acute WS-exposed Sham and OVX mice, particularly in TGF-β (hippocampus) and CCL-2 and CXCL-1 (cortex). Cortex GFAP optical density (OD) showed a notable elevation in male mice exposed to acute WS, compared to the control FA. Sham and OVX females demonstrated differential GFAP expression, depending on brain region. Overall, targeted lipidomics in phosphatidylcholine (PC) and phosphatidylethanolamine (PE) serum and brain lipids demonstrated more significant changes between control FA and acute WS exposure in female mice, compared to males. In summary, male and female mice show distinct neuroinflammatory markers in response to acute WS exposure. Furthermore, ovarian hormone deficiency may impact the neuroinflammatory response following an acute WS event.
Collapse
Affiliation(s)
- Kartika Wardhani
- College of Nursing, University of New Mexico-Health Sciences Center, Albuquerque, NM, 87131, USA
- Biochemistry and Biotechnology (B-TEK) Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, 87545, USA
| | - Sydnee Yazzie
- College of Nursing, University of New Mexico-Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Onamma Edeh
- College of Nursing, University of New Mexico-Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Martha Grimes
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico-Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Connor Dixson
- College of Nursing, University of New Mexico-Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Quiteria Jacquez
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico-Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Katherine E Zychowski
- College of Nursing, University of New Mexico-Health Sciences Center, Albuquerque, NM, 87131, USA.
| |
Collapse
|
4
|
Su H, Masters CL, Bush AI, Barnham KJ, Reid GE, Vella LJ. Exploring the significance of lipids in Alzheimer's disease and the potential of extracellular vesicles. Proteomics 2024; 24:e2300063. [PMID: 37654087 DOI: 10.1002/pmic.202300063] [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: 05/12/2023] [Revised: 08/07/2023] [Accepted: 08/14/2023] [Indexed: 09/02/2023]
Abstract
Lipids play a significant role in maintaining central nervous system (CNS) structure and function, and the dysregulation of lipid metabolism is known to occur in many neurological disorders, including Alzheimer's disease. Here we review what is currently known about lipid dyshomeostasis in Alzheimer's disease. We propose that small extracellular vesicle (sEV) lipids may provide insight into the pathophysiology and progression of Alzheimer's disease. This stems from the recognition that sEV likely contributes to disease pathogenesis, but also an understanding that sEV can serve as a source of potential biomarkers. While the protein and RNA content of sEV in the CNS diseases have been studied extensively, our understanding of the lipidome of sEV in the CNS is still in its infancy.
Collapse
Affiliation(s)
- Huaqi Su
- The Florey, The University of Melbourne, Parkville, Victoria, Australia
- School of Chemistry, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Colin L Masters
- The Florey, The University of Melbourne, Parkville, Victoria, Australia
| | - Ashley I Bush
- The Florey, The University of Melbourne, Parkville, Victoria, Australia
| | - Kevin J Barnham
- The Florey, The University of Melbourne, Parkville, Victoria, Australia
| | - Gavin E Reid
- School of Chemistry, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria, Australia
| | - Laura J Vella
- The Florey, The University of Melbourne, Parkville, Victoria, Australia
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
5
|
Peña-Bautista C, Álvarez-Sánchez L, García-Lluch G, Raga L, Quevedo P, Peretó M, Balaguer A, Baquero M, Cháfer-Pericás C. Relationship between Plasma Lipid Profile and Cognitive Status in Early Alzheimer Disease. Int J Mol Sci 2024; 25:5317. [PMID: 38791355 PMCID: PMC11120743 DOI: 10.3390/ijms25105317] [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: 04/11/2024] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Alzheimer disease (AD) is a heterogeneous and complex disease in which different pathophysiological mechanisms are involved. This heterogenicity can be reflected in different atrophy patterns or clinical manifestations. Regarding biochemical pathways involved in early AD, lipid metabolism plays an important role; therefore, lipid levels have been evaluated as potential AD diagnosis biomarkers, and their levels could be related to different AD clinical manifestations. Therefore, the aim of this work is to study AD lipid profiles from early AD patients and evaluate their clinical significance. For this purpose, untargeted plasma lipidomic analysis was carried out in early AD patients (n = 31) diagnosed with cerebrospinal fluid (CSF) biomarkers. Cluster analysis was carried out to define early AD subgroups according to the lipid levels. Then, the clinical significance of each lipid profile subgroup was studied, analyzing differences for other variables (cognitive status, CSF biomarkers, medication, comorbidities, age, and gender). The cluster analysis revealed two different groups of AD patients. Cluster 1 showed higher levels of plasma lipids and better cognitive status than Cluster 2. However, no differences were found for the other variables (age, gender, medication, comorbidities, cholesterol, and triglycerides levels) between both groups. Plasma lipid levels could differentiate two early AD subgroups, which showed different cognitive statuses. However, further research with a large cohort and longitudinal study evaluating the clinical evolution of these patients is required. In general, it would involve a relevant advance in the knowledge of AD pathological mechanisms, potential treatments, and precision medicine.
Collapse
Affiliation(s)
- Carmen Peña-Bautista
- Alzheimer’s Disease Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (C.P.-B.); (L.Á.-S.); (G.G.-L.); (L.R.); (M.P.)
| | - Lourdes Álvarez-Sánchez
- Alzheimer’s Disease Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (C.P.-B.); (L.Á.-S.); (G.G.-L.); (L.R.); (M.P.)
| | - Gemma García-Lluch
- Alzheimer’s Disease Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (C.P.-B.); (L.Á.-S.); (G.G.-L.); (L.R.); (M.P.)
| | - Luis Raga
- Alzheimer’s Disease Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (C.P.-B.); (L.Á.-S.); (G.G.-L.); (L.R.); (M.P.)
| | - Paola Quevedo
- Alzheimer’s Disease Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (C.P.-B.); (L.Á.-S.); (G.G.-L.); (L.R.); (M.P.)
| | - Mar Peretó
- Alzheimer’s Disease Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (C.P.-B.); (L.Á.-S.); (G.G.-L.); (L.R.); (M.P.)
| | - Angel Balaguer
- Faculty of Mathematical Sciences, University of Valencia, 46100 Burjassot, Spain;
| | - Miguel Baquero
- Alzheimer’s Disease Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (C.P.-B.); (L.Á.-S.); (G.G.-L.); (L.R.); (M.P.)
- Division of Neurology, Hospital Universitari I Politècnic La Fe, 46026 Valencia, Spain
| | - Consuelo Cháfer-Pericás
- Alzheimer’s Disease Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (C.P.-B.); (L.Á.-S.); (G.G.-L.); (L.R.); (M.P.)
| |
Collapse
|
6
|
Nie Y, Chu C, Qin Q, Shen H, Wen L, Tang Y, Qu M. Lipid metabolism and oxidative stress in patients with Alzheimer's disease and amnestic mild cognitive impairment. Brain Pathol 2024; 34:e13202. [PMID: 37619589 PMCID: PMC10711261 DOI: 10.1111/bpa.13202] [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: 02/01/2023] [Accepted: 07/26/2023] [Indexed: 08/26/2023] Open
Abstract
Lipid metabolism and oxidative stress are key mechanisms in Alzheimer's disease (AD). The link between plasma lipid metabolites and oxidative stress in AD patients is poorly understood. This study was to identify markers that distinguish AD and amnestic mild cognitive impairment (aMCI) from NC, and to reveal potential links between lipid metabolites and oxidative stress. We performed non-targeted lipid metabolism analysis of plasma from patients with AD, aMCI, and NC using LC-MS/MS. The plasma malondialdehyde (MDA), glutathione peroxidase (GSH-Px), and superoxide dismutase (SOD) levels were assessed. We found significant differences in lipid metabolism between patients with AD and aMCI compared to those in NC. AD severity is associated with lipid metabolites, especially TG (18:0_16:0_18:0) + NH4, TG (18:0_16:0_16:0) + NH4, LPC(16:1e)-CH3, and PE (20:0_20:4)-H. SPH (d16:0) + H, SPH (d18:1) + H, and SPH (d18:0) + H were high-performance markers to distinguish AD and aMCI from NC. The AUC of three SPHs combined to predict AD was 0.990, with specificity and sensitivity as 0.949 and 1, respectively; the AUC of three SPHs combined to predict aMCI was 0.934, with specificity and sensitivity as 0.900, 0.981, respectively. Plasma MDA concentrations were higher in the AD group than in the NC group (p = 0.003), whereas plasma SOD levels were lower in the AD (p < 0.001) and aMCI (p = 0.045) groups than in NC, and GSH-Px activity were higher in the AD group than in the aMCI group (p = 0.007). In addition, lipid metabolites and oxidative stress are widely associated. In conclusion, this study distinguished serum lipid metabolism in AD, aMCI, and NC subjects, highlighting that the three SPHs can distinguish AD and aMCI from NC. Additionally, AD patients showed elevated oxidative stress, and there are complex interactions between lipid metabolites and oxidative stress.
Collapse
Affiliation(s)
- Yuting Nie
- Department of NeurologyXuanwu Hospital, Capital Medical UniversityBeijingChina
| | - Changbiao Chu
- Department of NeurologyXuanwu Hospital, Capital Medical UniversityBeijingChina
| | - Qi Qin
- Department of NeurologyXuanwu Hospital, Capital Medical UniversityBeijingChina
| | - Huixin Shen
- Department of NeurologyXuanwu Hospital, Capital Medical UniversityBeijingChina
| | - Lulu Wen
- Department of NeurologyXuanwu Hospital, Capital Medical UniversityBeijingChina
| | - Yi Tang
- Department of NeurologyXuanwu Hospital, Capital Medical UniversityBeijingChina
| | - Miao Qu
- Department of NeurologyXuanwu Hospital, Capital Medical UniversityBeijingChina
| |
Collapse
|
7
|
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
|
8
|
Twait EL, Andaur Navarro CL, Gudnason V, Hu YH, Launer LJ, Geerlings MI. Dementia prediction in the general population using clinically accessible variables: a proof-of-concept study using machine learning. The AGES-Reykjavik study. BMC Med Inform Decis Mak 2023; 23:168. [PMID: 37641038 PMCID: PMC10463542 DOI: 10.1186/s12911-023-02244-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 07/18/2023] [Indexed: 08/31/2023] Open
Abstract
BACKGROUND Early identification of dementia is crucial for prompt intervention for high-risk individuals in the general population. External validation studies on prognostic models for dementia have highlighted the need for updated models. The use of machine learning in dementia prediction is in its infancy and may improve predictive performance. The current study aimed to explore the difference in performance of machine learning algorithms compared to traditional statistical techniques, such as logistic and Cox regression, for prediction of all-cause dementia. Our secondary aim was to assess the feasibility of only using clinically accessible predictors rather than MRI predictors. METHODS Data are from 4,793 participants in the population-based AGES-Reykjavik Study without dementia or mild cognitive impairment at baseline (mean age: 76 years, % female: 59%). Cognitive, biometric, and MRI assessments (total: 59 variables) were collected at baseline, with follow-up of incident dementia diagnoses for a maximum of 12 years. Machine learning algorithms included elastic net regression, random forest, support vector machine, and elastic net Cox regression. Traditional statistical methods for comparison were logistic and Cox regression. Model 1 was fit using all variables and model 2 was after feature selection using the Boruta package. A third model explored performance when leaving out neuroimaging markers (clinically accessible model). Ten-fold cross-validation, repeated ten times, was implemented during training. Upsampling was used to account for imbalanced data. Tuning parameters were optimized for recalibration automatically using the caret package in R. RESULTS 19% of participants developed all-cause dementia. Machine learning algorithms were comparable in performance to logistic regression in all three models. However, a slight added performance was observed in the elastic net Cox regression in the third model (c = 0.78, 95% CI: 0.78-0.78) compared to the traditional Cox regression (c = 0.75, 95% CI: 0.74-0.77). CONCLUSIONS Supervised machine learning only showed added benefit when using survival techniques. Removing MRI markers did not significantly worsen our model's performance. Further, we presented the use of a nomogram using machine learning methods, showing transportability for the use of machine learning models in clinical practice. External validation is needed to assess the use of this model in other populations. Identifying high-risk individuals will amplify prevention efforts and selection for clinical trials.
Collapse
Affiliation(s)
- Emma L Twait
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
- Department of General Practice, Amsterdam UMC, location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
- Amsterdam Public Health, Aging & Later life and Personalized Medicine, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration and Mood, Anxiety, Psychosis, Stress, and Sleep, Amsterdam, the Netherlands
| | - Constanza L Andaur Navarro
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - Vilmunur Gudnason
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- The Icelandic Heart Association, Kopavogur, Iceland
| | - Yi-Han Hu
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Baltimore, MD, USA
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Baltimore, MD, USA
| | - Mirjam I Geerlings
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands.
- Amsterdam Public Health, Aging & Later life and Personalized Medicine, Amsterdam, the Netherlands.
- Amsterdam Neuroscience, Neurodegeneration and Mood, Anxiety, Psychosis, Stress, and Sleep, Amsterdam, the Netherlands.
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Baltimore, MD, USA.
- Department of General Practice, Amsterdam UMC, location University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands.
| |
Collapse
|
9
|
Zhao L, Liu H, Wang W, Wang Y, Xiu M, Li S. Carnitine metabolites and cognitive improvement in patients with schizophrenia treated with olanzapine: a prospective longitudinal study. Front Pharmacol 2023; 14:1255501. [PMID: 37663259 PMCID: PMC10470116 DOI: 10.3389/fphar.2023.1255501] [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: 07/08/2023] [Accepted: 08/04/2023] [Indexed: 09/05/2023] Open
Abstract
Objective: Cognitive impairment is one of the core symptoms of schizophrenia, which is stable and lifelong. L-carnitine has been shown to improve cognitive function and decrease the rate of cognitive deterioration in patients with Alzheimer's disease. However, it remains unclear regarding the role of L-carnitine and its metabolites in cognitive functions in schizophrenia after treatment with olanzapine. The purpose of this study was to evaluate the relationship between changes in plasma levels of L-carnitine metabolites and cognitive improvement after olanzapine treatment. Methods: This was a prospective longitudinal study. In this study, we recruited 25 female patients with first episode schizophrenia (FES) who were drug naïve at baseline and received 4 weeks of olanzapine monotherapy. Cognitive function was assessed at baseline and 4-week follow-up using the RBANS. Plasma L-carnitine metabolite levels were determined by a metabolomics technology based on untargeted ultra-performance liquid chromatography-mass spectrometry (UPLC-MS). Results: We found that the immediate memory index, delayed memory index and RBANS composite score were significantly increased at the 4-week follow-up after treatment. A total of 7 differential L-carnitine metabolites were identified in FES patients after olanzapine monotherapy. In addition, we found that changes in butyrylcarnitine were positively correlated with improvements in language index and RBANS composite score. Further regression analyses confirmed the association between reduced butyrylcarnitine levels and cognitive improvement after olanzapine monotherapy in FES patients. Conclusion: Our study shows that cognitive improvement after olanzapine treatment was associated with changes in L-carnitine metabolite levels in patients with FES, suggesting a key role of L-carnitine in cognition in schizophrenia.
Collapse
Affiliation(s)
- Lei Zhao
- Qingdao Mental Health Center, Qingdao, Shandong, China
| | - Hua Liu
- Qingdao Mental Health Center, Qingdao, Shandong, China
| | - Wenjuan Wang
- Qingdao Mental Health Center, Qingdao, Shandong, China
| | - Youping Wang
- Department of Nutritional and Metabolic Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China
| | - Meihong Xiu
- Beijing HuiLongGuan Hospital, Peking University HuiLongGuan Clinical Medical School, Beijing, China
| | - Shuyun Li
- Department of Nutritional and Metabolic Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
10
|
Jové M, Mota-Martorell N, Obis È, Sol J, Martín-Garí M, Ferrer I, Portero-Otin M, Pamplona R. Ether Lipid-Mediated Antioxidant Defense in Alzheimer's Disease. Antioxidants (Basel) 2023; 12:293. [PMID: 36829852 PMCID: PMC9952080 DOI: 10.3390/antiox12020293] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/20/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
One of the richest tissues in lipid content and diversity of the human body is the brain. The human brain is constitutively highly vulnerable to oxidative stress. This oxidative stress is a determinant in brain aging, as well as in the onset and progression of sporadic (late-onset) Alzheimer's disease (sAD). Glycerophospholipids are the main lipid category widely distributed in neural cell membranes, with a very significant presence for the ether lipid subclass. Ether lipids have played a key role in the evolution of the human brain compositional specificity and functionality. Ether lipids determine the neural membrane structural and functional properties, membrane trafficking, cell signaling and antioxidant defense mechanisms. Here, we explore the idea that ether lipids actively participate in the pathogenesis of sAD. Firstly, we evaluate the quantitative relevance of ether lipids in the human brain composition, as well as their role in the human brain evolution. Then, we analyze the implications of ether lipids in neural cell physiology, highlighting their inherent antioxidant properties. Finally, we discuss changes in ether lipid content associated with sAD and their physiopathological implications, and propose a mechanism that, as a vicious cycle, explains the potential significance of ether lipids in sAD.
Collapse
Affiliation(s)
- Mariona Jové
- Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), Lleida University (UdL), E-25198 Lleida, Spain
| | - Natàlia Mota-Martorell
- Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), Lleida University (UdL), E-25198 Lleida, Spain
| | - Èlia Obis
- Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), Lleida University (UdL), E-25198 Lleida, Spain
| | - Joaquim Sol
- Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), Lleida University (UdL), E-25198 Lleida, Spain
- Research Support Unit (USR), Catalan Institute of Health (ICS), Fundació Institut Universitari per a la Recerca en Atenció Primària de Salut Jordi Gol i Gurina (IDIAP JGol), E-25007 Lleida, Spain
| | - Meritxell Martín-Garí
- Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), Lleida University (UdL), E-25198 Lleida, Spain
| | - Isidre Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona (UB), E-08907 Barcelona, Spain
- Neuropathology Group, Institute of Biomedical Research of Bellvitge (IDIBELL), E-08907 Barcelona, Spain
- Network Research Center of Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, E-08907 Barcelona, Spain
| | - Manuel Portero-Otin
- Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), Lleida University (UdL), E-25198 Lleida, Spain
| | - Reinald Pamplona
- Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), Lleida University (UdL), E-25198 Lleida, Spain
| |
Collapse
|
11
|
Sheng C, Chu X, He Y, Ding Q, Jia S, Shi Q, Sun R, Song L, Du W, Liang Y, Chen N, Yang Y, Wang X. Alterations in Peripheral Metabolites as Key Actors in Alzheimer's Disease. Curr Alzheimer Res 2023; 20:379-393. [PMID: 37622711 DOI: 10.2174/1567205020666230825091147] [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: 02/28/2023] [Revised: 06/24/2023] [Accepted: 07/20/2023] [Indexed: 08/26/2023]
Abstract
Growing evidence supports that Alzheimer's disease (AD) could be regarded as a metabolic disease, accompanying central and peripheral metabolic disturbance. Nowadays, exploring novel and potentially alternative hallmarks for AD is needed. Peripheral metabolites based on blood and gut may provide new biochemical insights about disease mechanisms. These metabolites can influence brain energy homeostasis, maintain gut mucosal integrity, and regulate the host immune system, which may further play a key role in modulating the cognitive function and behavior of AD. Recently, metabolomics has been used to identify key AD-related metabolic changes and define metabolic changes during AD disease trajectory. This review aims to summarize the key blood- and microbial-derived metabolites that are altered in AD and identify the potential metabolic biomarkers of AD, which will provide future targets for precision therapeutic modulation.
Collapse
Affiliation(s)
- Can Sheng
- Department of Neurology, The Affiliated Hospital of Jining Medical University, Jining, 272000, China
| | - Xu Chu
- Department of Neurology, The Affiliated Hospital of Jining Medical University, Jining, 272000, China
| | - Yan He
- Department of Neurology, The Affiliated Hospital of Jining Medical University, Jining, 272000, China
| | - Qingqing Ding
- Department of Neurology, The Affiliated Hospital of Jining Medical University, Jining, 272000, China
| | - Shulei Jia
- Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Qiguang Shi
- Department of Neurology, The Affiliated Hospital of Jining Medical University, Jining, 272000, China
| | - Ran Sun
- Department of Neurology, The Affiliated Hospital of Jining Medical University, Jining, 272000, China
| | - Li Song
- Department of Neurology, The Affiliated Hospital of Jining Medical University, Jining, 272000, China
| | - Wenying Du
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Yuan Liang
- Department of Clinical Medicine, Jining Medical University, Jining, 272067, China
| | - Nian Chen
- Department of Clinical Medicine, Jining Medical University, Jining, 272067, China
| | - Yan Yang
- Department of Neurology, The Affiliated Hospital of Jining Medical University, Jining, 272000, China
| | - Xiaoni Wang
- Department of Neurology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, China
| |
Collapse
|
12
|
Longitudinal Analysis of the Microbiome and Metabolome in the 5xfAD Mouse Model of Alzheimer's Disease. mBio 2022; 13:e0179422. [PMID: 36468884 PMCID: PMC9765021 DOI: 10.1128/mbio.01794-22] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Recent reports implicate gut microbiome dysbiosis in the onset and progression of Alzheimer's disease (AD), yet studies involving model animals overwhelmingly omit the microbial perspective. Here, we evaluate longitudinal microbiomes and metabolomes from a popular transgenic mouse model for familial AD (5xfAD). Cecal and fecal samples from 5xfAD and wild-type B6J (WT) mice from 4 to 18 months of age were subjected to shotgun Illumina sequencing. Metabolomics was performed on plasma and feces from a subset of the same animals. Significant genotype, sex, age, and cage-specific differences were observed in the microbiome, with the variance explained by genotype at 4 and 18 months of age rising from 0.9 to 9% and 0.3 to 8% for the cecal and fecal samples, respectively. Bacteria at significantly higher abundances in AD mice include multiple Alistipes spp., two Ligilactobacillus spp., and Lactobacillus sp. P38, while multiple species of Turicibacter, Lactobacillus johnsonii, and Romboutsia ilealis were less abundant. Turicibacter is similarly depleted in people with AD, and members of this genus both consume and induce the production of gut-derived serotonin. Contradicting previous findings in humans, serotonin is significantly more concentrated in the blood of older 5xfAD animals compared to their WT littermates. 5xfAD animals exhibited significantly lower plasma concentrations of carnosine and the lysophospholipid lysoPC a C18:1. Correlations between the microbiome and metabolome were also explored. Taken together, these findings strengthen the link between Turicibacter abundance and AD, provide a basis for further microbiome studies of murine models for AD, and suggest that greater control over animal model microbiomes is needed in AD research. IMPORTANCE Microorganisms residing within the gastrointestinal tract are implicated in the onset and progression of Alzheimer's disease (AD) through the mediation of inflammation, exchange of small-molecules across the blood-brain barrier, and stimulation of the vagus nerve. Unfortunately, most animal models for AD are housed under conditions that do not reflect real-world human microbial exposure and do not sufficiently account for (or meaningfully consider) variations in the microbiome. An improved understanding of AD model animal microbiomes will increase model efficacy and the translatability of research findings into humans. Here, we present the characterization of the microbiome and metabolome of the 5xfAD mouse model, which is one of the most common animal models for familial AD. The manuscript highlights the importance of considering the microbiome in study design and aims to lay the groundwork for future studies involving mouse models for AD.
Collapse
|
13
|
Kawakami J, Piccolo SR, Kauwe JK, Graves SW. Gender differences contribute to variability of serum lipid biomarkers for Alzheimer's disease. Biomark Med 2022; 16:1089-1100. [PMID: 36625236 DOI: 10.2217/bmm-2022-0462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Background: Alzheimer's disease (AD) cannot currently be diagnosed by a blood test. One reason may be gender differences. Another may be the statistical methods used. The authors evaluate these possibilities. Objective: The authors applied serum lipidomics to find AD biomarkers in men and women. They hypothesized that AD biomarkers would differ between genders and that machine-learning algorithms would improve diagnostic performance. Methods: Serum lipids were analyzed by mass spectrometry for a training set of AD cases and controls and in a blinded test set. Statistical analyses considered gender differences. Results: Lipids best classifying AD subjects differed significantly between men and women. Robust statistical algorithms did not improve diagnostic performance. Conclusion: Poor performance of AD biomarkers appears to be due primarily to inherent variability in AD patients.
Collapse
Affiliation(s)
- Jie Kawakami
- Department of Chemistry & Biochemistry, Brigham Young University, Provo, UT 84602, USA
| | - Stephen R Piccolo
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - John Ks Kauwe
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Steven W Graves
- Department of Chemistry & Biochemistry, Brigham Young University, Provo, UT 84602, USA
| |
Collapse
|
14
|
Chen J, Cao D, Jiang S, Liu X, Pan W, Cui H, Yang W, Liu Z, Jin J, Zhao Z. Triterpenoid saponins from Ilex pubescens promote blood circulation in blood stasis syndrome by regulating sphingolipid metabolism and the PI3K/AKT/eNOS signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 104:154242. [PMID: 35728385 DOI: 10.1016/j.phymed.2022.154242] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 05/27/2022] [Accepted: 06/04/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Blood stasis syndrome (BSS) is a severe disorder involving disturbances in glycerophosphocholine metabolism. Ilex pubescens (IP) can regulate the levels of lipids, such as lysophosphatidylcholine (LPC) and lysophosphatidylethanolamine (LPE); however, the main active constituent of IP and its corresponding mechanism in BSS treatment are still unclear. PURPOSE To explore the mechanisms by which triterpenoid saponins of IP (IPTS) promote blood circulation using system pharmacology-based approaches. METHODS Sprague-Dawley (SD) rat BSS model was prepared by oral administration of IPTS for 7 days followed by adrenaline hydrochloride injection before immersion in ice water. Coagulation parameters in plasma and thromboxane B2 (TXB2), endothelin (ET) and 6-keto-PGF1α in serum were measured. The possible influence on abdominal aortas was evaluated by histopathology assessment. Human vein endothelial cells (HUVECs) were incubated with ox-LDL, and the effects of IPTS on cell viability and LDH release were investigated. UPLC-QTOF-MS/MS was used for metabolic profile analysis of lipid-soluble components in rat plasma and intracellular metabolites in HUVECs. Network pharmacology was used to predict the relevant targets and model pathways of BSS and the main components of IPTS. Molecular docking, molecular dynamics (MD) simulation and biochemical assays were used to predict molecular interactions between the active components of IPTS and target proteins. RT-PCR was used to detect the mRNA level of target proteins. Western blotting and immunohistochemistry (IHC) were used to verify the mechanisms by which IPTS promotes blood circulation in BSS. RESULTS IPTS improved blood biochemical function in the process of BSS and played a role in vascular protection and maintenance of the normal morphology of blood vessels. Furthermore, metabolite pathways involved in steroid biosynthesis and sphingolipid metabolism were significantly perturbed. Both metabolomics analysis and network pharmacology results showed that IPTS ameliorates vascular injury and that lipid accumulation may be mediated by PI3K/AKT signaling pathway activation. MD simulation and enzyme inhibitory activity results suggested that the main components of IPTS can form stable complexes with PI3K, AKT and eNOS and that the complexes have significant binding affinity. PI3K, AKT, p-AKT, and eNOS mRNA and protein levels were considerably elevated in the IPTS-treated group. Thus, IPTS protects the vasculature by regulating the PI3K/AKT signaling pathway, activating eNOS and increasing the release of NO. CONCLUSION A possible mechanism by which IPTS prevents BSS is proposed: IPTS can promote blood circulation by modulating sphingolipid metabolism and activating the PI3K/AKT/eNOS signaling pathways.
Collapse
Affiliation(s)
- Jie Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Di Cao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; School of Pharmacy, Wannan Medical College, Anhui, 241002, China
| | - Shiqin Jiang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Xia Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Wencong Pan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Hui Cui
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Weiqun Yang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Zhongqiu Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Jing Jin
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China.
| | - Zhongxiang Zhao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| |
Collapse
|
15
|
Kadyrov M, Whiley L, Brown B, Erickson KI, Holmes E. Associations of the Lipidome with Ageing, Cognitive Decline and Exercise Behaviours. Metabolites 2022; 12:metabo12090822. [PMID: 36144226 PMCID: PMC9505967 DOI: 10.3390/metabo12090822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/22/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
One of the most recognisable features of ageing is a decline in brain health and cognitive dysfunction, which is associated with perturbations to regular lipid homeostasis. Although ageing is the largest risk factor for several neurodegenerative diseases such as dementia, a loss in cognitive function is commonly observed in adults over the age of 65. Despite the prevalence of normal age-related cognitive decline, there is a lack of effective methods to improve the health of the ageing brain. In light of this, exercise has shown promise for positively influencing neurocognitive health and associated lipid profiles. This review summarises age-related changes in several lipid classes that are found in the brain, including fatty acyls, glycerolipids, phospholipids, sphingolipids and sterols, and explores the consequences of age-associated pathological cognitive decline on these lipid classes. Evidence of the positive effects of exercise on the affected lipid profiles are also discussed to highlight the potential for exercise to be used therapeutically to mitigate age-related changes to lipid metabolism and prevent cognitive decline in later life.
Collapse
Affiliation(s)
- Maria Kadyrov
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, 5 Robin Warren Drive, Murdoch, WA 6150, Australia
- Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, 5 Robin Warren Drive, Murdoch, WA 6150, Australia
- Discipline of Exercise Science, College of Science, Health, Engineering and Education, Murdoch University, 90 South Street, Murdoch, WA 6150, Australia
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, 90 South Street, Murdoch, WA 6150, Australia
- Correspondence: (M.K.); (B.B.); (E.H.)
| | - Luke Whiley
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, 5 Robin Warren Drive, Murdoch, WA 6150, Australia
- Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, 5 Robin Warren Drive, Murdoch, WA 6150, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Belinda Brown
- Discipline of Exercise Science, College of Science, Health, Engineering and Education, Murdoch University, 90 South Street, Murdoch, WA 6150, Australia
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, 90 South Street, Murdoch, WA 6150, Australia
- School of Medical Sciences, Sarich Neuroscience Research Institute, Edith Cowan University, Nedlands, WA 6009, Australia
- Correspondence: (M.K.); (B.B.); (E.H.)
| | - Kirk I. Erickson
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA 15260, USA
- AdventHealth Research Institute, Neuroscience Institute, Orlando, FL 32804, USA
- PROFITH “PROmoting FITness and Health Through Physical Activity” Research Group, Sport and Health University Research Institute (iMUDS), Department of Physical Education and Sports, Faculty of Sport Sciences, University of Granada, 18071 Granada, Spain
| | - Elaine Holmes
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, 5 Robin Warren Drive, Murdoch, WA 6150, Australia
- Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, 5 Robin Warren Drive, Murdoch, WA 6150, Australia
- Division of Integrative Systems and Digestive Medicine, Department of Surgery and Cancer, Imperial College London, London SW7 2AZ, UK
- Correspondence: (M.K.); (B.B.); (E.H.)
| |
Collapse
|
16
|
Casas-Fernández E, Peña-Bautista C, Baquero M, Cháfer-Pericás C. Lipids as Early and Minimally Invasive Biomarkers for Alzheimer's Disease. Curr Neuropharmacol 2022; 20:1613-1631. [PMID: 34727857 PMCID: PMC9881089 DOI: 10.2174/1570159x19666211102150955] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/09/2021] [Accepted: 10/19/2021] [Indexed: 11/22/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder worldwide. Specifically, typical late-onset AD is a sporadic form with a complex etiology that affects over 90% of patients. The current gold standard for AD diagnosis is based on the determination of amyloid status by analyzing cerebrospinal fluid samples or brain positron emission tomography. These procedures can be used widely as they have several disadvantages (expensive, invasive). As an alternative, blood metabolites have recently emerged as promising AD biomarkers. Small molecules that cross the compromised AD blood-brain barrier could be determined in plasma to improve clinical AD diagnosis at early stages through minimally invasive techniques. Specifically, lipids could play an important role in AD since the brain has a high lipid content, and they are present ubiquitously inside amyloid plaques. Therefore, a systematic review was performed with the aim of identifying blood lipid metabolites as potential early AD biomarkers. In conclusion, some lipid families (fatty acids, glycerolipids, glycerophospholipids, sphingolipids, lipid peroxidation compounds) have shown impaired levels at early AD stages. Ceramide levels were significantly higher in AD subjects, and polyunsaturated fatty acids levels were significantly lower in AD. Also, high arachidonic acid levels were found in AD patients in contrast to low sphingomyelin levels. Consequently, these lipid biomarkers could be used for minimally invasive and early AD clinical diagnosis.
Collapse
Affiliation(s)
| | | | - Miguel Baquero
- Division of Neurology, University and Polytechnic Hospital La Fe, Valencia, Spain
| | - Consuelo Cháfer-Pericás
- Health Research Institute La Fe, Valencia, Spain;,Address correspondence to this author at the Health Research Institute La Fe, Avenida Fernando Abril Martorell 106, Valencia E46026, Spain;, Tel: +34-96 1246721; E-mail:
| |
Collapse
|
17
|
Microbial-derived metabolites as a risk factor of age-related cognitive decline and dementia. Mol Neurodegener 2022; 17:43. [PMID: 35715821 PMCID: PMC9204954 DOI: 10.1186/s13024-022-00548-6] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 05/30/2022] [Indexed: 02/06/2023] Open
Abstract
A consequence of our progressively ageing global population is the increasing prevalence of worldwide age-related cognitive decline and dementia. In the absence of effective therapeutic interventions, identifying risk factors associated with cognitive decline becomes increasingly vital. Novel perspectives suggest that a dynamic bidirectional communication system between the gut, its microbiome, and the central nervous system, commonly referred to as the microbiota-gut-brain axis, may be a contributing factor for cognitive health and disease. However, the exact mechanisms remain undefined. Microbial-derived metabolites produced in the gut can cross the intestinal epithelial barrier, enter systemic circulation and trigger physiological responses both directly and indirectly affecting the central nervous system and its functions. Dysregulation of this system (i.e., dysbiosis) can modulate cytotoxic metabolite production, promote neuroinflammation and negatively impact cognition. In this review, we explore critical connections between microbial-derived metabolites (secondary bile acids, trimethylamine-N-oxide (TMAO), tryptophan derivatives and others) and their influence upon cognitive function and neurodegenerative disorders, with a particular interest in their less-explored role as risk factors of cognitive decline.
Collapse
|
18
|
A retrotransposon storm marks clinical phenoconversion to late-onset Alzheimer's disease. GeroScience 2022; 44:1525-1550. [PMID: 35585302 PMCID: PMC9213607 DOI: 10.1007/s11357-022-00580-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 04/26/2022] [Indexed: 12/03/2022] Open
Abstract
Recent reports have suggested that the reactivation of otherwise transcriptionally silent transposable elements (TEs) might induce brain degeneration, either by dysregulating the expression of genes and pathways implicated in cognitive decline and dementia or through the induction of immune-mediated neuroinflammation resulting in the elimination of neural and glial cells. In the work we present here, we test the hypothesis that differentially expressed TEs in blood could be used as biomarkers of cognitive decline and development of AD. To this aim, we used a sample of aging subjects (age > 70) that developed late-onset Alzheimer’s disease (LOAD) over a relatively short period of time (12–48 months), for which blood was available before and after their phenoconversion, and a group of cognitive stable subjects as controls. We applied our developed and validated customized pipeline that allows the identification, characterization, and quantification of the differentially expressed (DE) TEs before and after the onset of manifest LOAD, through analyses of RNA-Seq data. We compared the level of DE TEs within more than 600,000 TE-mapping RNA transcripts from 25 individuals, whose specimens we obtained before and after their phenotypic conversion (phenoconversion) to LOAD, and discovered that 1790 TE transcripts showed significant expression differences between these two timepoints (logFC ± 1.5, logCMP > 5.3, nominal p value < 0.01). These DE transcripts mapped both over- and under-expressed TE elements. Occurring before the clinical phenoconversion, this TE storm features significant increases in DE transcripts of LINEs, LTRs, and SVAs, while those for SINEs are significantly depleted. These dysregulations end with signs of manifest LOAD. This set of highly DE transcripts generates a TE transcriptional profile that accurately discriminates the before and after phenoconversion states of these subjects. Our findings suggest that a storm of DE TEs occurs before phenoconversion from normal cognition to manifest LOAD in risk individuals compared to controls, and may provide useful blood-based biomarkers for heralding such a clinical transition, also suggesting that TEs can indeed participate in the complex process of neurodegeneration.
Collapse
|
19
|
Khan MJ, Chung NA, Hansen S, Dumitrescu L, Hohman TJ, Kamboh MI, Lopez OL, Robinson RAS. Targeted Lipidomics To Measure Phospholipids and Sphingomyelins in Plasma: A Pilot Study To Understand the Impact of Race/Ethnicity in Alzheimer's Disease. Anal Chem 2022; 94:4165-4174. [PMID: 35235294 PMCID: PMC9126486 DOI: 10.1021/acs.analchem.1c03821] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The number of people suffering from Alzheimer's disease (AD) is increasing rapidly every year. One aspect of AD that is often overlooked is the disproportionate incidence of AD among African American/Black populations. With the recent development of novel assays for lipidomics analysis in recent times, there has been a drastic increase in the number of studies focusing on changes of lipids in AD. However, very few of these studies have focused on or even included samples from African American/Black individuals samples. In this study, we aimed to determine if the lipidome in AD is universal across non-Hispanic White and African American/Black individuals. To accomplish this, a targeted mass spectrometry lipidomics analysis was performed on plasma samples (N = 113) obtained from cognitively normal (CN, N = 54) and AD (N = 59) individuals from African American/Black (N = 56) and non-Hispanic White (N = 57) backgrounds. Five lipids (PS 18:0_18:0, PS 18:0_20:0, PC 16:0_22:6, PC 18:0_22:6, and PS 18:1_22:6) were altered between AD and CN sample groups (p value < 0.05). Upon racial stratification, there were notable differences in lipids that were unique to African American/Black or non-Hispanic White individuals. PS 20:0_20:1 was reduced in AD in samples from non-Hispanic White but not African American/Black adults. We also tested whether race/ethnicity significantly modified the association between lipids and AD status by including a race × diagnosis interaction term in a linear regression model. PS 20:0_20:1 showed a significant interaction (p = 0.004). The discovery of lipid changes in AD in this study suggests that identifying relevant lipid biomarkers for diagnosis will require diversity in sample cohorts.
Collapse
Affiliation(s)
- Mostafa J Khan
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Nadjali A Chung
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Shania Hansen
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, Tennessee 37212, United States
| | - Logan Dumitrescu
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, Tennessee 37212, United States.,Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States.,Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States.,Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Timothy J Hohman
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, Tennessee 37212, United States
| | - M Ilyas Kamboh
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States.,Department of Human Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States.,Department of Epidemiology, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States
| | - Oscar L Lopez
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States.,Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States
| | - Renã A S Robinson
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States.,Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, Tennessee 37212, United States.,Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States.,Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States.,Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37232, United States
| |
Collapse
|
20
|
Munasinghe M, Afshari R, Heydarian D, Almotayri A, Dias DA, Thomas J, Jois M. Effects of cocoa on altered metabolite levels in purine metabolism pathways and urea cycle in Alzheimer's disease in C. elegans. TRANSLATIONAL MEDICINE OF AGING 2022. [DOI: 10.1016/j.tma.2022.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
21
|
Pathak N, Vimal SK, Tandon I, Agrawal L, Hongyi C, Bhattacharyya S. Neurodegenerative Disorders of Alzheimer, Parkinsonism, Amyotrophic Lateral Sclerosis and Multiple Sclerosis: An Early Diagnostic Approach for Precision Treatment. Metab Brain Dis 2022; 37:67-104. [PMID: 34719771 DOI: 10.1007/s11011-021-00800-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 07/11/2021] [Indexed: 12/21/2022]
Abstract
Neurodegenerative diseases (NDs) are characterised by progressive dysfunction of synapses, neurons, glial cells and their networks. Neurodegenerative diseases can be classified according to primary clinical features (e.g., dementia, parkinsonism, or motor neuron disease), anatomic distribution of neurodegeneration (e.g., frontotemporal degenerations, extrapyramidal disorders, or spinocerebellar degenerations), or principal molecular abnormalities. The most common neurodegenerative disorders are amyloidosis, tauopathies, a-synucleinopathy, and TAR DNA-binding protein 43 (TDP-43) proteopathy. The protein abnormalities in these disorders have abnormal conformational properties along with altered cellular mechanisms, and they exhibit motor deficit, mitochondrial malfunction, dysfunctions in autophagic-lysosomal pathways, synaptic toxicity, and more emerging mechanisms such as the roles of stress granule pathways and liquid-phase transitions. Finally, for each ND, microglial cells have been reported to be implicated in neurodegeneration, in particular, because the microglial responses can shift from neuroprotective to a deleterious role. Growing experimental evidence suggests that abnormal protein conformers act as seed material for oligomerization, spreading from cell to cell through anatomically connected neuronal pathways, which may in part explain the specific anatomical patterns observed in brain autopsy sample. In this review, we mention the human pathology of select neurodegenerative disorders, focusing on how neurodegenerative disorders (i.e., Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and multiple sclerosis) represent a great healthcare problem worldwide and are becoming prevalent because of the increasing aged population. Despite many studies have focused on their etiopathology, the exact cause of these diseases is still largely unknown and until now with the only available option of symptomatic treatments. In this review, we aim to report the systematic and clinically correlated potential biomarker candidates. Although future studies are necessary for their use in early detection and progression in humans affected by NDs, the promising results obtained by several groups leads us to this idea that biomarkers could be used to design a potential therapeutic approach and preclinical clinical trials for the treatments of NDs.
Collapse
Affiliation(s)
- Nishit Pathak
- Department of Pharmaceutical Sciences and Chinese Traditional Medicine, Southwest University, Beibei, Chongqing, 400715, People's Republic of China
| | - Sunil Kumar Vimal
- Department of Pharmaceutical Sciences and Chinese Traditional Medicine, Southwest University, Beibei, Chongqing, 400715, People's Republic of China
| | - Ishi Tandon
- Amity University Jaipur, Rajasthan, Jaipur, Rajasthan, India
| | - Lokesh Agrawal
- Graduate School of Comprehensive Human Sciences, Kansei Behavioural and Brain Sciences, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Cao Hongyi
- Department of Pharmaceutical Sciences and Chinese Traditional Medicine, Southwest University, Beibei, Chongqing, 400715, People's Republic of China
| | - Sanjib Bhattacharyya
- Department of Pharmaceutical Sciences and Chinese Traditional Medicine, Southwest University, Beibei, Chongqing, 400715, People's Republic of China.
| |
Collapse
|
22
|
Jia L, Yang J, Zhu M, Pang Y, Wang Q, Wei Q, Li Y, Li T, Li F, Wang Q, Li Y, Wei Y. A metabolite panel that differentiates Alzheimer's disease from other dementia types. Alzheimers Dement 2021; 18:1345-1356. [PMID: 34786838 PMCID: PMC9545206 DOI: 10.1002/alz.12484] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 08/24/2021] [Accepted: 08/30/2021] [Indexed: 11/12/2022]
Abstract
Introduction Alzheimer's disease (AD) is associated with altered metabolites. This study aimed to determine the validity of using circulating metabolites to differentiate AD from other dementias. Methods Blood metabolites were measured in three data sets. Data set 1 (controls, 27; AD, 28) was used for analyzing differential metabolites. Data set 2 (controls, 93; AD, 92) was used to establish a diagnostic AD model with use of a metabolite panel. The model was applied to Data set 3 (controls, 76; AD, 76; other dementias, 205) to verify its capacity for differentiating AD from other dementias. Results Data set 1 revealed 7 upregulated and 77 downregulated metabolites. In Data set 2, a panel of 11 metabolites was included in a model that could distinguish AD from controls. In Data set 3, this panel was used to successfully differentiate AD from other dementias. Discussion This study revealed an AD‐specific panel of 11 metabolites that may be used for AD diagnosis.
Collapse
Affiliation(s)
- Longfei Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Jianwei Yang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Min Zhu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Yana Pang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Qi Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Qin Wei
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Ying Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - TingTing Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Fangyu Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Qigeng Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Yan Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Yiping Wei
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| |
Collapse
|
23
|
Watanabe Y, Kasuga K, Tokutake T, Kitamura K, Ikeuchi T, Nakamura K. Alterations in Glycerolipid and Fatty Acid Metabolic Pathways in Alzheimer's Disease Identified by Urinary Metabolic Profiling: A Pilot Study. Front Neurol 2021; 12:719159. [PMID: 34777195 PMCID: PMC8578168 DOI: 10.3389/fneur.2021.719159] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 10/06/2021] [Indexed: 11/13/2022] Open
Abstract
An easily accessible and non-invasive biomarker for the early detection of Alzheimer's disease (AD) is needed. Evidence suggests that metabolic dysfunction underlies the pathophysiology of AD. While urine is a non-invasively collectable biofluid and a good source for metabolomics analysis, it is not yet widely used for this purpose. This small-scale pilot study aimed to examine whether the metabolic profile of urine from AD patients reflects the metabolic dysfunction reported to underlie AD pathology, and to identify metabolites that could distinguish AD patients from cognitively healthy controls. Spot urine of 18 AD patients (AD group) and 18 age- and sex-matched, cognitively normal controls (control group) were analyzed by mass spectrometry (MS). Capillary electrophoresis time-of-flight MS and liquid chromatography–Fourier transform MS were used to cover a larger range of molecules with ionic as well as lipid characteristics. A total of 304 ionic molecules and 81 lipid compounds of 12 lipid classes were identified. Of these, 26 molecules showed significantly different relative concentrations between the AD and control groups (Wilcoxon's rank-sum test). Moreover, orthogonal partial least-squares discriminant analysis revealed significant discrimination between the two groups. Pathway searches using the KEGG database, and pathway enrichment and topology analysis using Metaboanalyst software, suggested alterations in molecules relevant to pathways of glycerolipid and glycerophospholipid metabolism, thermogenesis, and caffeine metabolism in AD patients. Further studies of urinary metabolites will contribute to the early detection of AD and understanding of its pathogenesis.
Collapse
Affiliation(s)
- Yumi Watanabe
- Division of Preventive Medicine, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Kensaku Kasuga
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Takayoshi Tokutake
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Kaori Kitamura
- Division of Preventive Medicine, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Kazutoshi Nakamura
- Division of Preventive Medicine, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| |
Collapse
|
24
|
Discovery of a Metabolic Signature Predisposing High Risk Patients with Mild Cognitive Impairment to Converting to Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms222010903. [PMID: 34681563 PMCID: PMC8535253 DOI: 10.3390/ijms222010903] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 11/17/2022] Open
Abstract
Assessing dementia conversion in patients with mild cognitive impairment (MCI) remains challenging owing to pathological heterogeneity. While many MCI patients ultimately proceed to Alzheimer’s disease (AD), a subset of patients remain stable for various times. Our aim was to characterize the plasma metabolites of nineteen MCI patients proceeding to AD (P-MCI) and twenty-nine stable MCI (S-MCI) patients by untargeted metabolomics profiling. Alterations in the plasma metabolites between the P-MCI and S-MCI groups, as well as between the P-MCI and AD groups, were compared over the observation period. With the help of machine learning-based stratification, a 20-metabolite signature panel was identified that was associated with the presence and progression of AD. Furthermore, when the metabolic signature panel was used for classification of the three patient groups, this gave an accuracy of 73.5% using the panel. Moreover, when specifically classifying the P-MCI and S-MCI subjects, a fivefold cross-validation accuracy of 80.3% was obtained using the random forest model. Importantly, indole-3-propionic acid, a bacteria-generated metabolite from tryptophan, was identified as a predictor of AD progression, suggesting a role for gut microbiota in AD pathophysiology. Our study establishes a metabolite panel to assist in the stratification of MCI patients and to predict conversion to AD.
Collapse
|
25
|
Nielsen JE, Maltesen RG, Havelund JF, Færgeman NJ, Gotfredsen CH, Vestergård K, Kristensen SR, Pedersen S. Characterising Alzheimer's disease through integrative NMR- and LC-MS-based metabolomics. Metabol Open 2021; 12:100125. [PMID: 34622190 PMCID: PMC8479251 DOI: 10.1016/j.metop.2021.100125] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/10/2021] [Accepted: 09/10/2021] [Indexed: 12/14/2022] Open
Abstract
Background Alzheimer's Disease (AD) is a complex and multifactorial disease and novel approaches are needed to illuminate the underlying pathology. Metabolites comprise the end-product of genes, transcripts, and protein regulations and might reflect disease pathogenesis. Blood is a common biofluid used in metabolomics; however, since extracellular vesicles (EVs) hold cell-specific biological material and can cross the blood-brain barrier, their utilization as biological material warrants further investigation. We aimed to investigate blood- and EV-derived metabolites to add insigts to the pathological mechanisms of AD. Methods Blood samples were collected from 10 AD and 10 Mild Cognitive Impairment (MCI) patients, and 10 healthy controls. EVs were enriched from plasma using 100,000×g, 1 h, 4 °C with a wash. Metabolites from serum and EVs were measured using liquid chromatography-mass spectrometry (LC-MS) and nuclear magnetic resonance (NMR) spectroscopy. Multivariate and univariate analyses were employed to identify altered metabolites in cognitively impaired individuals. Results While no significant EV-derived metabolites were found differentiating patients from healthy individuals, six serum metabolites were found important; valine (p = 0.001, fold change, FC = 0.8), histidine (p = 0.001, FC = 0.9), allopurinol riboside (p = 0.002, FC = 0.2), inosine (p = 0.002, FC = 0.3), 4-pyridoxic acid (p = 0.006, FC = 1.6), and guanosine (p = 0.004, FC = 0.3). Pathway analysis revealed branched-chain amino acids, purine and histidine metabolisms to be downregulated, and vitamin B6 metabolism upregulated in patients compared to controls. Conclusion Using a combination of LC-MS and NMR methodologies we identified several altered mechanisms possibly related to AD pathology. EVs require additional optimization prior to their possible utilization as a biological material for AD-related metabolomics studies.
Collapse
Key Words
- ACE, Addenbrooke's cognitive examination
- AD, Alzheimer's Disease
- AUC, Area under the curve
- Alzheimer
- Aβ, Amyloid-β
- BBB, Blood-brain barrier
- BCAA, Branched-chain amino acid
- Blood
- CNS, Central nervous system
- CSF, Cerebrospinal fluid
- CV, Cross-validation
- EVs, Extracellular vesicles
- Extracellular vesicles
- FAQ, Functional activities questionnaire
- FDR, False discovery rate
- MCI, Mild cognitive impairment
- MMSE, Mini-mental state examination
- Mass spectrometry
- Metabolites
- Nuclear magnetic resonance
- PCA, Principal component analysis
- ROC, Receiver operating characteristics
- p-tau, Phospho-tau
- sPLS-DA, Sparse partial least squared discriminant analysis
- t-tau, Total-tau
Collapse
Affiliation(s)
- Jonas Ellegaard Nielsen
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.,Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
| | - Raluca Georgiana Maltesen
- Translational Radiation Biology and Oncology Laboratory, Centre for Cancer Research, Westmead Institute of Medical Research, Westmead, Australia.,Department of Anaesthesia and Intensive Care, Aalborg University Hospital, Aalborg, Denmark
| | - Jesper F Havelund
- Department of Biochemistry and Molecular Biology, Villum Center for Bioanalytical Sciences, University of Southern Denmark, Odense, Denmark
| | - Nils J Færgeman
- Department of Biochemistry and Molecular Biology, Villum Center for Bioanalytical Sciences, University of Southern Denmark, Odense, Denmark
| | | | | | - Søren Risom Kristensen
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.,Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
| | - Shona Pedersen
- Department of Basic Medical Sciences, College of Medicine, Qatar University, Qatar Health, Doha, Qatar
| |
Collapse
|
26
|
Bergland AK, Proitsi P, Kirsebom BE, Soennesyn H, Hye A, Larsen AI, Xu J, Legido-Quigley C, Rajendran L, Fladby T, Aarsland D. Exploration of Plasma Lipids in Mild Cognitive Impairment due to Alzheimer's Disease. J Alzheimers Dis 2021; 77:1117-1127. [PMID: 32804144 DOI: 10.3233/jad-200441] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Lipids have important structural roles in cell membranes and changes to these membrane lipids may influence β- and γ-secretase activities and thus contribute to Alzheimer's disease (AD) pathology. OBJECTIVE To explore baseline plasma lipid profiling in participants with mild cognitive impairment (MCI) with and without AD pathology. METHODS We identified 261 plasma lipids using reversed-phase liquid chromatography/mass spectrometry in cerebrospinal fluid amyloid positive (Aβ+) or negative (Aβ-) participants with MCI as compared to controls. Additionally, we analyzed the potential associations of plasma lipid profiles with performance on neuropsychological tests at baseline and after two years. RESULTS Sphingomyelin (SM) concentrations, particularly, SM(d43:2), were lower in MCI Aβ+ individuals compared to controls. Further, SM(d43:2) was also nominally reduced in MCI Aβ+ individuals compared to MCI Aβ-. No plasma lipids were associated with performance on primary neuropsychological tests at baseline or between the two time points after correction for multiple testing. CONCLUSION Reduced plasma concentrations of SM were associated with AD.
Collapse
Affiliation(s)
- Anne Katrine Bergland
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Petroula Proitsi
- Maurice Wohl Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Bjørn-Eivind Kirsebom
- Department of Neurology, University Hospital of North Norway, Tromsø, Norway.,Department of Psychology, Faculty of Health Sciences, UiT, The Arctic University of Norway, Tromsø, Norway
| | - Hogne Soennesyn
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Abdul Hye
- Maurice Wohl Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Alf Inge Larsen
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Cardiology, Stavanger University Hospital, Stavanger, Norway
| | - Jin Xu
- Maurice Wohl Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.,Institute of Pharmaceutical Science, King's College London, London, UK
| | - Cristina Legido-Quigley
- Institute of Pharmaceutical Science, King's College London, London, UK.,Systems Medicine, Steno Diabetes Centre, Copenhagen, Denmark
| | - Lawrence Rajendran
- UK Dementia Research Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Tormod Fladby
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway.,Institute of Clinical Medicine, Campus Ahus, University of Oslo, Oslo, Norway
| | - Dag Aarsland
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway.,UK Dementia Research Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| |
Collapse
|
27
|
Hampel H, Nisticò R, Seyfried NT, Levey AI, Modeste E, Lemercier P, Baldacci F, Toschi N, Garaci F, Perry G, Emanuele E, Valenzuela PL, Lucia A, Urbani A, Sancesario GM, Mapstone M, Corbo M, Vergallo A, Lista S. Omics sciences for systems biology in Alzheimer's disease: State-of-the-art of the evidence. Ageing Res Rev 2021; 69:101346. [PMID: 33915266 DOI: 10.1016/j.arr.2021.101346] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 04/06/2021] [Accepted: 04/22/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is characterized by non-linear, genetic-driven pathophysiological dynamics with high heterogeneity in biological alterations and disease spatial-temporal progression. Human in-vivo and post-mortem studies point out a failure of multi-level biological networks underlying AD pathophysiology, including proteostasis (amyloid-β and tau), synaptic homeostasis, inflammatory and immune responses, lipid and energy metabolism, oxidative stress. Therefore, a holistic, systems-level approach is needed to fully capture AD multi-faceted pathophysiology. Omics sciences - genomics, epigenomics, transcriptomics, proteomics, metabolomics, lipidomics - embedded in the systems biology (SB) theoretical and computational framework can generate explainable readouts describing the entire biological continuum of a disease. Such path in Neurology is encouraged by the promising results of omics sciences and SB approaches in Oncology, where stage-driven pathway-based therapies have been developed in line with the precision medicine paradigm. Multi-omics data integrated in SB network approaches will help detect and chart AD upstream pathomechanistic alterations and downstream molecular effects occurring in preclinical stages. Finally, integrating omics and neuroimaging data - i.e., neuroimaging-omics - will identify multi-dimensional biological signatures essential to track the clinical-biological trajectories, at the subpopulation or even individual level.
Collapse
|
28
|
Lipid Peroxidation Assessment in Preclinical Alzheimer Disease Diagnosis. Antioxidants (Basel) 2021; 10:antiox10071043. [PMID: 34209667 PMCID: PMC8300760 DOI: 10.3390/antiox10071043] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Alzheimer disease (AD) is an increasingly common neurodegenerative disease, especially in countries with aging populations. Its diagnosis is complex and is usually carried out in advanced stages of the disease. In addition, lipids and oxidative stress have been related to AD since the earliest stages. A diagnosis in the initial or preclinical stages of the disease could help in a more effective action of the treatments. METHODS Isoprostanoid biomarkers were determined in plasma samples from preclinical AD participants (n = 12) and healthy controls (n = 31) by chromatography and mass spectrometry (UPLC-MS/MS). Participants were accurately classified according to cerebrospinal fluid (CSF) biomarkers and neuropsychological examination. RESULTS Isoprostanoid levels did not show differences between groups. However, some of them correlated with CSF biomarkers (t-tau, p-tau) and with cognitive decline. In addition, a panel including 10 biomarkers showed an area under curve (AUC) of 0.96 (0.903-1) and a validation AUC of 0.90 in preclinical AD prediction. CONCLUSIONS Plasma isoprostanoids could be useful biomarkers in preclinical diagnosis for AD. However, these results would require a further validation with an external cohort.
Collapse
|
29
|
Llorente-Ovejero A, Martínez-Gardeazabal J, Moreno-Rodríguez M, Lombardero L, González de San Román E, Manuel I, Giralt MT, Rodríguez-Puertas R. Specific Phospholipid Modulation by Muscarinic Signaling in a Rat Lesion Model of Alzheimer's Disease. ACS Chem Neurosci 2021; 12:2167-2181. [PMID: 34037379 PMCID: PMC9162383 DOI: 10.1021/acschemneuro.1c00169] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
![]()
Alzheimer’s disease (AD) represents
the most common cause
of dementia worldwide and has been consistently associated with the
loss of basal forebrain cholinergic neurons (BFCNs) leading to impaired
cholinergic neurotransmission, aberrant synaptic function, and altered
structural lipid metabolism. In this sense, membrane phospholipids
(PLs) can be used for de novo synthesis of choline (Ch) for the further
obtaining of acetylcholine (ACh) when its availability is compromised.
Specific lipid species involved in the metabolism of Ch have been
identified as possible biomarkers of phenoconversion to AD. Using
a rat model of BFCN lesion, we have evaluated the lipid composition
and muscarinic signaling in brain areas related to cognitive processes.
The loss of BFCN resulted in alterations of varied lipid species related
to Ch metabolism at nucleus basalis magnocellularis (NMB) and cortical
projection areas. The activity of muscarinic receptors (mAChR) was
decreased in the NMB and increased in the hippocampus according to
the subcellular distribution of M1/M2 mAChR
which could explain the learning and memory impairment reported in
this AD rat model. These results suggest that the modulation of specific
lipid metabolic routes could represent an alternative therapeutic
strategy to potentiate cholinergic neurotransmission and preserve
cell membrane integrity in AD.
Collapse
Affiliation(s)
- Alberto Llorente-Ovejero
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain
| | - Jonatan Martínez-Gardeazabal
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain
| | - Marta Moreno-Rodríguez
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain
| | - Laura Lombardero
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain
| | - Estíbaliz González de San Román
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain
| | - Iván Manuel
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain
- Neurodegenerative Diseases, BioCruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - María Teresa Giralt
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain
| | - Rafael Rodríguez-Puertas
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain
- Neurodegenerative Diseases, BioCruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
| |
Collapse
|
30
|
Baumel BS, Doraiswamy PM, Sabbagh M, Wurtman R. Potential Neuroregenerative and Neuroprotective Effects of Uridine/Choline-Enriched Multinutrient Dietary Intervention for Mild Cognitive Impairment: A Narrative Review. Neurol Ther 2021; 10:43-60. [PMID: 33368017 PMCID: PMC8139993 DOI: 10.1007/s40120-020-00227-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 12/02/2020] [Indexed: 01/21/2023] Open
Abstract
In mild cognitive impairment (MCI) due to Alzheimer disease (AD), also known as prodromal AD, there is evidence for a pathologic shortage of uridine, choline, and docosahexaenoic acid [DHA]), which are key nutrients needed by the brain. Preclinical and clinical evidence shows the importance of nutrient bioavailability to support the development and maintenance of brain structure and function in MCI and AD. Availability of key nutrients is limited in MCI, creating a distinct nutritional need for uridine, choline, and DHA. Evidence suggests that metabolic derangements associated with ageing and disease-related pathology can affect the body's ability to generate and utilize nutrients. This is reflected in lower levels of nutrients measured in the plasma and brains of individuals with MCI and AD dementia, and progressive loss of cognitive performance. The uridine shortage cannot be corrected by normal diet, making uridine a conditionally essential nutrient in affected individuals. It is also challenging to correct the choline shortfall through diet alone, because brain uptake from the plasma significantly decreases with ageing. There is no strong evidence to support the use of single-agent supplements in the management of MCI due to AD. As uridine and choline work synergistically with DHA to increase phosphatidylcholine formation, there is a compelling rationale to combine these nutrients. A multinutrient enriched with uridine, choline, and DHA developed to support brain function has been evaluated in randomized controlled trials covering a spectrum of dementia from MCI to moderate AD. A randomized controlled trial in subjects with prodromal AD showed that multinutrient intervention slowed brain atrophy and improved some measures of cognition. Based on the available clinical evidence, nutritional intervention should be considered as a part of the approach to the management of individuals with MCI due to AD, including adherence to a healthy, balanced diet, and consideration of evidence-based multinutrient supplements.
Collapse
Affiliation(s)
- Barry S Baumel
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, USA.
| | - P Murali Doraiswamy
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Marwan Sabbagh
- Lou Ruvo Center for Brain Health, Cleveland Clinic, Las Vegas, NV, USA
| | - Richard Wurtman
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
31
|
Su H, Rustam YH, Masters CL, Makalic E, McLean CA, Hill AF, Barnham KJ, Reid GE, Vella LJ. Characterization of brain-derived extracellular vesicle lipids in Alzheimer's disease. J Extracell Vesicles 2021; 10:e12089. [PMID: 34012516 PMCID: PMC8111496 DOI: 10.1002/jev2.12089] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 03/29/2021] [Accepted: 04/13/2021] [Indexed: 12/13/2022] Open
Abstract
Lipid dyshomeostasis is associated with the most common form of dementia, Alzheimer's disease (AD). Substantial progress has been made in identifying positron emission tomography and cerebrospinal fluid biomarkers for AD, but they have limited use as front-line diagnostic tools. Extracellular vesicles (EVs) are released by all cells and contain a subset of their parental cell composition, including lipids. EVs are released from the brain into the periphery, providing a potential source of tissue and disease specific lipid biomarkers. However, the EV lipidome of the central nervous system is currently unknown and the potential of brain-derived EVs (BDEVs) to inform on lipid dyshomeostasis in AD remains unclear. The aim of this study was to reveal the lipid composition of BDEVs in human frontal cortex, and to determine whether BDEVs have an altered lipid profile in AD. Using semi-quantitative mass spectrometry, we describe the BDEV lipidome, covering four lipid categories, 17 lipid classes and 692 lipid molecules. BDEVs were enriched in glycerophosphoserine (PS) lipids, a characteristic of small EVs. Here we further report that BDEVs are enriched in ether-containing PS lipids, a finding that further establishes ether lipids as a feature of EVs. BDEVs in the AD frontal cortex offered improved detection of dysregulated lipids in AD over global lipid profiling of this brain region. AD BDEVs had significantly altered glycerophospholipid and sphingolipid levels, specifically increased plasmalogen glycerophosphoethanolamine and decreased polyunsaturated fatty acyl containing lipids, and altered amide-linked acyl chain content in sphingomyelin and ceramide lipids relative to CTL. The most prominent alteration was a two-fold decrease in lipid species containing anti-inflammatory/pro-resolving docosahexaenoic acid. The in-depth lipidome analysis provided in this study highlights the advantage of EVs over more complex tissues for improved detection of dysregulated lipids that may serve as potential biomarkers in the periphery.
Collapse
Affiliation(s)
- Huaqi Su
- The Florey Institute of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
- Department of Biochemistry and PharmacologyThe University of MelbourneParkvilleVictoriaAustralia
| | - Yepy H. Rustam
- Department of Biochemistry and PharmacologyThe University of MelbourneParkvilleVictoriaAustralia
| | - Colin L. Masters
- The Florey Institute of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
| | - Enes Makalic
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global HealthThe University of MelbourneParkvilleVictoriaAustralia
| | - Catriona A. McLean
- The Florey Institute of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
| | - Andrew F. Hill
- Department of Biochemistry and GeneticsLa Trobe Institute for Molecular Science, La Trobe UniversityBundooraVictoriaAustralia
| | - Kevin J. Barnham
- The Florey Institute of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
| | - Gavin E. Reid
- Department of Biochemistry and PharmacologyThe University of MelbourneParkvilleVictoriaAustralia
- School of Chemistry, Bio21 Molecular Science and Biotechnology InstituteThe University of MelbourneParkvilleVictoriaAustralia
| | - Laura J. Vella
- The Florey Institute of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
- Department of Surgery, The Royal Melbourne HospitalThe University of MelbourneParkvilleVictoriaAustralia
| |
Collapse
|
32
|
Lefèvre-Arbogast S, Hejblum BP, Helmer C, Klose C, Manach C, Low DY, Urpi-Sarda M, Andres-Lacueva C, González-Domínguez R, Aigner L, Altendorfer B, Lucassen PJ, Ruigrok SR, De Lucia C, Du Preez A, Proust-Lima C, Thuret S, Korosi A, Samieri C. Early signature in the blood lipidome associated with subsequent cognitive decline in the elderly: A case-control analysis nested within the Three-City cohort study. EBioMedicine 2021; 64:103216. [PMID: 33508744 PMCID: PMC7841305 DOI: 10.1016/j.ebiom.2021.103216] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 01/04/2021] [Accepted: 01/05/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Brain lipid metabolism appears critical for cognitive aging, but whether alterations in the lipidome relate to cognitive decline remains unclear at the system level. METHODS We studied participants from the Three-City study, a multicentric cohort of older persons, free of dementia at time of blood sampling, and who provided repeated measures of cognition over 12 subsequent years. We measured 189 serum lipids from 13 lipid classes using shotgun lipidomics in a case-control sample on cognitive decline (matched on age, sex and level of education) nested within the Bordeaux study center (discovery, n = 418). Associations with cognitive decline were investigated using bootstrapped penalized regression, and tested for validation in the Dijon study center (validation, n = 314). FINDINGS Among 17 lipids identified in the discovery stage, lower levels of the triglyceride TAG50:5, and of four membrane lipids (sphingomyelin SM40:2,2, phosphatidylethanolamine PE38:5(18:1/20:4), ether-phosphatidylethanolamine PEO34:3(16:1/18:2), and ether-phosphatidylcholine PCO34:1(16:1/18:0)), and higher levels of PCO32:0(16:0/16:0), were associated with greater odds of cognitive decline, and replicated in our validation sample. INTERPRETATION These findings indicate that in the blood lipidome of non-demented older persons, a specific profile of lipids involved in membrane fluidity, myelination, and lipid rafts, is associated with subsequent cognitive decline. FUNDING The complete list of funders is available at the end of the manuscript, in the Acknowledgement section.
Collapse
Affiliation(s)
- Sophie Lefèvre-Arbogast
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, UMR 1219, 146 rue Léo-Saignat, Bordeaux 33076, France
| | - Boris P Hejblum
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, UMR 1219, 146 rue Léo-Saignat, Bordeaux 33076, France; Inria SISTM, Bordeaux Sud-Ouest, Bordeaux 33000, France
| | - Catherine Helmer
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, UMR 1219, 146 rue Léo-Saignat, Bordeaux 33076, France
| | | | - Claudine Manach
- University of Clermont Auvergne, INRA, UMR1019, Human Nutrition Unit, Clermont Ferrand 63000, France
| | - Dorrain Y Low
- University of Clermont Auvergne, INRA, UMR1019, Human Nutrition Unit, Clermont Ferrand 63000, France
| | - Mireia Urpi-Sarda
- Nutrition, Food Science and Gastronomy Department, Faculty of Pharmacy and Food Science, CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, University of Barcelona, Barcelona 08028, Spain
| | - Cristina Andres-Lacueva
- Nutrition, Food Science and Gastronomy Department, Faculty of Pharmacy and Food Science, CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, University of Barcelona, Barcelona 08028, Spain
| | - Raúl González-Domínguez
- Nutrition, Food Science and Gastronomy Department, Faculty of Pharmacy and Food Science, CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, University of Barcelona, Barcelona 08028, Spain
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg 5020, Austria
| | - Barbara Altendorfer
- Institute of Molecular Regenerative Medicine, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg 5020, Austria
| | - Paul J Lucassen
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam 1098 XH, Netherlands
| | - Silvie R Ruigrok
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam 1098 XH, Netherlands
| | - Chiara De Lucia
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, United Kingdom
| | - Andrea Du Preez
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, United Kingdom
| | - Cécile Proust-Lima
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, UMR 1219, 146 rue Léo-Saignat, Bordeaux 33076, France
| | - Sandrine Thuret
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, United Kingdom; Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Aniko Korosi
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam 1098 XH, Netherlands
| | - Cécilia Samieri
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, UMR 1219, 146 rue Léo-Saignat, Bordeaux 33076, France.
| |
Collapse
|
33
|
Natarajan K, Ullgren A, Khoshnood B, Johansson C, Laffita-Mesa JM, Pannee J, Zetterberg H, Blennow K, Graff C. Plasma metabolomics of presymptomatic PSEN1-H163Y mutation carriers: a pilot study. Ann Clin Transl Neurol 2021; 8:579-591. [PMID: 33476461 PMCID: PMC7951103 DOI: 10.1002/acn3.51296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 12/04/2020] [Accepted: 12/10/2020] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND AND OBJECTIVE PSEN1-H163Y carriers, at the presymptomatic stage, have reduced 18 FDG-PET binding in the cerebrum of the brain (Scholl et al., Neurobiol Aging 32:1388-1399, 2011). This could imply dysfunctional energy metabolism in the brain. In this study, plasma of presymptomatic PSEN1 mutation carriers was analyzed to understand associated metabolic changes. METHODS We analyzed plasma from noncarriers (NC, n = 8) and presymptomatic PSEN1-H163Y mutation carriers (MC, n = 6) via untargeted metabolomics using gas and liquid chromatography coupled with mass spectrometry, which identified 1199 metabolites. All the metabolites were compared between MC and NC using univariate analysis, as well as correlated with the ratio of Aβ1-42/A β 1-40 , using Spearman's correlation. Altered metabolites were subjected to Ingenuity Pathway Analysis (IPA). RESULTS Based on principal component analysis the plasma metabolite profiles were divided into dataset A and dataset B. In dataset A, when comparing between presymptomatic MC and NC, the levels of 79 different metabolites were altered. Out of 79, only 14 were annotated metabolites. In dataset B, 37 metabolites were significantly altered between presymptomatic MC and NC and nine metabolites were annotated. In both datasets, annotated metabolites represent amino acids, fatty acyls, bile acids, hexoses, purine nucleosides, carboxylic acids, and glycerophosphatidylcholine species. 1-docosapentaenoyl-GPC was positively correlated, uric acid and glucose were negatively correlated with the ratio of plasma Aβ1-42 /Aβ1-40 (P < 0.05). INTERPRETATION This study finds dysregulated metabolite classes, which are changed before the disease symptom onset. Also, it provides an opportunity to compare with sporadic Alzheimer's Disease. Observed findings in this study need to be validated in a larger and independent Familial Alzheimer's Disease (FAD) cohort.
Collapse
Affiliation(s)
- Karthick Natarajan
- Division for Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Unit for Hereditary Dementias, Theme Aging, QA12, Karolinska University Hospital-Solna, Stockholm, Sweden
| | - Abbe Ullgren
- Division for Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Unit for Hereditary Dementias, Theme Aging, QA12, Karolinska University Hospital-Solna, Stockholm, Sweden
| | - Behzad Khoshnood
- Division for Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Unit for Hereditary Dementias, Theme Aging, QA12, Karolinska University Hospital-Solna, Stockholm, Sweden
| | - Charlotte Johansson
- Division for Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Unit for Hereditary Dementias, Theme Aging, QA12, Karolinska University Hospital-Solna, Stockholm, Sweden
| | - José M Laffita-Mesa
- Division for Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Unit for Hereditary Dementias, Theme Aging, QA12, Karolinska University Hospital-Solna, Stockholm, Sweden
| | - Josef Pannee
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Department of Molecular Neuroscience, UCL Institute of Neurology, London, WC1N 3BG, England
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Caroline Graff
- Division for Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Unit for Hereditary Dementias, Theme Aging, QA12, Karolinska University Hospital-Solna, Stockholm, Sweden
| |
Collapse
|
34
|
Nangare S, Patil P. Nanoarchitectured Bioconjugates and Bioreceptors Mediated Surface Plasmon Resonance Biosensor for In Vitro Diagnosis of Alzheimer’s Disease: Development and Future Prospects. Crit Rev Anal Chem 2021; 52:1139-1169. [DOI: 10.1080/10408347.2020.1864716] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Sopan Nangare
- Department of Pharmaceutical Chemistry, H. R. Patel Institute of Pharmaceutical Education and Research, Shirpur, India
| | - Pravin Patil
- Department of Pharmaceutical Chemistry, H. R. Patel Institute of Pharmaceutical Education and Research, Shirpur, India
| |
Collapse
|
35
|
Fote G, Wu J, Mapstone M, Macciardi F, Fiandaca MS, Federoff HJ. Plasma Sphingomyelins in Late-Onset Alzheimer's Disease. J Alzheimers Dis 2021; 83:1161-1171. [PMID: 34397408 PMCID: PMC9788856 DOI: 10.3233/jad-200871] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Altered plasma levels of sphingolipids, including sphingomyelins (SM), have been found in mouse models of Alzheimer's disease (AD) and in AD patient plasma samples. OBJECTIVE This study assesses fourteen plasma SM species in a late-onset AD (LOAD) patient cohort (n = 138). METHODS Specimens from control, preclinical, and symptomatic subjects were analyzed using targeted mass-spectrometry-based metabolomic methods. RESULTS Total plasma SM levels were not significantly affected by age or cognitive status. However, one metabolite that has been elevated in manifest AD in several recent studies, SM OHC14:1, was reduced significantly in pre-clinical AD and MCI relative to normal controls. CONCLUSION We recommend additional comprehensive plasma lipidomics in experimental and clinical biospecimens related to LOAD that might advance the utility of plasma sphingomyelin levels in molecular phenotyping and interpretations of pathobiological mechanisms.
Collapse
Affiliation(s)
- Gianna Fote
- UC Irvine Department of Biological Chemistry, Irvine, CA, USA,Correspondence to: Gianna M. Fote, UC Irvine School of Medicine, 385 S. Manchester Ave, Unit 2096, Orange, CA 92686, USA. Tel.: +1 310 924 4415; . and Howard Federoff, MD, PhD, Distinguished Professor, Neurology, UC Irvine School of Medicine, Orange, CA 92686, USA. Tel.: +1 240 281 2598;
| | - Jie Wu
- UC Irvine Department of Biological Chemistry, Irvine, CA, USA,UC Irvine Center for Complex Biological Systems, Irvine, CA, USA
| | | | - Fabio Macciardi
- Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, CA, USA
| | - Massimo S. Fiandaca
- Translational Laboratory and Biorepository, Department of Neurology, University of California Irvine School of Medicine, Irvine, CA, USA,Department of Neurological Surgery, University of California Irvine School of Medicine, Irvine, CA, USA,Department of Anatomy & Neurobiology, University of California Irvine School of Medicine, Irvine, CA, USA
| | - Howard J. Federoff
- UC Irvine Department of Neurology, Irvine, CA, USA,Correspondence to: Gianna M. Fote, UC Irvine School of Medicine, 385 S. Manchester Ave, Unit 2096, Orange, CA 92686, USA. Tel.: +1 310 924 4415; . and Howard Federoff, MD, PhD, Distinguished Professor, Neurology, UC Irvine School of Medicine, Orange, CA 92686, USA. Tel.: +1 240 281 2598;
| |
Collapse
|
36
|
Sagar R, Pathak P, Pandur B, Kim SJ, Li J, Mahairaki V. Biomarkers and Precision Medicine in Alzheimer’s Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1339:403-408. [DOI: 10.1007/978-3-030-78787-5_50] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
37
|
Abstract
Finding early disease markers using non-invasive and widely available methods is essential to develop a successful therapy for Alzheimer’s Disease. Few studies to date have examined urine, the most readily available biofluid. Here we report the largest study to date using comprehensive metabolic phenotyping platforms (NMR spectroscopy and UHPLC-MS) to probe the urinary metabolome in-depth in people with Alzheimer’s Disease and Mild Cognitive Impairment. Feature reduction was performed using metabolomic Quantitative Trait Loci, resulting in the list of metabolites associated with the genetic variants. This approach helps accuracy in identification of disease states and provides a route to a plausible mechanistic link to pathological processes. Using these mQTLs we built a Random Forests model, which not only correctly discriminates between people with Alzheimer’s Disease and age-matched controls, but also between individuals with Mild Cognitive Impairment who were later diagnosed with Alzheimer’s Disease and those who were not. Further annotation of top-ranking metabolic features nominated by the trained model revealed the involvement of cholesterol-derived metabolites and small-molecules that were linked to Alzheimer’s pathology in previous studies.
Collapse
|
38
|
Manzine PR, Vatanabe IP, Peron R, Grigoli MM, Pedroso RV, Nascimento CMC, Cominetti MR. Blood-based Biomarkers of Alzheimer's Disease: The Long and Winding Road. Curr Pharm Des 2020; 26:1300-1315. [PMID: 31942855 DOI: 10.2174/1381612826666200114105515] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 11/27/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Blood-based biomarkers can be very useful in formulating new diagnostic and treatment proposals in the field of dementia, especially in Alzheimer's disease (AD). However, due to the influence of several factors on the reproducibility and reliability of these markers, their clinical use is still very uncertain. Thus, up-to-date knowledge about the main blood biomarkers that are currently being studied is extremely important in order to discover clinically useful and applicable tools, which could also be used as novel pharmacological strategies for the AD treatment. METHODS A narrative review was performed based on the current candidates of blood-based biomarkers for AD to show the main results from different studies, focusing on their clinical applicability and association with AD pathogenesis. OBJECTIVE The aim of this paper was to carry out a literature review on the major blood-based biomarkers for AD, connecting them with the pathophysiology of the disease. RESULTS Recent advances in the search of blood-based AD biomarkers were summarized in this review. The biomarkers were classified according to the topics related to the main hallmarks of the disease such as inflammation, amyloid, and tau deposition, synaptic degeneration and oxidative stress. Moreover, molecules involved in the regulation of proteins related to these hallmarks were described, such as non-coding RNAs, neurotrophins, growth factors and metabolites. Cells or cellular components with the potential to be considered as blood-based AD biomarkers were described in a separate topic. CONCLUSION A series of limitations undermine new discoveries on blood-based AD biomarkers. The lack of reproducibility of findings due to the small size and heterogeneity of the study population, different analytical methods and other assay conditions make longitudinal studies necessary in this field to validate these structures, especially when considering a clinical evaluation that includes a broad panel of these potential and promising blood-based biomarkers.
Collapse
Affiliation(s)
- Patricia R Manzine
- Department of Gerontology, Federal University of Sao Carlos, Rod. Washington Luis, Km 235, Monjolinho, CEP 13565-905, Sao Carlos, SP, Brazil
| | - Izabela P Vatanabe
- Department of Gerontology, Federal University of Sao Carlos, Rod. Washington Luis, Km 235, Monjolinho, CEP 13565-905, Sao Carlos, SP, Brazil
| | - Rafaela Peron
- Department of Gerontology, Federal University of Sao Carlos, Rod. Washington Luis, Km 235, Monjolinho, CEP 13565-905, Sao Carlos, SP, Brazil
| | - Marina M Grigoli
- Department of Gerontology, Federal University of Sao Carlos, Rod. Washington Luis, Km 235, Monjolinho, CEP 13565-905, Sao Carlos, SP, Brazil
| | - Renata V Pedroso
- Department of Gerontology, Federal University of Sao Carlos, Rod. Washington Luis, Km 235, Monjolinho, CEP 13565-905, Sao Carlos, SP, Brazil
| | - Carla M C Nascimento
- Department of Gerontology, Federal University of Sao Carlos, Rod. Washington Luis, Km 235, Monjolinho, CEP 13565-905, Sao Carlos, SP, Brazil
| | - Marcia R Cominetti
- Department of Gerontology, Federal University of Sao Carlos, Rod. Washington Luis, Km 235, Monjolinho, CEP 13565-905, Sao Carlos, SP, Brazil
| |
Collapse
|
39
|
Huynh K, Lim WLF, Giles C, Jayawardana KS, Salim A, Mellett NA, Smith AAT, Olshansky G, Drew BG, Chatterjee P, Martins I, Laws SM, Bush AI, Rowe CC, Villemagne VL, Ames D, Masters CL, Arnold M, Nho K, Saykin AJ, Baillie R, Han X, Kaddurah-Daouk R, Martins RN, Meikle PJ. Concordant peripheral lipidome signatures in two large clinical studies of Alzheimer's disease. Nat Commun 2020; 11:5698. [PMID: 33173055 PMCID: PMC7655942 DOI: 10.1038/s41467-020-19473-7] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 10/15/2020] [Indexed: 11/22/2022] Open
Abstract
Changes to lipid metabolism are tightly associated with the onset and pathology of Alzheimer's disease (AD). Lipids are complex molecules comprising many isomeric and isobaric species, necessitating detailed analysis to enable interpretation of biological significance. Our expanded targeted lipidomics platform (569 species across 32 classes) allows for detailed lipid separation and characterisation. In this study we examined peripheral samples of two cohorts (AIBL, n = 1112 and ADNI, n = 800). We are able to identify concordant peripheral signatures associated with prevalent AD arising from lipid pathways including; ether lipids, sphingolipids (notably GM3 gangliosides) and lipid classes previously associated with cardiometabolic disease (phosphatidylethanolamine and triglycerides). We subsequently identified similar lipid signatures in both cohorts with future disease. Lastly, we developed multivariate lipid models that improved classification and prediction. Our results provide a holistic view between the lipidome and AD using a comprehensive approach, providing targets for further mechanistic investigation.
Collapse
Affiliation(s)
- Kevin Huynh
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Monash University, Melbourne, VIC, 3800, Australia
| | - Wei Ling Florence Lim
- School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia
- Cooperative research Centre (CRC) for Mental Health, Sydney, NSW, Australia
| | - Corey Giles
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | | | - Agus Salim
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Mathematics and Statistics, La Trobe University, Melbourne, VIC, Australia
- Melbourne School of Global and Population Health, The University of Melbourne, Melbourne, VIC, 3010, Australia
- School of Mathematics and Statistics, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | | | | | | | - Brian G Drew
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Monash University, Melbourne, VIC, 3800, Australia
| | - Pratishtha Chatterjee
- School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia
- Department of Biomedical Sciences, Macquarie University, Sydney, NSW, Australia
- KaRa Institute of Neurological Disease, Sydney, NSW, Australia
| | - Ian Martins
- School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia
- Cooperative research Centre (CRC) for Mental Health, Sydney, NSW, Australia
| | - Simon M Laws
- School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia
- Collaborative Genomics Group, School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia
- Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| | - Ashley I Bush
- The Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Christopher C Rowe
- The Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
- Department of Nuclear Medicine and Centre for PET, Austin Health, Heidelberg, VIC, Australia
| | - Victor L Villemagne
- Department of Nuclear Medicine and Centre for PET, Austin Health, Heidelberg, VIC, Australia
- Department of Medicine, Austin Health, The University of Melbourne, Heidelberg, VIC, Australia
| | - David Ames
- National Ageing Research Institute, Parkville, VIC, 3050, Australia
| | - Colin L Masters
- The Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Matthias Arnold
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Kwangsik Nho
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA.
- Duke Institute of Brain Sciences, Duke University, Durham, NC, USA.
- Department of Medicine, Duke University, Durham, NC, USA.
| | - Ralph N Martins
- School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia.
- Cooperative research Centre (CRC) for Mental Health, Sydney, NSW, Australia.
- Department of Biomedical Sciences, Macquarie University, Sydney, NSW, Australia.
- KaRa Institute of Neurological Disease, Sydney, NSW, Australia.
- School of Psychiatry and Clinical Neurosciences, The University of Western Australia, Perth, WA, Australia.
- Australian Alzheimer's Research Foundation, Nedlands, WA, Australia.
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.
- Monash University, Melbourne, VIC, 3800, Australia.
| |
Collapse
|
40
|
Yilmaz A, Ustun I, Ugur Z, Akyol S, Hu WT, Fiandaca MS, Mapstone M, Federoff H, Maddens M, Graham SF. A Community-Based Study Identifying Metabolic Biomarkers of Mild Cognitive Impairment and Alzheimer's Disease Using Artificial Intelligence and Machine Learning. J Alzheimers Dis 2020; 78:1381-1392. [PMID: 33164929 DOI: 10.3233/jad-200305] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BACKGROUND Currently, there is no objective, clinically available tool for the accurate diagnosis of Alzheimer's disease (AD). There is a pressing need for a novel, minimally invasive, cost friendly, and easily accessible tool to diagnose AD, assess disease severity, and prognosticate course. Metabolomics is a promising tool for discovery of new, biologically, and clinically relevant biomarkers for AD detection and classification. OBJECTIVE Utilizing artificial intelligence and machine learning, we aim to assess whether a panel of metabolites as detected in plasma can be used as an objective and clinically feasible tool for the diagnosis of mild cognitive impairment (MCI) and AD. METHODS Using a community-based sample cohort acquired from different sites across the US, we adopted an approach combining Proton Nuclear Magnetic Resonance Spectroscopy (1H NMR), Liquid Chromatography coupled with Mass Spectrometry (LC-MS) and various machine learning statistical approaches to identify a biomarker panel capable of identifying those patients with AD and MCI from healthy controls. RESULTS Of the 212 measured metabolites, 5 were identified as optimal to discriminate between controls, and individuals with MCI or AD. Our models performed with AUC values in the range of 0.72-0.76, with the sensitivity and specificity values ranging from 0.75-0.85 and 0.69-0.81, respectively. Univariate and pathway analysis identified lipid metabolism as the most perturbed biochemical pathway in MCI and AD. CONCLUSION A comprehensive method of acquiring metabolomics data, coupled with machine learning techniques, has identified a strong panel of diagnostic biomarkers capable of identifying individuals with MCI and AD. Further, our data confirm what other groups have reported, that lipid metabolism is significantly perturbed in those individuals suffering with dementia. This work may provide additional insight into AD pathogenesis and encourage more in-depth analysis of the AD lipidome.
Collapse
Affiliation(s)
- Ali Yilmaz
- Department of Obstetrics and Gynecology, Department of Internal Medicine, Oakland University-William Beaumont School of Medicine, Rochester, MI, USA.,Metabolomics Division, Beaumont Research Institute, Royal Oak, MI USA
| | - Ilyas Ustun
- Wayne State University, Civil and Environmental Engineering, Detroit, MI, USA
| | - Zafer Ugur
- Metabolomics Division, Beaumont Research Institute, Royal Oak, MI USA
| | - Sumeyya Akyol
- Metabolomics Division, Beaumont Research Institute, Royal Oak, MI USA
| | - William T Hu
- Department of Neurology, Emory University, Atlanta, GA, USA
| | - Massimo S Fiandaca
- Department of Neurology, University of California Irvine, Irvine, CA, USA
| | - Mark Mapstone
- Department of Neurology, University of California Irvine, Irvine, CA, USA
| | - Howard Federoff
- Department of Neurology, University of California Irvine, Irvine, CA, USA
| | - Michael Maddens
- Department of Obstetrics and Gynecology, Department of Internal Medicine, Oakland University-William Beaumont School of Medicine, Rochester, MI, USA
| | - Stewart F Graham
- Department of Obstetrics and Gynecology, Department of Internal Medicine, Oakland University-William Beaumont School of Medicine, Rochester, MI, USA.,Metabolomics Division, Beaumont Research Institute, Royal Oak, MI USA
| |
Collapse
|
41
|
Handen BL, Lott IT, Christian BT, Schupf N, OBryant S, Mapstone M, Fagan AM, Lee JH, Tudorascu D, Wang M, Head E, Klunk W, Ances B, Lai F, Zaman S, Krinsky‐McHale S, Brickman AM, Rosas HD, Cohen A, Andrews H, Hartley S, Silverman W. The Alzheimer's Biomarker Consortium-Down Syndrome: Rationale and methodology. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2020; 12:e12065. [PMID: 32775597 PMCID: PMC7396809 DOI: 10.1002/dad2.12065] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/05/2020] [Accepted: 06/08/2020] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Adults with Down syndrome (DS) are at exceptionally high risk for Alzheimer's disease (AD), with virtually all individuals developing key neuropathological features by age 40. Identifying biomarkers of AD progression in DS can provide valuable insights into pathogenesis and suggest targets for disease modifying treatments. METHODS We describe the development of a multi-center, longitudinal study of biomarkers of AD in DS. The protocol includes longitudinal examination of clinical, cognitive, blood and cerebrospinal fluid-based biomarkers, magnetic resonance imaging and positron emission tomography measures (at 16-month intervals), as well as genetic modifiers of AD risk and progression. RESULTS Approximately 400 individuals will be enrolled in the study (more than 370 to date). The methodological approach from the administrative, clinical, neuroimaging, omics, neuropathology, and statistical cores is provided. DISCUSSION This represents the largest U.S.-based, multi-site, biomarker initiative of AD in DS. Findings can inform other multidisciplinary networks studying AD in the general population.
Collapse
Affiliation(s)
- Benjamin L. Handen
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Ira T. Lott
- IrvineSchool of MedicineDepartment of PediatricsUniversity of CaliforniaOrangeCaliforniaUSA
| | | | - Nicole Schupf
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Sid OBryant
- Department of Pharmacology and Neuroscience and Institute for Translational ResearchUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Mark Mapstone
- IrvineDepartment of NeurologyUniversity of CaliforniaIrvineCaliforniaUSA
| | - Anne M. Fagan
- Department of NeurologyWashington University in St. LouisSt LouisMissouriUSA
| | - Joseph H. Lee
- Department of Neurology Center, Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityCollege of Physicians and SurgeonsNew YorkNew YorkUSA
| | - Dana Tudorascu
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Mei‐Cheng Wang
- Johns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | - Elizabeth Head
- IrvineDepartment of PathologyUniversity of CaliforniaIrvineCaliforniaUSA
| | - William Klunk
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Beau Ances
- Washingston University School of Medicine in St. LouisSt. LouisMissouriUSA
| | - Florence Lai
- Massachusetts General HospitalDepartment of NeurologyHarvard Medical SchoolCharlestownMassachusettsUSA
| | - Shahid Zaman
- School of Clinical MedicineDepartment of PsychiatryUniversity of CambridgeCambridgeUK
| | - Sharon Krinsky‐McHale
- Department of PsychologyNYS Institute for Basic Research in Developmental DisabilitiesStaten IslandNew YorkUSA
| | - Adam M. Brickman
- Department of Neurology Center, Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityCollege of Physicians and SurgeonsNew YorkNew YorkUSA
| | - H. Diana Rosas
- Massachusetts General HospitalDepartments of Neurology and RadiologyHarvard Medical SchoolCharlestownMassachusettsUSA
| | - Annie Cohen
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Howard Andrews
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Sigan Hartley
- Waisman CenterUniversity of Wisconsin MadisonMadisonWisconsinUSA
| | - Wayne Silverman
- IrvineSchool of MedicineDepartment of PediatricsUniversity of CaliforniaOrangeCaliforniaUSA
| | | |
Collapse
|
42
|
Chatterjee P, Cheong Y, Bhatnagar A, Goozee K, Wu Y, McKay M, Martins IJ, Lim WLF, Pedrini S, Tegg M, Villemagne VL, Asih PR, Dave P, Shah TM, Dias CB, Fuller SJ, Hillebrandt H, Gupta S, Hone E, Taddei K, Zetterberg H, Blennow K, Sohrabi HR, Martins RN. Plasma metabolites associated with biomarker evidence of neurodegeneration in cognitively normal older adults. J Neurochem 2020; 159:389-402. [DOI: 10.1111/jnc.15128] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/29/2020] [Accepted: 07/07/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Pratishtha Chatterjee
- Department of Biomedical Sciences Macquarie University North Ryde NSW Australia
- School of Medical and Health Sciences Edith Cowan University, Patricia Sarich Neuroscience Research Institute Nedlands WA Australia
| | - Yeo‐Jin Cheong
- Department of Biomedical Sciences Macquarie University North Ryde NSW Australia
| | - Atul Bhatnagar
- Department of Molecular Sciences Macquarie University North Ryde NSW Australia
| | - Kathryn Goozee
- Department of Biomedical Sciences Macquarie University North Ryde NSW Australia
- KaRa Institute of Neurological Disease Sydney NSW Australia
- Clinical Research Department Anglicare, Sydney NSW Australia
- School of Psychiatry and Clinical Neurosciences University of Western Australia, Crawley WA Australia
| | - Yunqi Wu
- Department of Molecular Sciences Macquarie University North Ryde NSW Australia
| | - Matthew McKay
- Department of Molecular Sciences Macquarie University North Ryde NSW Australia
| | - Ian J. Martins
- School of Medical and Health Sciences Edith Cowan University, Patricia Sarich Neuroscience Research Institute Nedlands WA Australia
| | - Wei L. F. Lim
- School of Medical and Health Sciences Edith Cowan University, Patricia Sarich Neuroscience Research Institute Nedlands WA Australia
| | - Steve Pedrini
- School of Medical and Health Sciences Edith Cowan University, Patricia Sarich Neuroscience Research Institute Nedlands WA Australia
| | - Michelle Tegg
- School of Medical and Health Sciences Edith Cowan University, Patricia Sarich Neuroscience Research Institute Nedlands WA Australia
| | - Victor L. Villemagne
- The Florey Institute of Neuroscience and Mental Health University of Melbourne VA Australia
| | - Prita R. Asih
- School of Medical and Health Sciences Edith Cowan University, Patricia Sarich Neuroscience Research Institute Nedlands WA Australia
| | - Preeti Dave
- Department of Biomedical Sciences Macquarie University North Ryde NSW Australia
- Clinical Research Department Anglicare, Sydney NSW Australia
| | - Tejal M. Shah
- Department of Biomedical Sciences Macquarie University North Ryde NSW Australia
- School of Medical and Health Sciences Edith Cowan University, Patricia Sarich Neuroscience Research Institute Nedlands WA Australia
- Australian Alzheimer’s Research Foundation Nedlands WA Australia
| | - Cintia B. Dias
- Department of Biomedical Sciences Macquarie University North Ryde NSW Australia
| | - Stephanie J. Fuller
- Department of Biomedical Sciences Macquarie University North Ryde NSW Australia
| | - Heidi Hillebrandt
- Department of Biomedical Sciences Macquarie University North Ryde NSW Australia
| | - Sunil Gupta
- Department of Biomedical Sciences Macquarie University North Ryde NSW Australia
| | - Eugene Hone
- School of Medical and Health Sciences Edith Cowan University, Patricia Sarich Neuroscience Research Institute Nedlands WA Australia
| | - Kevin Taddei
- School of Medical and Health Sciences Edith Cowan University, Patricia Sarich Neuroscience Research Institute Nedlands WA Australia
- Australian Alzheimer’s Research Foundation Nedlands WA Australia
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry Institute of Neuroscience and Physiology University of Gothenburg Mölndal Sweden
- Clinical Neurochemistry Laboratory Sahlgrenska University Hospital Mölndal Sweden
- Department of Neurodegenerative Disease UCL Institute of NeurologyQueen Square London UK
- UK Dementia Research Institute at UCL London UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry Institute of Neuroscience and Physiology University of Gothenburg Mölndal Sweden
- Clinical Neurochemistry Laboratory Sahlgrenska University Hospital Mölndal Sweden
| | - Hamid R. Sohrabi
- Department of Biomedical Sciences Macquarie University North Ryde NSW Australia
- School of Medical and Health Sciences Edith Cowan University, Patricia Sarich Neuroscience Research Institute Nedlands WA Australia
- Australian Alzheimer’s Research Foundation Nedlands WA Australia
- Centre for Healthy Ageing School of Psychology and Exercise Science College of Science Health, Engineering and Education Murdoch University Murdoch WA Australia
| | - Ralph N. Martins
- Department of Biomedical Sciences Macquarie University North Ryde NSW Australia
- School of Medical and Health Sciences Edith Cowan University, Patricia Sarich Neuroscience Research Institute Nedlands WA Australia
- KaRa Institute of Neurological Disease Sydney NSW Australia
- School of Psychiatry and Clinical Neurosciences University of Western Australia, Crawley WA Australia
- Australian Alzheimer’s Research Foundation Nedlands WA Australia
| |
Collapse
|
43
|
Costa AC, Joaquim HPG, Forlenza OV, Gattaz WF, Talib LL. Three plasma metabolites in elderly patients differentiate mild cognitive impairment and Alzheimer's disease: a pilot study. Eur Arch Psychiatry Clin Neurosci 2020; 270:483-488. [PMID: 31218445 DOI: 10.1007/s00406-019-01034-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 06/15/2019] [Indexed: 12/11/2022]
Abstract
The metabolomic profile of patients with Alzheimer's disease (AD) and mild cognitive impairment (MCI) may suggest potential diagnostic biomarkers and provide information on the pathophysiology of dementia. Our aim was to quantify plasmatic metabolites of AD patients, MCI and controls. We investigated the metabolomic profile-using the AbsoluteIDQ®p180 assay-of 79 older adults with primary cognitive impairment (34 AD and 20 MCI) and 25 healthy elders (controls). A cluster analysis revealed that a combination C12-DC, C12 and PCaaC26:0 could differentiate the patients according to diagnostic. Future studies should combine metabolomic profiles with other biomarkers to identify diagnostic groups.
Collapse
Affiliation(s)
- Alana C Costa
- Laboratório de Neurociências (LIM-27), Departamento e Instituto de Psiquiatria, Faculdade de Medicina da Universidade de São Paulo, Rua Dr. Ovídio Pires de Campos, 785, 3º andar, Sao Paulo, 05403-010, Brazil.,Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBioN), Conselho Nacional de Desenvolvimento Cientifico e Tecnológico, Sao Paulo, Brazil
| | - Helena P G Joaquim
- Laboratório de Neurociências (LIM-27), Departamento e Instituto de Psiquiatria, Faculdade de Medicina da Universidade de São Paulo, Rua Dr. Ovídio Pires de Campos, 785, 3º andar, Sao Paulo, 05403-010, Brazil.,Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBioN), Conselho Nacional de Desenvolvimento Cientifico e Tecnológico, Sao Paulo, Brazil
| | - Orestes V Forlenza
- Laboratório de Neurociências (LIM-27), Departamento e Instituto de Psiquiatria, Faculdade de Medicina da Universidade de São Paulo, Rua Dr. Ovídio Pires de Campos, 785, 3º andar, Sao Paulo, 05403-010, Brazil.,Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBioN), Conselho Nacional de Desenvolvimento Cientifico e Tecnológico, Sao Paulo, Brazil
| | - Wagner F Gattaz
- Laboratório de Neurociências (LIM-27), Departamento e Instituto de Psiquiatria, Faculdade de Medicina da Universidade de São Paulo, Rua Dr. Ovídio Pires de Campos, 785, 3º andar, Sao Paulo, 05403-010, Brazil.,Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBioN), Conselho Nacional de Desenvolvimento Cientifico e Tecnológico, Sao Paulo, Brazil
| | - Leda L Talib
- Laboratório de Neurociências (LIM-27), Departamento e Instituto de Psiquiatria, Faculdade de Medicina da Universidade de São Paulo, Rua Dr. Ovídio Pires de Campos, 785, 3º andar, Sao Paulo, 05403-010, Brazil. .,Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBioN), Conselho Nacional de Desenvolvimento Cientifico e Tecnológico, Sao Paulo, Brazil.
| |
Collapse
|
44
|
Graham SF, Turkoglu O, Yilmaz A, Ustun I, Ugur Z, Bjorndhal T, Han B, Mandal R, Wishart D, Bahado-Singh RO. Targeted metabolomics highlights perturbed metabolism in the brain of autism spectrum disorder sufferers. Metabolomics 2020; 16:59. [PMID: 32333121 DOI: 10.1007/s11306-020-01685-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Accepted: 04/17/2020] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Autism spectrum disorder (ASD) is a group of neurodevelopmental disorders characterized by deficiencies in social interactions and communication, combined with restricted and repetitive behavioral issues. OBJECTIVES As little is known about the etiopathophysiology of ASD and early diagnosis is relatively subjective, we aim to employ a targeted, fully quantitative metabolomics approach to biochemically profile post-mortem human brain with the overall goal of identifying metabolic pathways that may have been perturbed as a result of the disease while uncovering potential central diagnostic biomarkers. METHODS Using a combination of 1H NMR and DI/LC-MS/MS we quantitatively profiled the metabolome of the posterolateral cerebellum from post-mortem human brain harvested from people who suffered with ASD (n = 11) and compared them with age-matched controls (n = 10). RESULTS We accurately identified and quantified 203 metabolites in post-mortem brain extracts and performed a metabolite set enrichment analyses identifying 3 metabolic pathways as significantly perturbed (p < 0.05). These include Pyrimidine, Ubiquinone and Vitamin K metabolism. Further, using a variety of machine-based learning algorithms, we identified a panel of central biomarkers (9-hexadecenoylcarnitine (C16:1) and the phosphatidylcholine PC ae C36:1) capable of discriminating between ASD and controls with an AUC = 0.855 with a sensitivity and specificity equal to 0.80 and 0.818, respectively. CONCLUSION For the first time, we report the use of a multi-platform metabolomics approach to biochemically profile brain from people with ASD and report several metabolic pathways which are perturbed in the diseased brain of ASD sufferers. Further, we identified a panel of biomarkers capable of distinguishing ASD from control brains. We believe that these central biomarkers may be useful for diagnosing ASD in more accessible biomatrices.
Collapse
Affiliation(s)
- Stewart F Graham
- Oakland University-William Beaumont School of Medicine, Rochester, MI, 48309, USA.
- Research Institute, Metabolomics Division, Beaumont Health, Royal Oak, MI, 48073, USA.
| | - Onur Turkoglu
- Oakland University-William Beaumont School of Medicine, Rochester, MI, 48309, USA
| | - Ali Yilmaz
- Oakland University-William Beaumont School of Medicine, Rochester, MI, 48309, USA
- Research Institute, Metabolomics Division, Beaumont Health, Royal Oak, MI, 48073, USA
| | - Ilyas Ustun
- Wayne State University, Civil and Environmental Engineering, Detroit, MI, USA
| | - Zafer Ugur
- Oakland University-William Beaumont School of Medicine, Rochester, MI, 48309, USA
- Research Institute, Metabolomics Division, Beaumont Health, Royal Oak, MI, 48073, USA
| | - Trent Bjorndhal
- Department of Biological and Computing Sciences, University of Alberta, Edmonton, AB, Canada
| | - BeomSoo Han
- Department of Biological and Computing Sciences, University of Alberta, Edmonton, AB, Canada
| | - Rupa Mandal
- Department of Biological and Computing Sciences, University of Alberta, Edmonton, AB, Canada
| | - David Wishart
- Department of Biological and Computing Sciences, University of Alberta, Edmonton, AB, Canada
| | - Ray O Bahado-Singh
- Oakland University-William Beaumont School of Medicine, Rochester, MI, 48309, USA
| |
Collapse
|
45
|
Stathopoulou C, Nikoleri D, Bertsias G. Immunometabolism: an overview and therapeutic prospects in autoimmune diseases. Immunotherapy 2020; 11:813-829. [PMID: 31120393 DOI: 10.2217/imt-2019-0002] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Metabolism is a critical immune regulator under physiologic and pathologic conditions. Culminating evidence has disentangled the contribution of distinct metabolic pathways, namely glucolysis, pentose phosphate, fatty acid oxidation, glutaminolysis, Krebs cycle and oxidative phosphorylation, in modulating innate and adaptive immune cells based on their activation/differentiation state. Metabolic aberrations and changes in the intracellular levels of specific metabolites are linked to the inflammatory phenotype of immune cells implicated in autoimmune disorders such as systemic lupus erythematosus, rheumatoid arthritis, multiple sclerosis and diabetes. Notably, targeting metabolism such as the mTOR by rapamycin, hexokinase by 2-deoxy-D-glucose, AMP-activated protein kinase by metformin, may be used to ameliorate autoimmune inflammation. Accordingly, research in immunometabolism is expected to offer novel opportunities for monitoring and treating immune-mediated diseases.
Collapse
Affiliation(s)
- Chrysoula Stathopoulou
- Department of Rheumatology, Clinical Immunology & Allergy, University Hospital of Heraklion, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece.,Laboratory of Rheumatology, Autoimmunity & Inflammation, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece.,Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, N. Plastira 100, 70013 Heraklion, Greece
| | - Dimitra Nikoleri
- Department of Rheumatology, Clinical Immunology & Allergy, University Hospital of Heraklion, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece.,Laboratory of Rheumatology, Autoimmunity & Inflammation, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece.,Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, N. Plastira 100, 70013 Heraklion, Greece
| | - George Bertsias
- Department of Rheumatology, Clinical Immunology & Allergy, University Hospital of Heraklion, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece.,Laboratory of Rheumatology, Autoimmunity & Inflammation, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece.,Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, N. Plastira 100, 70013 Heraklion, Greece
| |
Collapse
|
46
|
Mapstone M, Gross TJ, Macciardi F, Cheema AK, Petersen M, Head E, Handen BL, Klunk WE, Christian BT, Silverman W, Lott IT, Schupf N. Metabolic correlates of prevalent mild cognitive impairment and Alzheimer's disease in adults with Down syndrome. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2020; 12:e12028. [PMID: 32258359 PMCID: PMC7131985 DOI: 10.1002/dad2.12028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/14/2020] [Accepted: 02/19/2020] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Disruption of metabolic function is a recognized feature of late onset Alzheimer's disease (LOAD). We sought to determine whether similar metabolic pathways are implicated in adults with Down syndrome (DS) who have increased risk for Alzheimer's disease (AD). METHODS We examined peripheral blood from 292 participants with DS who completed baseline assessments in the Alzheimer's Biomarkers Consortium-Down Syndrome (ABC-DS) using untargeted mass spectrometry (MS). Our sample included 38 individuals who met consensus criteria for AD (DS-AD), 43 who met criteria for mild cognitive impairment (DS-MCI), and 211 who were cognitively unaffected and stable (CS). RESULTS We measured relative abundance of 8,805 features using MS and 180 putative metabolites were differentially expressed (DE) among the groups at false discovery rate-corrected q< 0.05. From the DE features, a nine-feature classifier model classified the CS and DS-AD groups with receiver operating characteristic area under the curve (ROC AUC) of 0.86 and a two-feature model classified the DS-MCI and DS-AD groups with ROC AUC of 0.88. Metabolite set enrichment analysis across the three groups suggested alterations in fatty acid and carbohydrate metabolism. DISCUSSION Our results reveal metabolic alterations in DS-AD that are similar to those seen in LOAD. The pattern of results in this cross-sectional DS cohort suggests a dynamic time course of metabolic dysregulation which evolves with clinical progression from non-demented, to MCI, to AD. Metabolomic markers may be useful for staging progression of DS-AD.
Collapse
Affiliation(s)
- Mark Mapstone
- Department of NeurologyUniversity of California‐IrvineIrvineCaliforniaUSA
| | - Thomas J Gross
- Department of NeurologyUniversity of California‐IrvineIrvineCaliforniaUSA
| | - Fabio Macciardi
- Department of Psychiatry and Human BehaviorUniversity of California‐IrvineIrvineCaliforniaUSA
| | - Amrita K Cheema
- Departments of Biochemistry and Molecular & Cellular BiologyGeorgetown University Medical CenterWashingtonDCUSA
| | - Melissa Petersen
- Institute for Translational ResearchUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Elizabeth Head
- Department of Pathology and Laboratory MedicineUniversity of California‐IrvineIrvineCaliforniaUSA
| | - Benjamin L Handen
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - William E Klunk
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Bradley T Christian
- Departments of Medical Physics and PsychiatryWaisman CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Wayne Silverman
- Department of PediatricsUniversity of California‐ IrvineIrvineCaliforniaUSA
| | - Ira T Lott
- Department of PediatricsUniversity of California‐ IrvineIrvineCaliforniaUSA
| | - Nicole Schupf
- Taub Institute for Research in Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
- Department of NeurologyColumbia University and the New York Presbyterian HospitalNew YorkNew YorkUSA
- Department of EpidemiologyJoseph P. Mailman School of Public HealthColumbia UniversityNew YorkNew YorkUSA
- Gertrude H. Sergievsky CenterColumbia UniversityNew YorkNew YorkUSA
| | | |
Collapse
|
47
|
Obrocki P, Khatun A, Ness D, Senkevich K, Hanrieder J, Capraro F, Mattsson N, Andreasson U, Portelius E, Ashton NJ, Blennow K, Schöll M, Paterson RW, Schott JM, Zetterberg H. Perspectives in fluid biomarkers in neurodegeneration from the 2019 biomarkers in neurodegenerative diseases course-a joint PhD student course at University College London and University of Gothenburg. ALZHEIMERS RESEARCH & THERAPY 2020; 12:20. [PMID: 32111242 PMCID: PMC7049194 DOI: 10.1186/s13195-020-00586-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 02/12/2020] [Indexed: 12/12/2022]
Abstract
Until relatively recently, a diagnosis of probable Alzheimer's disease (AD) and other neurodegenerative disorders was principally based on clinical presentation, with post-mortem examination remaining a gold standard for disease confirmation. This is in sharp contrast to other areas of medicine, where fluid biomarkers, such as troponin levels in myocardial infarction, form an integral part of the diagnostic and treatment criteria. There is a pressing need for such quantifiable and easily accessible tools in neurodegenerative diseases.In this paper, based on lectures given at the 2019 Biomarkers in Neurodegenerative Diseases Course, we provide an overview of a range of cerebrospinal fluid (CSF) and blood biomarkers in neurodegenerative disorders, including the 'core' AD biomarkers amyloid β (Aβ) and tau, as well as other disease-specific and general markers of neuroaxonal injury. We then highlight the main challenges in the field, and how those could be overcome with the aid of new methodological advances, such as assay automation, mass spectrometry and ultrasensitive immunoassays.As we hopefully move towards an era of disease-modifying treatments, reliable biomarkers will be essential to increase diagnostic accuracy, allow for earlier diagnosis, better participant selection and disease activity and treatment effect monitoring.
Collapse
Affiliation(s)
- Pawel Obrocki
- Department of Medicine, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, UK.
| | - Ayesha Khatun
- Dementia Research Centre, Department of Neurodegeneration, UCL Institute of Neurology, London, UK
| | - Deborah Ness
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Konstantin Senkevich
- First Pavlov State Medical University of St. Petersburg, St. Petersburg, Russia.,Petersburg Nuclear Physics Institute named by B.P. Konstantinov of National Research Center, Kurchatov Institute, Gatchina, Russia
| | - Jörg Hanrieder
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Federica Capraro
- The Francis Crick Institute, London, UK.,Department of Neuromuscular Diseases, University College London Queen Square Institute of Neurology, London, UK
| | - Niklas Mattsson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden.,Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Ulf Andreasson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Erik Portelius
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK.,NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Michael Schöll
- Dementia Research Centre, Department of Neurodegeneration, UCL Institute of Neurology, London, UK.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Ross W Paterson
- Dementia Research Centre, Department of Neurodegeneration, UCL Institute of Neurology, London, UK
| | - Jonathan M Schott
- Dementia Research Centre, Department of Neurodegeneration, UCL Institute of Neurology, London, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,UK Dementia Research Institute, University College London, London, UK.,Department of Neurodegenerative Disease, University College London Institute of Neurology, London, UK
| |
Collapse
|
48
|
Gresenz CR, Mitchell JM, Marrone J, Federoff HJ. Effect of early-stage Alzheimer's disease on household financial outcomes. HEALTH ECONOMICS 2020; 29:18-29. [PMID: 31650668 DOI: 10.1002/hec.3962] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 09/02/2019] [Accepted: 09/15/2019] [Indexed: 06/10/2023]
Abstract
Significant limitations and rapid declines in financial capacity are a hallmark of patients with early-stage Alzheimer's disease (AD). We use linked Health and Retirement Study and Medicare claims data spanning 1992-2014 to examine the effect of early-stage AD, from the start of first symptoms to diagnosis, on household financial outcomes. We estimate household fixed-effects models and examine continuous measures of liquid assets and net wealth, as well as dichotomous indicators for a large change in either outcome. We find robust evidence that early-stage AD places households at significant risk for large adverse changes in liquid assets. Further, we find some, but more limited, evidence that early-stage AD reduces net wealth. Our findings are consequential because financial vulnerability during the disease's early-stage impacts the ability of afflicted individuals and their families to pay for care in the disease's later stage. Additionally, the findings speak to the value that earlier diagnosis may provide by helping avert adverse financial outcomes that occur before the disease is currently diagnosable with available tools. These results also point to a potentially important role for financial institutions in helping reduce exposure of vulnerable elderly to poor outcomes.
Collapse
Affiliation(s)
- Carole Roan Gresenz
- Department of Health Systems Administration, Georgetown University, Washington, D.C., USA
| | - Jean M Mitchell
- McCourt School of Public Policy, Georgetown University, Washington, D.C., USA
| | | | - Howard J Federoff
- Department of Neurology, School of Medicine, Henry and Susan Samueli College of Health Sciences, University of California-Irvine, Irvine, California
| |
Collapse
|
49
|
Moretti R, Peinkhofer C. B Vitamins and Fatty Acids: What Do They Share with Small Vessel Disease-Related Dementia? Int J Mol Sci 2019; 20:E5797. [PMID: 31752183 PMCID: PMC6888477 DOI: 10.3390/ijms20225797] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 10/21/2019] [Accepted: 11/12/2019] [Indexed: 12/12/2022] Open
Abstract
Many studies have been written on vitamin supplementation, fatty acid, and dementia, but results are still under debate, and no definite conclusion has yet been drawn. Nevertheless, a significant amount of lab evidence confirms that vitamins of the B group are tightly related to gene control for endothelium protection, act as antioxidants, play a co-enzymatic role in the most critical biochemical reactions inside the brain, and cooperate with many other elements, such as choline, for the synthesis of polyunsaturated phosphatidylcholine, through S-adenosyl-methionine (SAM) methyl donation. B-vitamins have anti-inflammatory properties and act in protective roles against neurodegenerative mechanisms, for example, through modulation of the glutamate currents and a reduction of the calcium currents. In addition, they also have extraordinary antioxidant properties. However, laboratory data are far from clinical practice. Many studies have tried to apply these results in everyday clinical activity, but results have been discouraging and far from a possible resolution of the associated mysteries, like those represented by Alzheimer's disease (AD) or small vessel disease dementia. Above all, two significant problems emerge from the research: No consensus exists on general diagnostic criteria-MCI or AD? Which diagnostic criteria should be applied for small vessel disease-related dementia? In addition, no general schema exists for determining a possible correct time of implementation to have effective results. Here we present an up-to-date review of the literature on such topics, shedding some light on the possible interaction of vitamins and phosphatidylcholine, and their role in brain metabolism and catabolism. Further studies should take into account all of these questions, with well-designed and world-homogeneous trials.
Collapse
Affiliation(s)
- Rita Moretti
- Neurology Clinic, Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy;
| | | |
Collapse
|
50
|
St John-Williams L, Mahmoudiandehkordi S, Arnold M, Massaro T, Blach C, Kastenmüller G, Louie G, Kueider-Paisley A, Han X, Baillie R, Motsinger-Reif AA, Rotroff D, Nho K, Saykin AJ, Risacher SL, Koal T, Moseley MA, Tenenbaum JD, Thompson JW, Kaddurah-Daouk R. Bile acids targeted metabolomics and medication classification data in the ADNI1 and ADNIGO/2 cohorts. Sci Data 2019; 6:212. [PMID: 31624257 PMCID: PMC6797798 DOI: 10.1038/s41597-019-0181-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 07/10/2019] [Indexed: 12/28/2022] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. The mechanism of disease development and progression is not well understood, but increasing evidence suggests multifactorial etiology, with a number of genetic, environmental, and aging-related factors. There is a growing body of evidence that metabolic defects may contribute to this complex disease. To interrogate the relationship between system level metabolites and disease susceptibility and progression, the AD Metabolomics Consortium (ADMC) in partnership with AD Neuroimaging Initiative (ADNI) is creating a comprehensive biochemical database for patients in the ADNI1 cohort. We used the Biocrates Bile Acids platform to evaluate the association of metabolic levels with disease risk and progression. We detail the quantitative metabolomics data generated on the baseline samples from ADNI1 and ADNIGO/2 (370 cognitively normal, 887 mild cognitive impairment, and 305 AD). Similar to our previous reports on ADNI1, we present the tools for data quality control and initial analysis. This data descriptor represents the third in a series of comprehensive metabolomics datasets from the ADMC on the ADNI.
Collapse
Affiliation(s)
- Lisa St John-Williams
- Proteomics and Metabolomics Shared Resource, Center for Genomics and Computational Biology, Duke University, Durham, NC, USA
| | | | - Matthias Arnold
- Department of Psychiatry & Behavioral Sciences, Duke University, Durham, NC, USA
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Tyler Massaro
- Duke Clinical Research Institute, Duke University, Durham, NC, USA
| | - Colette Blach
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - Gabi Kastenmüller
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Gregory Louie
- Department of Psychiatry & Behavioral Sciences, Duke University, Durham, NC, USA
| | | | - Xianlin Han
- University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | | | - Alison A Motsinger-Reif
- Bioinformatics Research Center, Department of Statistics, North Carolina State University, Raleigh, NC, USA
| | - Daniel Rotroff
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Kwangsik Nho
- Department of Radiology and Imaging Sciences and the Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences and the Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shannon L Risacher
- Department of Radiology and Imaging Sciences and the Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - M Arthur Moseley
- Proteomics and Metabolomics Shared Resource, Center for Genomics and Computational Biology, Duke University, Durham, NC, USA
| | - Jessica D Tenenbaum
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - J Will Thompson
- Proteomics and Metabolomics Shared Resource, Center for Genomics and Computational Biology, Duke University, Durham, NC, USA
| | - Rima Kaddurah-Daouk
- Department of Psychiatry & Behavioral Sciences, Duke University, Durham, NC, USA.
- Duke Institute for Brain Sciences, Duke University, Durham, NC, USA.
| |
Collapse
|