1
|
Beydoun MA, Beydoun HA, Hu YH, Li Z, Georgescu MF, Noren Hooten N, Bouhrara M, Weiss J, Launer LJ, Evans MK, Zonderman AB. Mediating and moderating effects of plasma proteomic biomarkers on the association between poor oral health problems and brain white matter microstructural integrity: the UK Biobank study. Mol Psychiatry 2025; 30:388-401. [PMID: 39080466 DOI: 10.1038/s41380-024-02678-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 01/22/2025]
Abstract
The plasma proteome can mediate associations between periodontal disease (Pd) and brain white matter integrity (WMI). We screened 5089 UK Biobank participants aged 40-70 years for poor oral health problems (POHP). We examined the association between POHP and WMI (fractional anisotropy (FA), mean diffusivity (MD), Intracellular Volume Fraction (ICVF), Isotropic Volume Fraction (ISOVF) and Orientation Diffusion (OD)), decomposing the total effect through the plasma proteome of 1463 proteins into pure mediation, pure interaction, neither, while adjusting for socio-demographic and cardiovascular health factors. Similarly, structural equations modeling (SEM) was conducted. POHP was more prevalent among men (12.3% vs. 9.6%), and was associated with lower WMI on most metrics, in a sex-specific manner. Of 15 proteins strongly associated with POHP, growth differentiation factor 15 (GDF15) and WAP four-disulfide core domain 2 (WFDC2; also known as human epididymis protein 4; HE4) were consistent mediators. Both proteins mediated 7-8% of total POHP effect on FAmean. SEM yielded significant total effects for FAmean, MDmean and ISOVFmean in full models, with %mediated by common latent factor (GDF15 and WFDC2) ranging between 13% (FAmean) and 19% (ISOVFmean). For FA, mediation by this common factor was found for 16 of 49 tract-specific and global mean metrics. Protein metabolism, immune system, and signal transduction were the most common pathways for mediational effects. POHP was associated with poorer WMI, which was partially mediated by GDF15 and WFDC2.
Collapse
Affiliation(s)
- May A Beydoun
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD, 21224, USA.
| | - Hind A Beydoun
- VA National Center on Homelessness Among Veterans, U.S. Department of Veterans Affairs, Washington, DC, 20420, USA
- Department of Management, Policy, and Community Health, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Yi-Han Hu
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD, 21224, USA
| | - Zhiguang Li
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD, 21224, USA
| | - Michael F Georgescu
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD, 21224, USA
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD, 21224, USA
| | - Mustapha Bouhrara
- Laboratory of Clinical Investigation, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD, 21224, USA
| | - Jordan Weiss
- Stanford Center on Longevity, Stanford University, Stanford, CA, 94301, USA
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD, 21224, USA
| | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD, 21224, USA
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD, 21224, USA
| |
Collapse
|
2
|
Ao J, Picard C, Auld D, Zetterberg H, Brinkmalm A, Blennow K, Villeneuve S, Breitner JCS, Poirier J. Novel synaptic markers predict early tau pathology and cognitive deficit in an asymptomatic population at risk of Alzheimer's disease. Mol Psychiatry 2025:10.1038/s41380-024-02884-z. [PMID: 39827219 DOI: 10.1038/s41380-024-02884-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 12/11/2024] [Accepted: 12/27/2024] [Indexed: 01/22/2025]
Abstract
Cognitive dysfunction in Alzheimer's disease (AD) correlates closely with pathology in the neuronal microtubule-associated protein tau. Tau pathology may spread via neural synapses. In a population of cognitively unimpaired elderly at elevated risk of AD, we investigated four cerebrospinal (CSF) markers of synaptic dysfunction and degeneration. Three of these (SYT1, SNAP25, and ADAM23) are derived from pre-synaptic structures, while ADAM22 reflects post-synaptic changes. All four markers correlated strongly with tau protein measures. In statistical models, SYT1 accounted for more than half the total variance in both total- and P(181)-tau levels. Observed correlations with CSF levels of Alzheimer amyloid-β (Aβ42) were somewhat weaker. In longitudinal data, baseline levels of ADAM22 and ADAM23 robustly predicted increase over time in both total- and P-tau. CSF SYT1 levels also correlated with PET image uptake of tau and (at a trend level) Aβ in areas of interest for early AD pathology. CSF SYT1 and SNAP25 levels correlated inversely with a global psychometric score and several of its domain subscales. In quantitative trait loci analyses, all four synaptic markers were associated with at least one AD genetic risk locus. Upon "staging" participants by their evidence of amyloid and tau pathology (A/T/N framework), the CSF synaptic markers were unexpectedly reduced in participants with CSF evidence of amyloid but not tau pathology. They were clearly elevated, however, in the CSF of persons with indications of both tau and amyloid pathology. These observations provide evidence for clear pre-synaptic degeneration in cognitively unimpaired persons with biomarker evidence of early AD pathology.
Collapse
Affiliation(s)
- Jiarui Ao
- Douglas Mental Health University Institute, Montréal, QC, Canada
- Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, QC, Canada
| | - Cynthia Picard
- Douglas Mental Health University Institute, Montréal, QC, Canada
- Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, QC, Canada
| | - Daniel Auld
- Victor Phillip Dahdaleh Institute of Genomic Medicine at McGill University, Montréal, QC, Canada
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Lab, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Ann Brinkmalm
- Clinical Neurochemistry Lab, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Kaj Blennow
- Clinical Neurochemistry Lab, Sahlgrenska University Hospital, Mölndal, Sweden
- Inst. of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, PR China
| | - Sylvia Villeneuve
- Douglas Mental Health University Institute, Montréal, QC, Canada
- Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, QC, Canada
| | - John C S Breitner
- Douglas Mental Health University Institute, Montréal, QC, Canada
- Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, QC, Canada
| | - Judes Poirier
- Douglas Mental Health University Institute, Montréal, QC, Canada.
- Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, QC, Canada.
| |
Collapse
|
3
|
Beydoun MA, Beydoun HA, Noren Hooten N, Li Z, Hu YH, Georgescu MF, Hossain S, Tanaka T, Bouhrara M, Maino Vieytes CA, Fanelli-Kuczmarski MT, Launer LJ, Evans MK, Zonderman AB. Plasma proteomic biomarkers as mediators or moderators for the association between poor cardiovascular health and white matter microstructural integrity: The UK Biobank study. Alzheimers Dement 2025. [PMID: 39822062 DOI: 10.1002/alz.14507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 11/16/2024] [Accepted: 12/03/2024] [Indexed: 01/19/2025]
Abstract
INTRODUCTION The plasma proteome's mediating or moderating roles in the association between poor cardiovascular health (CVH) and brain white matter (WM) microstructural integrity are largely unknown. METHODS Data from 3953 UK Biobank participants were used (40-70 years, 2006-2010), with a neuroimaging visit between 2014 and 2021. Poor CVH was determined using Life's Essential 8 (LE8) and reversing standardized z-scores (LE8z _rev). The plasma proteome was examined as a potential mediator or moderator of LE8z _rev's effects on quantitative diffusion-weighted magnetic resonance imaging (dMRI) metrics. RESULTS LE8z_rev was significantly associated with deteriorated WM microstructural integrity, as reflected by lower tract-averaged fractional anisotropy (dMRI-FAmean), (β ± standared error (SE): -0.00152 ± 0.0003, p < 0.001) and higher tract-averaged orientation dispersion (dMRI-ODmean), (β ± SE:+0.00081 ± 0.00017, p < 0.001). Ten strongly mediating plasma proteins of 1463 were identified, with leptin as the principal driver. DISCUSSION Poor CVH is linked to poor WM microstructural integrity measures (lower FAmean and higher ODmean), mostly mediated through leptin. HIGHLIGHTS Up to 3953 UK Biobank participants were selected for this study. Poor cardiovascular health (CVH) was determined using Life's Essential 8. The plasma proteome was examined as a potential mediator or moderator of poor CVH's effect on dMRI metrics. Ten plasma proteins were identified with strong mediating effects, with leptin being the principal driver.
Collapse
Affiliation(s)
- May A Beydoun
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, Maryland, USA
| | - Hind A Beydoun
- VA National Center on Homelessness Among Veterans, U.S. Department of Veterans Affairs, Washington, DC, USA
- Department of Management, Policy, and Community Health, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, Maryland, USA
| | - Zhiguang Li
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, Maryland, USA
| | - Yi-Han Hu
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, Maryland, USA
| | - Michael F Georgescu
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, Maryland, USA
| | - Sharmin Hossain
- Department of Human Services (DHS), State of Maryland, Baltimore, Maryland, USA
| | - Toshiko Tanaka
- Translational Gerontology Branch, National Institute on Aging, NIA/NIH/IRP, Baltimore, Maryland, USA
| | - Mustapha Bouhrara
- Laboratory of Clinical Investigation, National Institute on Aging, NIA/NIH/IRP, Baltimore, Maryland, USA
| | - Christian A Maino Vieytes
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, Maryland, USA
| | - Marie T Fanelli-Kuczmarski
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, Maryland, USA
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, Maryland, USA
| | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, Maryland, USA
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, Maryland, USA
| |
Collapse
|
4
|
Tiet MY, Guțu BI, Springall-Jeggo P, Coman D, Willemsen M, Van Os N, Doria M, Donath H, Schubert R, Dineen RA, Biagiotti S, Prayle AP, Group ATBW, Hensiek AE, Horvath R. Biomarkers in Ataxia-Telangiectasia: a Systematic Review. J Neurol 2025; 272:110. [PMID: 39812834 PMCID: PMC11735505 DOI: 10.1007/s00415-024-12766-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 10/24/2024] [Accepted: 10/26/2024] [Indexed: 01/16/2025]
Abstract
Ataxia-Telangiectasia (A-T) is a very rare multisystem disease of DNA repair, associated with progressive disabling neurological symptoms, respiratory failure, immunodeficiency and cancer predisposition, leading to premature death. There are no curative treatments available for A-T but clinical trials have begun. A major limiting factor in effectively evaluating therapies for A-T is the lack of suitable outcome measures and biomarkers. We have performed a systematic review to collect the information currently available on biomarkers for A-T both in patients and preclinical studies. We have identified 56 reports discussing potential A-T biomarkers in both pre-clinical models and patients. These studies report on diagnostic biomarkers but prognostic biomarkers and responsive markers of clinical status are currently lacking. Some biomarkers of neurodegeneration in A-T show promise, including non-invasive neuroimaging biomarkers. Some biomarkers of oxidative stress and responsive markers to radiotherapy and steroid treatment have potential value in clinical trials. The formation of the A-T biomarker working group with international experts is an important step forward to facilitate the sharing of materials, data and expertise with the common goal of finding effective biomarkers for A-T.
Collapse
Affiliation(s)
- M Y Tiet
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, University of Cambridge, Robinson Way, Cambridge, CB2 0PY, UK
| | - B-I Guțu
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, University of Cambridge, Robinson Way, Cambridge, CB2 0PY, UK
| | | | - D Coman
- Queensland Children's Hospital, 501 Stanley Street, South Brisbane, Australia
| | - M Willemsen
- Department of Pediatrics, Pediatric Neurology, Radboud University Medical Centre, Amalia Children's Hospital, Nijmegen, Netherlands
| | - N Van Os
- Department of Pediatrics, Pediatric Neurology, Radboud University Medical Centre, Amalia Children's Hospital, Nijmegen, Netherlands
| | - M Doria
- Primary Immunodeficiency Unit, Bambino Gesù Children's Hospital, Rome, Italy
| | - H Donath
- Division of Pneumology, Allergology, Infectiology and Gastroenterology, Department of Children and Adolescent Medicine, University Hospital, Goethe University, Frankfurt, Germany
| | - R Schubert
- Division of Pneumology, Allergology, Infectiology and Gastroenterology, Department of Children and Adolescent Medicine, University Hospital, Goethe University, Frankfurt, Germany
| | - R A Dineen
- Mental Health and Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham, UK
| | - S Biagiotti
- Department of Biomolecular Sciences, University of Urbino, Urbino, Italy
| | - A P Prayle
- NIHR Nottingham Biomedical Research Centre, Nottingham, UK
- Division of Child Health, Obstetrics and Gynaecology, University of Nottingham, Nottingham, UK
| | | | - A E Hensiek
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, University of Cambridge, Robinson Way, Cambridge, CB2 0PY, UK
| | - R Horvath
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, University of Cambridge, Robinson Way, Cambridge, CB2 0PY, UK.
| |
Collapse
|
5
|
Nehra G, Maloney BJ, Smith RR, Chumboatong W, Abner EL, Nelson PT, Bauer B, Hartz AMS. Plasma S100β is a predictor for pathology and cognitive decline in Alzheimer's disease. Fluids Barriers CNS 2025; 22:4. [PMID: 39789614 PMCID: PMC11720585 DOI: 10.1186/s12987-024-00615-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 12/20/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Blood-brain barrier dysfunction is one characteristic of Alzheimer's disease (AD) and is recognized as both a cause and consequence of the pathological cascade leading to cognitive decline. The goal of this study was to assess markers for barrier dysfunction in postmortem tissue samples from research participants who were either cognitively normal individuals (CNI) or diagnosed with AD at the time of autopsy and determine to what extent these markers are associated with AD neuropathologic changes (ADNC) and cognitive impairment. METHODS We used postmortem brain tissue and plasma samples from 19 participants: 9 CNI and 10 AD dementia patients who had come to autopsy from the University of Kentucky AD Research Center (UK-ADRC) community-based cohort; all cases with dementia had confirmed severe ADNC. Plasma samples were obtained within 2 years of autopsy. Aβ40, Aβ42, and tau levels in brain tissue samples were quantified by ELISA. Cortical brain sections were cleared using the X-CLARITY™ system and immunostained for neurovascular unit-related proteins. Brain slices were then imaged using confocal microscopy and analyzed for microvascular diameters and immunoreactivity coverage using Fiji/ImageJ. Isolated human brain microvessels were assayed for tight-junction protein expression using the JESS™ automated Western blot system. S100 calcium-binding protein B (S100β), matrix metalloproteinase (MMP)-2, MMP-9, and neuron-specific enolase (NSE) levels in plasma were quantified by ELISA. All outcomes were assessed for linear associations with global cognitive function (MMSE, CDR) and cerebral atrophy scores by Pearson, polyserial, or polychoric correlation, as appropriate, along with generalized linear modeling or generalized linear mixed-level modeling. RESULTS As expected, we detected elevated Aβ and tau pathology in brain tissue sections from AD patients compared to CNI. However, we found no differences in microvascular diameters in cleared AD and CNI brain tissue sections. We also observed no differences in claudin-5 protein levels in capillaries isolated from AD and CNI tissue samples. Plasma biomarker analysis showed that AD patients had 12.4-fold higher S100β plasma levels, twofold lower NSE plasma levels, 2.4-fold higher MMP-9 plasma levels, and 1.2-fold lower MMP-2 plasma levels than CNI. Data analysis revealed that elevated S100β plasma levels were predictive of AD pathology and cognitive impairment. CONCLUSION Our data suggest that among different markers relevant to barrier dysfunction, plasma S100β is the most promising diagnostic biomarker for ADNC. Further investigation is necessary to assess how plasma S100β levels relate to these changes and whether they may predict clinical outcomes, particularly in the prodromal and early stages of AD.
Collapse
Affiliation(s)
- Geetika Nehra
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, 760 Press Ave, 124 HKRB, Lexington, KY, 40536-0679, USA
| | - Bryan J Maloney
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, 760 Press Ave, 124 HKRB, Lexington, KY, 40536-0679, USA
| | - Rebecca R Smith
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, 760 Press Ave, 124 HKRB, Lexington, KY, 40536-0679, USA
| | - Wijitra Chumboatong
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, 760 Press Ave, 124 HKRB, Lexington, KY, 40536-0679, USA
| | - Erin L Abner
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, 760 Press Ave, 124 HKRB, Lexington, KY, 40536-0679, USA
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, USA
- Department of Epidemiology and Environmental Health, College of Public Health, University of Kentucky, Lexington, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, 760 Press Ave, 124 HKRB, Lexington, KY, 40536-0679, USA
- Department of Pathology, College of Medicine, University of Kentucky, Lexington, USA
| | - Björn Bauer
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, 760 Press Ave, 124 HKRB, Lexington, KY, 40536-0679, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, USA
| | - Anika M S Hartz
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, 760 Press Ave, 124 HKRB, Lexington, KY, 40536-0679, USA.
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, USA.
| |
Collapse
|
6
|
Radosinska D, Radosinska J. The Link Between Matrix Metalloproteinases and Alzheimer's Disease Pathophysiology. Mol Neurobiol 2025; 62:885-899. [PMID: 38935232 PMCID: PMC11711632 DOI: 10.1007/s12035-024-04315-0] [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: 01/10/2024] [Accepted: 06/16/2024] [Indexed: 06/28/2024]
Abstract
Alzheimer's disease (AD) is a major contributor to dementia and the most common neurodegenerative disorder. In AD pathophysiology, matrix metalloproteinases (MMPs)-proteolytic enzymes, best known to be responsible for remodeling and degradation of the extracellular matrix-were suggested to play an important role. Due to the diverse nature of the published data and frequent inconsistent results presented in available papers, it was considered essential to analyze all aspects of MMP literature with respect to AD pathophysiology and attempt to outline a unifying concept for understanding their role in AD. Thus, the main contribution of this review article is to summarize the most recent research on the participation of MMP in AD pathophysiology obtained using the cell cultures to understand the molecular principles of their action. Furthermore, an updated comprehensive view regarding this topic based exclusively on papers from human studies is provided as well. It can be concluded that determining the exact role of any particular MMPs in the AD pathophysiology holds promise for establishing their role as potential biomarkers reflecting the severity or progression of this disease or for developing new therapeutic agents targeting the processes that lead to AD.
Collapse
Affiliation(s)
- Dominika Radosinska
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovak Republic
| | - Jana Radosinska
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 2, 81372, Bratislava, Slovak Republic.
| |
Collapse
|
7
|
Hunter TR, Santos LE, Tovar-Moll F, De Felice FG. Alzheimer's disease biomarkers and their current use in clinical research and practice. Mol Psychiatry 2025; 30:272-284. [PMID: 39232196 DOI: 10.1038/s41380-024-02709-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 09/06/2024]
Abstract
While blood-based tests are readily available for various conditions, including cardiovascular diseases, type 2 diabetes, and common cancers, Alzheimer's disease (AD) and other neurodegenerative diseases lack an early blood-based screening test that can be used in primary care. Major efforts have been made towards the investigation of approaches that may lead to minimally invasive, cost-effective, and reliable tests capable of measuring brain pathological status. Here, we review past and current technologies developed to investigate biomarkers of AD, including novel blood-based approaches and the more established cerebrospinal fluid and neuroimaging biomarkers of disease. The utility of blood as a source of AD-related biomarkers in both clinical practice and interventional trials is discussed, supported by a comprehensive list of clinical trials for AD drugs and interventions that list biomarkers as primary or secondary endpoints. We highlight that identifying individuals in early preclinical AD using blood-based biomarkers will improve clinical trials and the optimization of therapeutic treatments as they become available. Lastly, we discuss challenges that remain in the field and address new approaches being developed, such as the examination of cargo packaged within extracellular vesicles of neuronal origin isolated from peripheral blood.
Collapse
Affiliation(s)
- Tai R Hunter
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Luis E Santos
- D'Or Institute for Research and Education, Rio de Janeiro, RJ, Brazil.
| | | | - Fernanda G De Felice
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada.
- D'Or Institute for Research and Education, Rio de Janeiro, RJ, Brazil.
- Centre for Neuroscience Studies and Department of Psychiatry, Queen's University, Kingston, ON, Canada.
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
8
|
Puerta R, Cano A, García-González P, García-Gutiérrez F, Capdevila M, de Rojas I, Olivé C, Blázquez-Folch J, Sotolongo-Grau O, Miguel A, Montrreal L, Martino-Adami P, Khan A, Orellana A, Sung YJ, Frikke-Schmidt R, Marchant N, Lambert JC, Rosende-Roca M, Alegret M, Fernández MV, Marquié M, Valero S, Tárraga L, Cruchaga C, Ramírez A, Boada M, Smets B, Cabrera-Socorro A, Ruiz A. Head-to-Head Comparison of Aptamer- and Antibody-Based Proteomic Platforms in Human Cerebrospinal Fluid Samples from a Real-World Memory Clinic Cohort. Int J Mol Sci 2024; 26:286. [PMID: 39796148 PMCID: PMC11720409 DOI: 10.3390/ijms26010286] [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: 10/29/2024] [Revised: 12/16/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
High-throughput proteomic platforms are crucial to identify novel Alzheimer's disease (AD) biomarkers and pathways. In this study, we evaluated the reproducibility and reliability of aptamer-based (SomaScan® 7k) and antibody-based (Olink® Explore 3k) proteomic platforms in cerebrospinal fluid (CSF) samples from the Ace Alzheimer Center Barcelona real-world cohort. Intra- and inter-platform reproducibility were evaluated through correlations between two independent SomaScan® assays analyzing the same samples, and between SomaScan® and Olink® results. Association analyses were performed between proteomic measures, CSF biological traits, sample demographics, and AD endophenotypes. Our 12-category metric of reproducibility combining correlation analyses identified 2428 highly reproducible SomaScan CSF measures, with over 600 proteins well reproduced on another proteomic platform. The association analyses among AD clinical phenotypes revealed that the significant associations mainly involved reproducible proteins. The validation of reproducibility in these novel proteomics platforms, measured using this scarce biomaterial, is essential for accurate analysis and proper interpretation of innovative results. This classification metric could enhance confidence in multiplexed proteomic platforms and improve the design of future panels.
Collapse
Affiliation(s)
- Raquel Puerta
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
- PhD Program in Biotecnology, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain
| | - Amanda Cano
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos III, 28029 Madrid, Spain
| | - Pablo García-González
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos III, 28029 Madrid, Spain
| | - Fernando García-Gutiérrez
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
| | - Maria Capdevila
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
- Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, 08007 Barcelona, Spain
| | - Itziar de Rojas
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos III, 28029 Madrid, Spain
| | - Clàudia Olivé
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
| | - Josep Blázquez-Folch
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
| | - Oscar Sotolongo-Grau
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
| | - Andrea Miguel
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
| | - Laura Montrreal
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
| | - Pamela Martino-Adami
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (P.M.-A.); (A.R.)
| | - Asif Khan
- Janssen Pharmaceutica NV, a Johnson & Johnson Company, 2340 Beerse, Belgium; (A.K.); (B.S.); (A.C.-S.)
| | - Adelina Orellana
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos III, 28029 Madrid, Spain
| | - Yun Ju Sung
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO 63108, USA; (Y.J.S.); (C.C.)
- Hope Center for Neurological Disorders, Washington University, St. Louis, MO 63110, USA
| | - Ruth Frikke-Schmidt
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark;
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Natalie Marchant
- Division of Psychiatry, University College London, London W1T 7NK, UK;
| | - Jean Charles Lambert
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Université de Lille, F-59000 Lille, France;
- Institut Pasteur de Lille, Inserm U1167, CHU de Lille, LabEx DISTALZ, Université de Lille, F-59000 Lille, France
| | - Maitée Rosende-Roca
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
| | - Montserrat Alegret
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos III, 28029 Madrid, Spain
| | - Maria Victoria Fernández
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos III, 28029 Madrid, Spain
| | - Marta Marquié
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos III, 28029 Madrid, Spain
| | - Sergi Valero
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos III, 28029 Madrid, Spain
| | - Lluís Tárraga
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos III, 28029 Madrid, Spain
| | - Carlos Cruchaga
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO 63108, USA; (Y.J.S.); (C.C.)
- Hope Center for Neurological Disorders, Washington University, St. Louis, MO 63110, USA
| | - Alfredo Ramírez
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (P.M.-A.); (A.R.)
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, Medical Faculty, University Hospital Bonn, 53127 Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
- Department of Psychiatry and Glenn, Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, TX 78229, USA
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Mercè Boada
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos III, 28029 Madrid, Spain
| | - Bart Smets
- Janssen Pharmaceutica NV, a Johnson & Johnson Company, 2340 Beerse, Belgium; (A.K.); (B.S.); (A.C.-S.)
| | - Alfredo Cabrera-Socorro
- Janssen Pharmaceutica NV, a Johnson & Johnson Company, 2340 Beerse, Belgium; (A.K.); (B.S.); (A.C.-S.)
| | - Agustín Ruiz
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08029 Barcelona, Spain; (R.P.); (A.C.); (P.G.-G.); (F.G.-G.); (M.C.); (I.d.R.); (C.O.); (J.B.-F.); (O.S.-G.); (A.M.); (L.M.); (A.O.); (M.R.-R.); (M.A.); (M.V.F.); (M.M.); (S.V.); (L.T.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos III, 28029 Madrid, Spain
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center, San Antonio, TX 77204, USA
| |
Collapse
|
9
|
Zhang T, Si H, Liao J, Ma R. Association of plasma BDNF and MMP-9 levels with mild cognitive impairment: a matched case-control study. Sci Rep 2024; 14:30911. [PMID: 39730669 DOI: 10.1038/s41598-024-81895-w] [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: 08/28/2024] [Accepted: 11/29/2024] [Indexed: 12/29/2024] Open
Abstract
The prevalence of Alzheimer's disease (AD) is on the rise globally, and everyone who develops AD eventually experiences mild cognitive impairment (MCI) first. Timely intervention at an early stage of the disease may mitigate disease progression. Recent studies indicate that BDNF and MMP-9 play a significant role in the pathogenesis of AD. Therefore, this study aims to ascertain whether there are differences in plasma BDNF and MMP-9 levels between individuals with mild cognitive impairment due to AD and those with normal cognition, and to analyze the factors influencing mild cognitive impairment.This case-control study included 102 individuals with mild cognitive impairment and 102 controls, matched by age and sex. Participants completed a series of questionnaires, neuropsychological assessments, and clinical examinations. Plasma concentrations of BDNF and MMP-9 of the participants were quantified using ELISA. Subsequently, the factors influencing MCI were analyzed using univariate and multivariate logistic regression. The differences in plasma BDNF levels, MOCA total scores, and scores in various cognitive domains (including visuospatial and executive abilities, abstract thinking, attention, language, naming, and delayed memory) between the MCI and the control groups showed statistically significant (p < 0.05). Logistic regression analysis revealed that plasma BDNF levels and years of formal education were significantly negatively associated with MCI. This study indicates that plasma BDNF and years of formal education are protective factors influencing cognitive function.
Collapse
Affiliation(s)
- Tingyu Zhang
- Department of Public Health, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, The Xinjiang Production and Construction Corps. Shihezi University, Shihezi, Xinjiang, China
| | - Huili Si
- Department of Neurology, Shihezi People's Hospital, Shihezi, Xinjiang, China
| | - Jiali Liao
- Department of Public Health, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, The Xinjiang Production and Construction Corps. Shihezi University, Shihezi, Xinjiang, China
| | - Rulin Ma
- Department of Public Health, School of Medicine, Shihezi University, Shihezi, Xinjiang, China.
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, The Xinjiang Production and Construction Corps. Shihezi University, Shihezi, Xinjiang, China.
| |
Collapse
|
10
|
Lin CC'J, Tian Y, Tanzi RE, Jorfi M. Approaches for studying neuroimmune interactions in Alzheimer's disease. Trends Immunol 2024; 45:971-986. [PMID: 39537528 PMCID: PMC11624993 DOI: 10.1016/j.it.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024]
Abstract
Peripheral immune cells play an important role in the pathology of Alzheimer's disease (AD), impacting processes such as amyloid and tau protein aggregation, glial activation, neuronal integrity, and cognitive decline. Here, we examine cutting-edge strategies - encompassing animal and cellular models - used to investigate the roles of peripheral immune cells in AD. Approaches such as antibody-mediated depletion, genetic ablation, and bone marrow chimeras in mouse models have been instrumental in uncovering T, B, and innate immune cell disease-modifying functions. However, challenges such as specificity, off-target effects, and differences between human and mouse immune systems underscore the need for more human-relevant models. Emerging multicellular models replicating critical aspects of human brain tissue and neuroimmune interactions increasingly offer fresh insights into the role of immune cells in AD pathogenesis. Refining these methodologies can deepen our understanding of immune cell contributions to AD and support the development of novel immune-related therapeutic interventions.
Collapse
Affiliation(s)
- Chih-Chung 'Jerry' Lin
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Yuyao Tian
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.
| | - Mehdi Jorfi
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.
| |
Collapse
|
11
|
Prabha S, Sajad M, Anjum F, Hassan MI, Shamsi A, Thakur SC. Investigating gene expression datasets of hippocampus tissue to discover Alzheimer's disease-associated molecular markers. J Alzheimers Dis 2024; 102:994-1016. [PMID: 39604273 DOI: 10.1177/13872877241297335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is an advancing neurodegenerative disorder distinguished by the formation of amyloid plaques and neurofibrillary tangles in the human brain. Nevertheless, the lack of peripheral biomarkers that can detect the development of AD remains a significant limitation. OBJECTIVE The main aim of this work was to discover the molecular markers associated with AD. METHODS We conducted a comprehensive microarray analysis of gene expression data from hippocampus tissue in AD patients and control samples using three microarray datasets (GSE1297, GSE28146, and GSE29378) collected from Gene Expression Omnibus (GEO). The datasets were pre-processed and normalized, revealing 346 significant genes, 103 of which were upregulated and 243 downregulated. The PPI network of significant genes was constructed to detect the top 50 hub genes, which were then further analyzed using Gene Ontology (GO) terms, Kyoto Encyclopedia of Genes and Genomes pathway (KEGG), and GSEA, revealing 47 key genes involved in AD-related pathways. These key genes were then subjected to feed forward loop (FFL) motif analysis for the prediction of transcriptional factors (TFs) and microRNAs (miRNAs) mediated gene regulatory networks. RESULTS The interaction of AD-associated TFs HNF4A, SPI1, EGR1, STAT3, and MYC and miRNAs hsa-miR-155-5p and hsa-miR-16-5p in the transcriptional and post-transcriptional events of 3 upregulated and 10 downregulated genes: H2AFZ, MCM3, MYO1C, AXIN1, CCND1, ETS2, MYH9, RELA, RHEB, SOCS3, TBL1X, TBP, TXNIP, and YWHAZ, respectively, has been identified. The miRNA/TF-mediated three types of the FFL motifs, i.e., miRNA-FFL, TF-FFL, and composite-FFL, were constructed, and seven common genes among these FFL were identified: CCND1, MYH9, SOCS3, RHEB, MYO1C, TXNIP, AXIN1, and TXNIP. CONCLUSIONS These findings may provide insights into the development of potential molecular markers for therapeutic management of AD.
Collapse
Affiliation(s)
- Sneh Prabha
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Mohd Sajad
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Farah Anjum
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Anas Shamsi
- Center of Medical and Bio-Allied Health Sciences Research (CMBHSR), Ajman University, Ajman, United Arab Emirates
| | - Sonu Chand Thakur
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
12
|
Morató X, Puerta R, Cano A, Orellana A, de Rojas I, Capdevila M, Montrreal L, Rosende-Roca M, García-González P, Olivé C, García-Gutiérrez F, Blázquez J, Miguel A, Núñez-Llaves R, Pytel V, Alegret M, Fernández MV, Marquié M, Valero S, Cavazos JE, Mañes S, Boada M, Cabrera-Socorro A, Ruiz A. Associations of plasma SMOC1 and soluble IL6RA levels with the progression from mild cognitive impairment to dementia. Brain Behav Immun Health 2024; 42:100899. [PMID: 39640195 PMCID: PMC11617377 DOI: 10.1016/j.bbih.2024.100899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 08/12/2024] [Accepted: 10/27/2024] [Indexed: 12/07/2024] Open
Abstract
Despite the central role attributed to neuroinflammation in the etiology and pathobiology of Alzheimer's disease (AD), the direct link between levels of inflammatory mediators in blood and cerebrospinal fluid (CSF) compartments, as well as their potential implications for AD diagnosis and progression, remains inconclusive. Moreover, there is debate on whether inflammation has a protective or detrimental effect on disease onset and progression. Indeed, distinct immunological mechanisms may govern protective and damaging effects at early and late stages, respectively. This study aims to (i) identify inflammatory mediators demonstrating robust correlations between peripheral and central nervous system (CNS) compartments by means of plasma and CSF analysis, respectively, and (ii) assess their potential significance in the context of AD and disease progression from mild cognitive impairment (MCI) to dementia. To achieve this, we have examined the inflammatory profile of a well-defined subcohort comprising 485 individuals from the Ace Alzheimer Center Barcelona (ACE). Employing a hierarchical clustering approach, we thoroughly evaluated the intercompartmental correlations of 63 distinct inflammation mediators, quantified in paired CSF and plasma samples, using advanced SOMAscan technology. Of the array of mediators investigated, only six mediators (CRP, IL1RAP, ILRL1, IL6RA, PDGFRB, and YKL-40) exhibited robust correlations between the central and peripheral compartments (proximity scores <400). To strengthen the validity of our findings, these identified mediators were subsequently validated in a second subcohort of individuals from ACE (n = 873). The observed plasma correlations across the entire cohort consistently have a Spearman rho value above 0.51 (n = 1,360, p < 1.77E-93). Of the high CSF-plasma correlated proteins, only soluble IL6RA (sIL6RA) displayed a statistically significant association with the conversion from MCI to dementia. This association remained robust even after applying a stringent Bonferroni correction (Cox proportional hazard ratio [HR] = 1.936 per standard deviation; p = 0.0018). This association retained its significance when accounting for various factors, including CSF amyloid (Aβ42) and Thr181-phosphorylated tau (p-tau) levels, age, sex, baseline Mini-Mental State Examination (MMSE) score, and potential sampling biases identified through principal component analysis (PCA) modeling. Furthermore, our study confirmed the association of both plasma and CSF levels of SPARC-related modular calcium-binding protein 1 (SMOC1) with amyloid and tau accumulation, indicating their role as early surrogate biomarkers for AD pathology. Despite the lack of a statistically significant correlation between SMOC1 levels in CSF and plasma, both acted as independent biomarkers of disease progression (HR > 1.3, p < 0.002). In conclusion, our study unveils that sIL6RA and SMOC1 are associated with MCI progression. The absence of correlations among inflammatory mediators between the central and peripheral compartments appears to be a common pattern, with only a few intriguing exceptions.
Collapse
Affiliation(s)
- Xavier Morató
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Raquel Puerta
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
- Universitat de Barcelona (UB), Spain
| | - Amanda Cano
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Spain
| | - Adelina Orellana
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Itziar de Rojas
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - María Capdevila
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Spain
| | - Laura Montrreal
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
| | - Maitée Rosende-Roca
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
| | - Pablo García-González
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
| | - Claudia Olivé
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
| | | | - Josep Blázquez
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
| | - Andrea Miguel
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
| | - Raúl Núñez-Llaves
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
| | - Vanesa Pytel
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
| | - Montserrat Alegret
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Marta Marquié
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Sergi Valero
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Jose Enrique Cavazos
- South Texas Medical Science Training Program, University of Texas Health San Antonio, San Antonio, TX, USA
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center, San Antonio, TX, USA
| | - Santos Mañes
- Department of Immunology and Oncology, Centro Nacional Biotecnología (CNB-CSIC), 28049, Madrid, Spain
| | - Mercè Boada
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Agustín Ruiz
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center, San Antonio, TX, USA
| |
Collapse
|
13
|
Ding R, Wu L, Wei S, Lu H, Qin X, Liu X, Wang Y, Liu W, Li H, Luo B, Xie T, Chen Z. Multi-targeted olink proteomics analyses of cerebrospinal fluid from patients with aneurysmal subarachnoid hemorrhage. Proteome Sci 2024; 22:11. [PMID: 39604965 PMCID: PMC11600900 DOI: 10.1186/s12953-024-00236-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND The complexity of delayed cerebral ischemia (DCI) after aneurysmal subarachnoid hemorrhage (aSAH) may require the simultaneous analysis of variant types of protein biomarkers to describe it more accurately. In this study, we analyzed for the first time the alterations of cerebrospinal fluid (CSF) proteins in patients with aSAH by multi-targeted Olink proteomics, aiming to reveal the pathophysiology of DCI and provide insights into the diagnosis and treatment of aSAH. METHODS Six aSAH patients and six control patients were selected, and CSF samples were analyzed by Olink Proteomics (including 96-neurology panel and 96-inflammation panel) based on Proximity Extension Assay (PEA). Differentially expressed proteins (DEPs) were acquired and bioinformatics analysis was performed. RESULTS PCA analysis revealed better intra- and inter-group reproducibility of CSF samples in the control and aSAH groups. 23 neurology-related and 31 inflammation-relevant differential proteins were identified. In the neurology panel, compared to controls, the up-regulated proteins in the CSF of SAH patients predominantly included macrophage scavenger receptor 1 (MSR1), siglec-1, siglec-9, cathepsin C (CTSC), cathepsin S (CTSS), etc. Meanwhile, in the inflammation group, the incremental proteins mainly contained interleukin-6 (IL-6), MCP-1, CXCL10, CXCL-9, TRAIL, etc. Cluster analysis exhibited significant differences in differential proteins between the two groups. GO function enrichment analysis hinted that the differential proteins pertinent to neurology in the CSF of SAH patients were mainly involved in the regulation of defense response, vesicle-mediated transport and regulation of immune response; while the differential proteins related to inflammation were largely connected with the cellular response to chemokine, response to chemokine and chemokine-mediated signaling pathway. Additionally, in the neurology panel, KEGG enrichment analysis indicated that the differential proteins were significantly enriched in the phagosome, apoptosis and microRNAs in cancer pathway. And in the inflammation panel, the differential proteins were mainly enriched in the chemokine signaling pathway, viral protein interaction with cytokine and cytokine receptor and toll-like receptor signaling pathway. CONCLUSIONS These identified differential proteins reveal unique pathophysiological characteristics secondary to aSAH. Further characterization of these proteins and aberrant pathways in future research could enable their application as potential therapeutic targets and biomarkers for DCI after aSAH.
Collapse
Affiliation(s)
- Rui Ding
- Department of Neurosurgery, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Street, Wuhan, 430060, China
| | - Liquan Wu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Street, Wuhan, 430060, China
| | - Shanshan Wei
- Department of Oncology, Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, 430063, China
| | - Haoran Lu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Street, Wuhan, 430060, China
| | - Xiaohong Qin
- Department of Neurosurgery, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Street, Wuhan, 430060, China
| | - Xizhi Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Street, Wuhan, 430060, China
| | - Yanhua Wang
- Department of Neurosurgery, Hanchuan Renmin Hospital, Hanchuan, Hubei, 431600, China
| | - Wen Liu
- Department of Neurosurgery, Hanchuan Renmin Hospital, Hanchuan, Hubei, 431600, China
| | - Huibing Li
- Department of Neurosurgery, Hanchuan Renmin Hospital, Hanchuan, Hubei, 431600, China
| | - Baochang Luo
- Department of Neurosurgery, Hanchuan Renmin Hospital, Hanchuan, Hubei, 431600, China
| | - Teng Xie
- Department of Neurosurgery, Hanchuan Renmin Hospital, Hanchuan, Hubei, 431600, China.
| | - Zhibiao Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Street, Wuhan, 430060, China.
- Department of Neurosurgery, Hanchuan Renmin Hospital, Hanchuan, Hubei, 431600, China.
| |
Collapse
|
14
|
Beydoun MA, Beydoun HA, Li Z, Hu YH, Noren Hooten N, Ding J, Hossain S, Maino Vieytes CA, Launer LJ, Evans MK, Zonderman AB. Alzheimer's Disease polygenic risk, the plasma proteome, and dementia incidence among UK older adults. GeroScience 2024:10.1007/s11357-024-01413-8. [PMID: 39586964 DOI: 10.1007/s11357-024-01413-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 10/23/2024] [Indexed: 11/27/2024] Open
Abstract
Alzheimer's Disease (AD) is a complex polygenic neurodegenerative disorder. Its genetic risk's relationship with all-cause dementia may be influenced by the plasma proteome. Up to 40,139 UK Biobank participants aged ≥ 50y at baseline assessment (2006-2010) were followed-up for ≤ 15 y for dementia incidence. Plasma proteomics were performed on a sub-sample of UK Biobank participants (k = 1,463 plasma proteins). AD polygenic risk scores (PRS) were used as the primary exposure and Cox proportional hazards models were conducted to examine the AD PRS-dementia relationship. A four-way decomposition model then partitioned the total effect (TE) of AD PRS on dementia into an effect due to mediation only, an effect due to interaction only, neither or both. The study found that AD PRS tertiles significantly increased the risk for all-cause dementia, particularly among women. The study specifically found that AD PRS was associated with a 79% higher risk for all-cause dementia for each unit increase (HR = 1.79, 95% CI: 1.70-1.87, P < 0.001). Eighty-six plasma proteins were significantly predicted by AD PRS, including a positive association with PLA2G7, BRK1, the glial acidic fibrillary protein (GFAP), neurofilament light chain (NfL), and negative with TREM2. Both GFAP and NfL significantly interacted synergistically with AD PRS to increase all-dementia risk (> 10% of TE is pure interaction), while GFAP was also an important consistent mediator in the AD PRS-dementia relationship. In summary, we detected significant interactions of NfL and GFAP with AD PRS, in relation to dementia incidence, suggesting potential for personalized dementia prevention and management.
Collapse
Affiliation(s)
- May A Beydoun
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA.
| | - Hind A Beydoun
- VA National Center On Homelessness Among Veterans, U.S. Department of Veterans Affairs, Washington, DC, 20420, USA
- Department of Management, Policy, and Community Health, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Zhiguang Li
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Yi-Han Hu
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Jun Ding
- Translational Gerontology Branch, National Institute On Aging, NIA/NIH/IRP, Baltimore, MD, 21224, USA
| | - Sharmin Hossain
- Department of Human Services (DHS), State of Maryland, Baltimore, MD, 21202, USA
| | - Christian A Maino Vieytes
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| |
Collapse
|
15
|
Peña-Bautista C, Álvarez-Sánchez L, Balaguer Á, Raga L, García-Vallés L, Baquero M, Cháfer-Pericás C. Defining Alzheimer's Disease through Proteomic CSF Profiling. J Proteome Res 2024; 23:5096-5106. [PMID: 39373095 DOI: 10.1021/acs.jproteome.4c00590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Alzheimer disease (AD) is the main cause of dementia, and its complexity is not yet completely understood. Proteomic profiles can provide useful information to explore the pathways involved and the heterogeneity among AD patients. A proteomic analysis was performed in cerebrospinal fluid (CSF) samples from mild cognitive impairment due to AD (MCI-AD) and control individuals; both groups were classified by amyloid β42/amyloid β40 levels in CSF (data available in BioStudies database (S-BSST1456)). The analysis based on PLS regression and volcano plot identified 7 proteins (FOLR2, PPP3CA, SMOC2, STMN1, TAGLN3, TMEM132B, and UCHL1) mainly related to protein phosphorylation, structure maintenance, inflammation, and protein degradation. Enrichment analysis revealed the involvement of different biological processes related to neuronal mechanisms and synapses, lipid and carbohydrate metabolism, immune system and inflammation, vascular, hormones, and response to stimuli, and cell signaling and adhesion. In addition, the proteomic profile showed some association with the levels of AD biomarkers in CSF. Regarding the subtypes, two MCI-AD subgroups were identified: one could be related to synapsis and neuronal functions and the other to innate immunity. The study of the proteomic profile in the CSF of AD patients reflects the heterogeneity of biochemical pathways involved in AD.
Collapse
Affiliation(s)
- Carmen Peña-Bautista
- Alzheimer's Disease Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Lourdes Álvarez-Sánchez
- Alzheimer's Disease Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Ángel Balaguer
- Faculty of Mathematical Sciences, University of Valencia, 46100 Burjassot, Valencia, Spain
| | - Luis Raga
- Alzheimer's Disease Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Lorena García-Vallés
- Alzheimer's Disease Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Miguel Baquero
- 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
| |
Collapse
|
16
|
Dong Y, Song X, Wang X, Wang S, He Z. The early diagnosis of Alzheimer's disease: Blood-based panel biomarker discovery by proteomics and metabolomics. CNS Neurosci Ther 2024; 30:e70060. [PMID: 39572036 PMCID: PMC11581788 DOI: 10.1111/cns.70060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/28/2024] [Accepted: 09/10/2024] [Indexed: 11/25/2024] Open
Abstract
Diagnosis and prediction of Alzheimer's disease (AD) are increasingly pressing in the early stage of the disease because the biomarker-targeted therapies may be most effective. Diagnosis of AD largely depends on the clinical symptoms of AD. Currently, cerebrospinal fluid biomarkers and neuroimaging techniques are considered for clinical detection and diagnosis. However, these clinical diagnosis results could provide indications of the middle and/or late stages of AD rather than the early stage, and another limitation is the complexity attached to limited access, cost, and perceived invasiveness. Therefore, the prediction of AD still poses immense challenges, and the development of novel biomarkers is needed for early diagnosis and urgent intervention before the onset of obvious phenotypes of AD. Blood-based biomarkers may enable earlier diagnose and aid detection and prognosis for AD because various substances in the blood are vulnerable to AD pathophysiology. The application of a systematic biological paradigm based on high-throughput techniques has demonstrated accurate alterations of molecular levels during AD onset processes, such as protein levels and metabolite levels, which may facilitate the identification of AD at an early stage. Notably, proteomics and metabolomics have been used to identify candidate biomarkers in blood for AD diagnosis. This review summarizes data on potential blood-based biomarkers identified by proteomics and metabolomics that are closest to clinical implementation and discusses the current challenges and the future work of blood-based candidates to achieve the aim of early screening for AD. We also provide an overview of early diagnosis, drug target discovery and even promising therapeutic approaches for AD.
Collapse
Affiliation(s)
- Yun Dong
- College of PharmacyShenzhen Technology UniversityShenzhenChina
| | - Xun Song
- College of PharmacyShenzhen Technology UniversityShenzhenChina
| | - Xiao Wang
- Department of PharmacyShenzhen People's Hospital (The Second Clinical Medical College, The First Affiliated Hospital, Jinan University, Southern University of Science and Technology)ShenzhenChina
| | - Shaoxiang Wang
- School of Pharmaceutical Sciences, Health Science CenterShenzhen UniversityShenzhenChina
| | - Zhendan He
- College of PharmacyShenzhen Technology UniversityShenzhenChina
| |
Collapse
|
17
|
Aumont-Rodrigue G, Picard C, Labonté A, Poirier J. Apolipoprotein B gene expression and regulation in relation to Alzheimer's disease pathophysiology. J Lipid Res 2024; 65:100667. [PMID: 39395793 PMCID: PMC11602985 DOI: 10.1016/j.jlr.2024.100667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 10/14/2024] Open
Abstract
Apolipoprotein B (APOB), a receptor-binding protein present in cholesterol-rich lipoproteins, has been implicated in Alzheimer's disease (AD). High levels of APOB-containing low-density lipoproteins (LDL) are linked to the pathogenesis of both early-onset familial and late-onset sporadic AD. Rare coding mutations in the APOB gene are associated with familial AD, suggesting a role for APOB-bound lipoproteins in the central nervous system. This research explores APOB gene regulation across the AD spectrum using four cohorts: BRAINEAC (elderly control brains), DBCBB (controls, AD brains), ROSMAP (controls, MCI, AD brains), and ADNI (control, MCI, AD clinical subjects). APOB protein levels, measured via mass spectrometry and ELISA, positively correlated with AD pathology indices and cognition, while APOB mRNA levels showed negative correlations. Brain APOB protein levels are also correlated with cortical Aβ levels. A common coding variant in the APOB gene locus affected its expression but didn't impact AD risk or brain cholesterol concentrations, except for 24-S-hydroxycholesterol. Polymorphisms in the CYP27A1 gene, notably rs4674344, were associated with APOB protein levels. A negative correlation was observed between brain APOB gene expression and AD biomarker levels. CSF APOB correlated with Tau pathology in presymptomatic subjects, while cortical APOB was strongly associated with cortical Aβ deposition in late-stage AD. The study discusses the potential link between blood-brain barrier dysfunction and AD symptoms in relation to APOB neurobiology. Overall, APOB's involvement in lipoprotein metabolism appears to influence AD pathology across different stages of the disease.
Collapse
Affiliation(s)
- Gabriel Aumont-Rodrigue
- Douglas Mental Health University Institute, Neurosciences divison, Montréal, Québec, Canada; Centre for the Studies on Prevention of Alzheimer's Disease, Montréal, Québec, Canada; McGill University, Psychiatry department, Montréal, Québec, Canada
| | - Cynthia Picard
- Douglas Mental Health University Institute, Neurosciences divison, Montréal, Québec, Canada; Centre for the Studies on Prevention of Alzheimer's Disease, Montréal, Québec, Canada
| | - Anne Labonté
- Douglas Mental Health University Institute, Neurosciences divison, Montréal, Québec, Canada; Centre for the Studies on Prevention of Alzheimer's Disease, Montréal, Québec, Canada
| | - Judes Poirier
- Douglas Mental Health University Institute, Neurosciences divison, Montréal, Québec, Canada; Centre for the Studies on Prevention of Alzheimer's Disease, Montréal, Québec, Canada; McGill University, Psychiatry department, Montréal, Québec, Canada.
| |
Collapse
|
18
|
Theme 7 Pre-Clinical Therapeutic Strategies. Amyotroph Lateral Scler Frontotemporal Degener 2024; 25:197-217. [PMID: 39508670 DOI: 10.1080/21678421.2024.2403304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
|
19
|
Guo Q, Ping L, Dammer EB, Duong DM, Yin L, Xu K, Shantaraman A, Fox EJ, Golde TE, Johnson ECB, Roberts BR, Lah JJ, Levey AI, Seyfried NT. Heparin-enriched plasma proteome is significantly altered in Alzheimer's disease. Mol Neurodegener 2024; 19:67. [PMID: 39380021 PMCID: PMC11460197 DOI: 10.1186/s13024-024-00757-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 09/24/2024] [Indexed: 10/10/2024] Open
Abstract
INTRODUCTION Heparin binding proteins (HBPs) with roles in extracellular matrix assembly are strongly correlated to β-amyloid (Aβ) and tau pathology in Alzheimer's disease (AD) brain and cerebrospinal fluid (CSF). However, it remains challenging to detect these proteins in plasma using standard mass spectrometry-based proteomic approaches. METHODS We employed heparin-affinity chromatography, followed by off-line fractionation and tandem mass tag mass spectrometry (TMT-MS), to enrich HBPs from plasma obtained from AD (n = 62) and control (n = 47) samples. These profiles were then correlated to Aβ, tau and phosphorylated tau (pTau) CSF biomarkers and plasma pTau181 from the same individuals, as well as a consensus brain proteome network to assess the overlap with AD brain pathophysiology. RESULTS Heparin enrichment from plasma was highly reproducible, enriched well-known HBPs like APOE and thrombin, and depleted high-abundant proteins such as albumin. A total of 2865 proteins, spanning 10 orders of magnitude in abundance, were measured across 109 samples. Compared to the consensus AD brain protein co-expression network, we observed that specific plasma proteins exhibited consistent direction of change in both brain and plasma, whereas others displayed divergent changes, highlighting the complex interplay between the two compartments. Elevated proteins in AD plasma, when compared to controls, included members of the matrisome module in brain that accumulate with Aβ deposits, such as SMOC1, SMOC2, SPON1, MDK, OLFML3, FRZB, GPNMB, and the APOE4 proteoform. Additionally, heparin-enriched proteins in plasma demonstrated significant correlations with conventional AD CSF biomarkers, including Aβ, total tau, pTau, and plasma pTau181. A panel of five plasma proteins classified AD from control individuals with an area under the curve (AUC) of 0.85. When combined with plasma pTau181, the panel significantly improved the classification performance of pTau181 alone, increasing the AUC from 0.93 to 0.98. This suggests that the heparin-enriched plasma proteome captures additional variance in cognitive dementia beyond what is explained by pTau181. CONCLUSION These findings support the utility of a heparin-affinity approach coupled with TMT-MS for enriching amyloid-associated proteins, as well as a wide spectrum of plasma biomarkers that reflect pathological changes in the AD brain.
Collapse
Affiliation(s)
- Qi Guo
- Department of Biochemistry, School of Medicine, Emory School of Medicine, 505J Whitehead Biomedical Research Building, 615 Michael St, Atlanta, GA, 30322, USA
- Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Lingyan Ping
- Department of Biochemistry, School of Medicine, Emory School of Medicine, 505J Whitehead Biomedical Research Building, 615 Michael St, Atlanta, GA, 30322, USA
- Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Eric B Dammer
- Department of Biochemistry, School of Medicine, Emory School of Medicine, 505J Whitehead Biomedical Research Building, 615 Michael St, Atlanta, GA, 30322, USA
- Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Duc M Duong
- Department of Biochemistry, School of Medicine, Emory School of Medicine, 505J Whitehead Biomedical Research Building, 615 Michael St, Atlanta, GA, 30322, USA
- Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Luming Yin
- Department of Biochemistry, School of Medicine, Emory School of Medicine, 505J Whitehead Biomedical Research Building, 615 Michael St, Atlanta, GA, 30322, USA
- Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Kaiming Xu
- Department of Biochemistry, School of Medicine, Emory School of Medicine, 505J Whitehead Biomedical Research Building, 615 Michael St, Atlanta, GA, 30322, USA
- Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Anantharaman Shantaraman
- Department of Biochemistry, School of Medicine, Emory School of Medicine, 505J Whitehead Biomedical Research Building, 615 Michael St, Atlanta, GA, 30322, USA
- Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Edward J Fox
- Department of Biochemistry, School of Medicine, Emory School of Medicine, 505J Whitehead Biomedical Research Building, 615 Michael St, Atlanta, GA, 30322, USA
- Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Todd E Golde
- Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Erik C B Johnson
- Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Blaine R Roberts
- Department of Biochemistry, School of Medicine, Emory School of Medicine, 505J Whitehead Biomedical Research Building, 615 Michael St, Atlanta, GA, 30322, USA
- Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - James J Lah
- Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Allan I Levey
- Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Nicholas T Seyfried
- Department of Biochemistry, School of Medicine, Emory School of Medicine, 505J Whitehead Biomedical Research Building, 615 Michael St, Atlanta, GA, 30322, USA.
- Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
20
|
Beydoun HA, Beydoun MA, Noren Hooten N, Weiss J, Li Z, Georgescu MF, Maino Vieytes CA, Meirelles O, Launer LJ, Evans MK, Zonderman AB. Mediating and moderating effects of plasma proteomic biomarkers on the association between poor oral health problems and incident dementia: The UK Biobank study. GeroScience 2024; 46:5343-5363. [PMID: 38809392 PMCID: PMC11336161 DOI: 10.1007/s11357-024-01202-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 05/12/2024] [Indexed: 05/30/2024] Open
Abstract
The plasma proteome can mediate poor oral health problems (POHP)'s link to incident dementia. We screened 37,269 UK Biobank participants 50-74 years old (2006-2010) for prevalent POHP, further tested against 1463 plasma proteins and incident dementia over up to 15 years of follow-up. Total effect (TE) of POHP-dementia through plasma proteomic markers was decomposed into pure indirect effect (PIE), interaction referent (INTREF), controlled direct effect (CDE), or mediated interaction (INTMED). POHP increased the risk of all-cause dementia by 17% (P < 0.05). Growth differentiation factor 15 (GDF15) exhibited the strongest mediating effects (PIE > 0, P < 0.001), explaining 28% the total effect of POHP on dementia, as a pure indirect effect. A first principal component encompassing top 4 mediators (GDF15, IL19, MMP12, and ACVRL1), explained 11% of the POHP-dementia effect as a pure indirect effect. Pathway analysis including all mediators (k = 173 plasma proteins) revealed the involvement of the immune system, signal transduction, metabolism, disease, and gene expression, while STRING analysis indicated that top mediators within the first principal component were also represented in the two largest proteomic clusters. The dominant biological GO pathway for the GDF15 cluster was GO:0007169 labeled as "transmembrane receptor protein tyrosine kinase signaling pathway." Dementia is linked to POHP mediated by GDF15 among several proteomic markers.
Collapse
Affiliation(s)
- Hind A Beydoun
- US Department of Veterans Affairs, VA National Center On Homelessness Among Veterans, Washington, DC, 20420, USA
- Department of Management, Policy, and Community Health, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - May A Beydoun
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA.
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Jordan Weiss
- Stanford Center on Longevity, Stanford University, Palo Alto, CA, 94305, USA
| | - Zhiguang Li
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Michael F Georgescu
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Christian A Maino Vieytes
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Osorio Meirelles
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| |
Collapse
|
21
|
Ropri AS, Lam TG, Kalia V, Buchanan HM, Bartosch AMW, Youth EHH, Xiao H, Ross SK, Jain A, Chakrabarty JK, Kang MS, Boyett D, Spinazzi EF, Iodice G, McGovern RA, Honig LS, Brown LM, Miller GW, McKhann GM, Teich AF. Alzheimer's disease CSF biomarkers correlate with early pathology and alterations in neuronal and glial gene expression. Alzheimers Dement 2024; 20:7090-7103. [PMID: 39192661 PMCID: PMC11485399 DOI: 10.1002/alz.14194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 07/18/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024]
Abstract
INTRODUCTION Normal pressure hydrocephalus (NPH) patients undergoing cortical shunting frequently show early Alzheimer's disease (AD) pathology on cortical biopsy, which is predictive of progression to clinical AD. The objective of this study was to use samples from this cohort to identify cerebrospinal fluid (CSF) biomarkers for AD-related central nervous system (CNS) pathophysiologic changes using tissue and fluids with early pathology, free of post mortem artifact. METHODS We analyzed Simoa, proteomic, and metabolomic CSF data from 81 patients with previously documented pathologic and transcriptomic changes. RESULTS AD pathology on biopsy correlates with CSF β-amyloid-42/40, neurofilament light chain (NfL), and phospho-tau-181(p-tau181)/β-amyloid-42, while several gene expression modules correlate with NfL. Proteomic analysis highlights seven core proteins that correlate with pathology and gene expression changes on biopsy, and metabolomic analysis of CSF identifies disease-relevant groups that correlate with biopsy data. DISCUSSION As additional biomarkers are added to AD diagnostic panels, our work provides insight into the CNS pathophysiology these markers are tracking. HIGHLIGHTS AD CSF biomarkers correlate with CNS pathology and transcriptomic changes. Seven proteins correlate with CNS pathology and gene expression changes. Inflammatory and neuronal gene expression changes correlate with YKL-40 and NPTXR, respectively. CSF metabolomic analysis identifies pathways that correlate with biopsy data. Fatty acid metabolic pathways correlate with β-amyloid pathology.
Collapse
Affiliation(s)
- Ali S. Ropri
- Department of Pathology and Cell BiologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Tiffany G. Lam
- Department of Pathology and Cell BiologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Vrinda Kalia
- Department of Environmental Health Sciences, Mailman School of Public HealthColumbia UniversityNew YorkNew YorkUSA
| | - Heather M. Buchanan
- Department of Pathology and Cell BiologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Anne Marie W. Bartosch
- Department of Pathology and Cell BiologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Elliot H. H. Youth
- Department of Pathology and Cell BiologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Harrison Xiao
- Department of Pathology and Cell BiologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Sophie K. Ross
- Department of Pathology and Cell BiologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Anu Jain
- Quantitative Proteomics and Metabolomics Center, Department of Biological SciencesColumbia UniversityNew YorkNew YorkUSA
| | - Jayanta K. Chakrabarty
- Quantitative Proteomics and Metabolomics Center, Department of Biological SciencesColumbia UniversityNew YorkNew YorkUSA
| | - Min Suk Kang
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
- Department of NeurologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Deborah Boyett
- Department of NeurosurgeryColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Eleonora F. Spinazzi
- Department of NeurosurgeryColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Gail Iodice
- Ankyra TherapeuticsCambridgeMassachusettsUSA
| | - Robert A. McGovern
- Department of NeurosurgeryUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Lawrence S. Honig
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
- Department of NeurologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Lewis M. Brown
- Quantitative Proteomics and Metabolomics Center, Department of Biological SciencesColumbia UniversityNew YorkNew YorkUSA
| | - Gary W. Miller
- Department of Environmental Health Sciences, Mailman School of Public HealthColumbia UniversityNew YorkNew YorkUSA
| | - Guy M. McKhann
- Department of NeurosurgeryColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Andrew F. Teich
- Department of Pathology and Cell BiologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
- Department of NeurologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
| |
Collapse
|
22
|
Guo Y, Chen SD, You J, Huang SY, Chen YL, Zhang Y, Wang LB, He XY, Deng YT, Zhang YR, Huang YY, Dong Q, Feng JF, Cheng W, Yu JT. Multiplex cerebrospinal fluid proteomics identifies biomarkers for diagnosis and prediction of Alzheimer's disease. Nat Hum Behav 2024; 8:2047-2066. [PMID: 38987357 DOI: 10.1038/s41562-024-01924-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 06/10/2024] [Indexed: 07/12/2024]
Abstract
Recent expansion of proteomic coverage opens unparalleled avenues to unveil new biomarkers of Alzheimer's disease (AD). Among 6,361 cerebrospinal fluid (CSF) proteins analysed from the ADNI database, YWHAG performed best in diagnosing both biologically (AUC = 0.969) and clinically (AUC = 0.857) defined AD. Four- (YWHAG, SMOC1, PIGR and TMOD2) and five- (ACHE, YWHAG, PCSK1, MMP10 and IRF1) protein panels greatly improved the accuracy to 0.987 and 0.975, respectively. Their superior performance was validated in an independent external cohort and in discriminating autopsy-confirmed AD versus non-AD, rivalling even canonical CSF ATN biomarkers. Moreover, they effectively predicted the clinical progression to AD dementia and were strongly associated with AD core biomarkers and cognitive decline. Synaptic, neurogenic and infectious pathways were enriched in distinct AD stages. Mendelian randomization did not support the significant genetic link between CSF proteins and AD. Our findings revealed promising high-performance biomarkers for AD diagnosis and prediction, with implications for clinical trials targeting different pathomechanisms.
Collapse
Affiliation(s)
- Yu Guo
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shi-Dong Chen
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jia You
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Shu-Yi Huang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi-Lin Chen
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lin-Bo Wang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Xiao-Yu He
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yue-Ting Deng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ya-Ru Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu-Yuan Huang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Wei Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
23
|
Zhao J, Lu F, Yu H, Cao J, Su Z, Zhao J. Proteomic analysis of baicalin intervention on protein expression and modification in the hippocampus of Alzheimer's disease model rat. Int J Neurosci 2024:1-10. [PMID: 39136404 DOI: 10.1080/00207454.2024.2332963] [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: 02/20/2024] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 09/03/2024]
Abstract
OBJECTIVE We aimed to explore the treatment effect and therapeutic mechanisms of baicalin in Alzheimer's disease (AD). METHODS The AD rat model was established by intracerebroventricular injection of Aβ1-40, with rats in the baicalin group receiving baicalin intraventricular injections. Morris Water Maze and Hematoxylin-eosin (H&E) Staining were employed to detect the successful model construction and baicalin treatment effect. The proteins extracted from the hippocampus were subjected to proteomics analysis. Bioinformatics technology was employed for differential protein screening, functional classification, and enrichment. Western Blot was employed to validate the expressions of differentially expressed proteins (DEPs) and the protein modification alternations. RESULTS Water maze test confirmed the successful AD model construction and baicalin can improve learning and memory abilities. A total of 26 DEPs associated with 28 Gene Ontology (GO) functions were identified in the model and 32 DEPs were obtained between the baicalin group and the model. Bioinformatics analysis demonstrated that AD occurrence resulted in neuronal dysfunction and was associated with immune responses. The baicalin therapeutic effect on AD may be associated with metabolic processes, vitamin response, angiogenesis regulation, and fatty acid response. Immunoglobulin heavy constant mu (Ighm) and Immunoglobulin G2a (IgG2a) exhibited significant increases in AD and baicalin attenuated their expressions, while Fatty acid desaturase 1 (Fads1) exhibited a significantly diminished expression and baicalin could reverse the trend. Succinylation detection exhibited the differentially expressed at 35 kD between the model and baicalin group. CONCLUSION Baicalin intervention may ameliorate cognitive impairment in AD rats by modulating the expressions of proteins and the succinylation modifications.
Collapse
Affiliation(s)
- Jiwei Zhao
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Fan Lu
- Shandong Public Health Clinical Center, Shandong University, Jinan, Shandong, China
| | - Hongli Yu
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Jingwei Cao
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Zhiqiang Su
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Jingkun Zhao
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| |
Collapse
|
24
|
Hok-A-Hin YS, Vermunt L, Peeters CF, van der Ende EL, de Boer SC, Meeter LH, van Swieten JC, Hu WT, Lleó A, Alcolea D, Engelborghs S, Sieben A, Chen-Plotkin A, Irwin DJ, van der Flier WM, Pijnenburg YA, Teunissen CE, del Campo M. Large-scale CSF proteome profiling identifies biomarkers for accurate diagnosis of Frontotemporal Dementia. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.19.24312100. [PMID: 39228745 PMCID: PMC11370532 DOI: 10.1101/2024.08.19.24312100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Diagnosis of Frontotemporal dementia (FTD) and the specific underlying neuropathologies (frontotemporal lobar degeneration; FTLD- Tau and FTLD-TDP) is challenging, and thus fluid biomarkers are needed to improve diagnostic accuracy. We used proximity extension assays to analyze 665 proteins in cerebrospinal fluid (CSF) samples from a multicenter cohort including patients with FTD (n = 189), Alzheimer's Disease dementia (AD; n = 232), and cognitively unimpaired individuals (n = 196). In a subset, FTLD neuropathology was determined based on phenotype or genotype (FTLD-Tau = 87 and FTLD-TDP = 68). Forty three proteins were differentially regulated in FTD compared to controls and AD, reflecting axon development, regulation of synapse assembly, and cell-cell adhesion mediator activity pathways. Classification analysis identified a 14- and 13-CSF protein panel that discriminated FTD from controls (AUC: 0.96) or AD (AUC: 0.91). Custom multiplex panels confirmed the highly accurate discrimination between FTD and controls (AUCs > 0.96) or AD (AUCs > 0.88) in three validation cohorts, including one with autopsy confirmation (AUCs > 0.90). Six proteins were differentially regulated between FTLD-TDP and FTLD-Tau, but no reproducible classification model could be generated (AUC: 0.80). Overall, this study introduces novel FTD-specific biomarker panels with potential use in diagnostic setting.
Collapse
Affiliation(s)
- Yanaika S. Hok-A-Hin
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Lisa Vermunt
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Alzheimer Center and Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
| | - Carel F.W. Peeters
- Mathematical & Statistical Methods group – Biometris, Wageningen University & Research, Wageningen, The Netherlands
| | - Emma L. van der Ende
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Sterre C.M. de Boer
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Alzheimer Center and Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
- School of Psychology and Brain & Mind Centre, The University of Sydney, Sydney, Australia
| | - Lieke H. Meeter
- Alzheimer center and department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - John C. van Swieten
- Alzheimer center and department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - William T. Hu
- Department of Neurology, Center for Neurodegenerative Diseases Research, Emory University School of Medicine, Atlanta, USA
| | - Alberto Lleó
- Department of Neurology, Institut d’Investigacions Biomèdiques Sant Pau (IIB SANT PAU) - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Hospital de la Santa Creu i Sant Pau, Barcelona, Catalunya, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Daniel Alcolea
- Department of Neurology, Institut d’Investigacions Biomèdiques Sant Pau (IIB SANT PAU) - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Hospital de la Santa Creu i Sant Pau, Barcelona, Catalunya, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Sebastiaan Engelborghs
- Reference Center for Biological Markers of Dementia (BIODEM), Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Vrije Universiteit Brussel, Center for Neurosciences (C4N), Neuroprotection and Neuromodulation Research Group (NEUR), Brussels, Belgium
- Universitair Ziekenhuis Brussel, Department of Neurology, Brussels, Belgium
| | - Anne Sieben
- Lab of neuropathology, Neurobiobank, Institute Born-Bunge, Antwerp University, Edegem, Belgium
| | - Alice Chen-Plotkin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David J. Irwin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wiesje M. van der Flier
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Alzheimer Center and Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
| | - Yolande A.L. Pijnenburg
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Alzheimer Center and Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
| | - Charlotte E. Teunissen
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Marta del Campo
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
- Barcelonaßeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San PabloCEU, CEU Universities, Madrid, Spain
| |
Collapse
|
25
|
Hu Q, Shi M, Li Y, Zhao X. Elevated plasma neurofilament light was associated with multi-modal neuroimaging features in Alzheimer's disease signature regions and predicted future tau deposition. BMC Neurol 2024; 24:236. [PMID: 38971733 PMCID: PMC11227162 DOI: 10.1186/s12883-024-03728-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 06/14/2024] [Indexed: 07/08/2024] Open
Abstract
BACKGROUND Neurofilament Light (NfL) is a biomarker for early neurodegeneration in Alzheimer's disease (AD). This study aims to examine the association between plasma NfL and multi-modal neuroimaging features across the AD spectrum and whether NfL predicts future tau deposition. METHODS The present study recruited 517 participants comprising Aβ negative cognitively normal (CN-) participants (n = 135), Aβ positive cognitively normal (CN +) participants (n = 64), individuals with amnestic mild cognitive impairment (aMCI) (n = 212), and those diagnosed with AD dementia (n = 106). All the participants underwent multi-modal neuroimaging examinations. Cross-sectional and longitudinal associations between plasma NfL and multi-modal neuro-imaging features were evaluated using partial correlation analysis and linear mixed effects models. We also used linear regression analysis to investigate the association of baseline plasma NfL with future PET tau load. Mediation analysis was used to explore whether the effect of NfL on cognition was mediated by these imaging biomarkers. RESULTS The results showed that baseline NfL levels and the rate of change were associated with Aβ deposition, brain atrophy, brain connectome, glucose metabolism, and brain perfusion in AD signature regions (P<0.05). In both Aβ positive CN and MCI participants, baseline NfL showed a significant predictive value of elevating tau burden in the left medial orbitofrontal cortex and para-hippocampus (β = 0.336, P = 0.032; β = 0.313, P = 0.047). Lastly, the multi-modal neuroimaging features mediated the association between plasma NfL and cognitive performance. CONCLUSIONS The study supports the association between plasma NfL and multi-modal neuroimaging features in AD-vulnerable regions and its predictive value for future tau deposition.
Collapse
Affiliation(s)
- Qili Hu
- Department of Imaging, The Fifth People's Hospital of Shanghai, Fudan University, No.128 Ruili Road, Minhang District, Shanghai, 200240, China
| | - Mengqiu Shi
- Department of Imaging, The Fifth People's Hospital of Shanghai, Fudan University, No.128 Ruili Road, Minhang District, Shanghai, 200240, China
| | - Yunfei Li
- Department of Imaging, The Fifth People's Hospital of Shanghai, Fudan University, No.128 Ruili Road, Minhang District, Shanghai, 200240, China
| | - Xiaohu Zhao
- Department of Imaging, The Fifth People's Hospital of Shanghai, Fudan University, No.128 Ruili Road, Minhang District, Shanghai, 200240, China.
| |
Collapse
|
26
|
Beydoun MA, Beydoun HA, Hu YH, Maino Vieytes CA, Noren Hooten N, Song M, Georgescu MF, Fanelli-Kuczmarski MT, Meirelles O, Launer LJ, Evans MK, Zonderman AB. Plasma proteomic biomarkers and the association between poor cardiovascular health and incident dementia: The UK Biobank study. Brain Behav Immun 2024; 119:995-1007. [PMID: 38710337 PMCID: PMC11285716 DOI: 10.1016/j.bbi.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/04/2024] [Accepted: 05/02/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND The study examined how plasma proteome indicators may explain the link between poor cardiovascular health (CVH) and dementia risk. METHODS The present study involved 28,974 UK Biobank participants aged 50-74y at baseline (2006-2010) who were followed-up for ≤ 15 y for incidence of dementia. CVH was calculated using Life's Essential 8 (LE8) total scores. The scores were standardized and reverse coded to reflect poor CVH (LE8z_rev). OLINK proteomics was available on this sample (k = 1,463 plasma proteins). The study primarily tested the mediating effects of the plasma proteome in LE8z_rev-dementia effect. The total effect was decomposed into "mediation only" or pure indirect effect (PIE), "interaction only" or interaction referent (INTREF), "neither mediation nor interaction" or controlled direct effect (CDE), and "both mediation and interaction" or mediated interaction (INTMED). RESULTS The study found poorer CVH assessed by LE8z_rev increased the risk of all-cause dementia by 11 % [per 1 SD, hazard ratio, (HR) = 1.11, 95 % CI: 1.03-1.20, p = 0.005). The study identified 11 plasma proteins with strong mediating effects, with GDF15 having the strongest association with dementia risk (per 1 SD, HR = 1.24, 95 % CI: 1.16, 1.33, P < 0.001 when LE8z_rev is set at its mean value) and the largest proportion mediated combining PIE and INTMED (62.6 %; 48 % of TE is PIE), followed by adrenomedullin or ADM. A first principal component with 10 top mediators (TNFRSF1A, GDF15, FSTL3, COL6A3, PLAUR, ADM, GFRAL, ACVRL1, TNFRSF6B, TGFA) mediated 53.6 % of the LE8z_rev-dementia effect. Using all the significant PIE (k = 526) proteins, we used OLINK Insight pathway analysis to identify key pathways, which revealed the involvement of the immune system, signal transduction, metabolism, disease, protein metabolism, hemostasis, membrane trafficking, extracellular matrix organization, developmental biology, and gene expression among others. STRING analysis revealed that five top consistent proteomic mediators were represented in two larger clusters reflecting numerous interconnected biological gene ontology pathways, most notably cytokine-mediated signaling pathway for GDF15 cluster (GO:0019221) and regulation of peptidyl-tyrosine phosphorylation for the ADM cluster (GO:0050730). CONCLUSION Dementia is linked to poor CVH mediated by GDF15 and ADM among several key proteomic markers which collectively explained ∼ 54 % of the total effect.
Collapse
Affiliation(s)
- May A Beydoun
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States.
| | - Hind A Beydoun
- VA National Center on Homelessness Among Veterans, U.S. Department of Veterans Affairs, Washington, DC 20420, United States; Department of Management, Policy, and Community Health, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX 77030, United States
| | - Yi-Han Hu
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States
| | - Christian A Maino Vieytes
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States
| | - Minkyo Song
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States
| | - Michael F Georgescu
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States
| | - Marie T Fanelli-Kuczmarski
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States
| | - Osorio Meirelles
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States
| | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States
| |
Collapse
|
27
|
Jäkel L, De Kort AM, Stellingwerf A, Hernández Utrilla C, Kersten I, Vervuurt M, Vermeiren Y, Küsters B, Schreuder FHBM, Klijn CJM, Kuiperij HB, Verbeek MM. Altered brain expression and cerebrospinal fluid levels of TIMP4 in cerebral amyloid angiopathy. Acta Neuropathol Commun 2024; 12:103. [PMID: 38915119 PMCID: PMC11194996 DOI: 10.1186/s40478-024-01823-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: 04/19/2024] [Accepted: 06/17/2024] [Indexed: 06/26/2024] Open
Abstract
Cerebral amyloid angiopathy (CAA) is a highly prevalent and progressive pathology, involving amyloid-β (Aβ) deposition in the cerebral blood vessel walls. CAA is associated with an increased risk for intracerebral hemorrhages (ICH). Insight into the molecular mechanisms associated with CAA pathology is urgently needed, to develop additional diagnostic tools to allow for reliable and early diagnosis of CAA and to obtain novel leads for the development of targeted therapies. Tissue inhibitor of matrix metalloproteinases 4 (TIMP4) is associated with cardiovascular functioning and disease and has been linked to vascular dementia. Using immunohistochemistry, we studied occipital brain tissue samples of 57 patients with CAA (39 without ICH and 18 with ICH) and 42 controls, and semi-quantitatively assessed expression levels of TIMP4. Patients with CAA had increased vascular expression of TIMP4 compared to controls (p < 0.001), and in these patients, TIMP4 expression correlated with CAA severity (τb = 0.38; p = 0.001). Moreover, TIMP4 expression was higher in CAA-ICH compared to CAA-non-ICH cases (p = 0.024). In a prospective cross-sectional study of 38 patients with CAA and 37 age- and sex-matched controls, we measured TIMP4 levels in cerebrospinal fluid (CSF) and serum using ELISA. Mean CSF levels of TIMP4 were decreased in patients with CAA compared to controls (3.36 ± 0.20 vs. 3.96 ± 0.22 ng/ml, p = 0.033), whereas median serum levels were increased in patients with CAA (4.51 ng/ml [IQR 3.75-5.29] vs 3.60 ng/ml [IQR 3.11-4.85], p-9.013). Moreover, mean CSF TIMP4 levels were lower in CAA patients who had experienced a symptomatic hemorrhage compared to CAA patients who did not (2.13 ± 0.24 vs. 3.57 ± 0.24 ng/ml, p = 0.007). CSF TIMP4 levels were associated with CSF levels of Aβ40 (spearman r (rs) = 0.321, p = 0.009). In summary, we show that TIMP4 is highly associated with CAA and CAA-related ICH, which is reflected by higher levels in the cerebral vasculature and lower levels in CSF. With these findings we provide novel insights into the pathophysiology of CAA, and more specifically in CAA-associated ICH.
Collapse
Affiliation(s)
- Lieke Jäkel
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 830 TML, P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
- Radboud Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anna M De Kort
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 830 TML, P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
- Radboud Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Arno Stellingwerf
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 830 TML, P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
- Radboud Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Carla Hernández Utrilla
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 830 TML, P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
- Radboud Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Iris Kersten
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 830 TML, P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Marc Vervuurt
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 830 TML, P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
- Radboud Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Yannick Vermeiren
- Division of Human Nutrition and Health, Chair Group Nutritional Biology, Wageningen University and Research (WUR), Wageningen, The Netherlands
- Faculty of Medicine and Health Sciences, Translational Neurosciences, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Benno Küsters
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Floris H B M Schreuder
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 830 TML, P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Catharina J M Klijn
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 830 TML, P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - H Bea Kuiperij
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 830 TML, P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
- Radboud Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marcel M Verbeek
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 830 TML, P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands.
- Radboud Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
28
|
Hällqvist J, Bartl M, Dakna M, Schade S, Garagnani P, Bacalini MG, Pirazzini C, Bhatia K, Schreglmann S, Xylaki M, Weber S, Ernst M, Muntean ML, Sixel-Döring F, Franceschi C, Doykov I, Śpiewak J, Vinette H, Trenkwalder C, Heywood WE, Mills K, Mollenhauer B. Plasma proteomics identify biomarkers predicting Parkinson's disease up to 7 years before symptom onset. Nat Commun 2024; 15:4759. [PMID: 38890280 PMCID: PMC11189460 DOI: 10.1038/s41467-024-48961-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 05/20/2024] [Indexed: 06/20/2024] Open
Abstract
Parkinson's disease is increasingly prevalent. It progresses from the pre-motor stage (characterised by non-motor symptoms like REM sleep behaviour disorder), to the disabling motor stage. We need objective biomarkers for early/pre-motor disease stages to be able to intervene and slow the underlying neurodegenerative process. Here, we validate a targeted multiplexed mass spectrometry assay for blood samples from recently diagnosed motor Parkinson's patients (n = 99), pre-motor individuals with isolated REM sleep behaviour disorder (two cohorts: n = 18 and n = 54 longitudinally), and healthy controls (n = 36). Our machine-learning model accurately identifies all Parkinson patients and classifies 79% of the pre-motor individuals up to 7 years before motor onset by analysing the expression of eight proteins-Granulin precursor, Mannan-binding-lectin-serine-peptidase-2, Endoplasmatic-reticulum-chaperone-BiP, Prostaglaindin-H2-D-isomaerase, Interceullular-adhesion-molecule-1, Complement C3, Dickkopf-WNT-signalling pathway-inhibitor-3, and Plasma-protease-C1-inhibitor. Many of these biomarkers correlate with symptom severity. This specific blood panel indicates molecular events in early stages and could help identify at-risk participants for clinical trials aimed at slowing/preventing motor Parkinson's disease.
Collapse
Affiliation(s)
- Jenny Hällqvist
- UCL Institute of Child Health and Great Ormond Street Hospital, London, UK.
- UCL Queen Square Institute of Neurology, Clinical and Movement Neurosciences, London, UK.
| | - Michael Bartl
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany.
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Goettingen, Goettingen, Germany.
| | - Mohammed Dakna
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
| | | | - Paolo Garagnani
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | | | - Chiara Pirazzini
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Kailash Bhatia
- National Hospital for Neurology & Neurosurgery, Queen Square, WC1N3BG, London, UK
| | | | - Mary Xylaki
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
| | - Sandrina Weber
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
| | - Marielle Ernst
- Institute of Diagnostic and Interventional Neuroradiology, University Medical Center Goettingen, Goettingen, Germany
| | | | - Friederike Sixel-Döring
- Paracelsus-Elena-Klinik, Kassel, Germany
- Department of Neurology, Philipps-University, Marburg, Germany
| | - Claudio Franceschi
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Ivan Doykov
- UCL Institute of Child Health and Great Ormond Street Hospital, London, UK
| | - Justyna Śpiewak
- UCL Institute of Child Health and Great Ormond Street Hospital, London, UK
| | - Héloїse Vinette
- UCL Institute of Child Health and Great Ormond Street Hospital, London, UK
- UCL: Food, Microbiomes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Claudia Trenkwalder
- Paracelsus-Elena-Klinik, Kassel, Germany
- Department of Neurosurgery, University Medical Center Goettingen, Goettingen, Germany
| | - Wendy E Heywood
- UCL Institute of Child Health and Great Ormond Street Hospital, London, UK
| | - Kevin Mills
- UCL Queen Square Institute of Neurology, Clinical and Movement Neurosciences, London, UK
| | - Brit Mollenhauer
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
- Paracelsus-Elena-Klinik, Kassel, Germany
| |
Collapse
|
29
|
Ropri AS, Lam TG, Kalia V, Buchanan HM, Bartosch AMW, Youth EHH, Xiao H, Ross SK, Jain A, Chakrabarty JK, Kang MS, Boyett D, Spinazzi EF, Iodice G, McGovern RA, Honig LS, Brown LM, Miller GW, McKhann GM, Teich AF. Alzheimer's disease CSF biomarkers correlate with early pathology and alterations in neuronal and glial gene expression. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.11.24308706. [PMID: 38947015 PMCID: PMC11213077 DOI: 10.1101/2024.06.11.24308706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
INTRODUCTION Normal pressure hydrocephalus (NPH) patients undergoing cortical shunting frequently show early AD pathology on cortical biopsy, which is predictive of progression to clinical AD. The objective of this study was to use samples from this cohort to identify CSF biomarkers for AD-related CNS pathophysiologic changes using tissue and fluids with early pathology, free of post-mortem artifact. METHODS We analyzed Simoa, proteomic, and metabolomic CSF data from 81 patients with previously documented pathologic and transcriptomic changes. RESULTS AD pathology on biopsy correlates with CSF β-amyloid-40/42, neurofilament light chain (NfL), and phospho-tau-181(p-tau181)/β-amyloid-42, while several gene expression modules correlate with NfL. Proteomic analysis highlights 7 core proteins that correlate with pathology and gene expression changes on biopsy, and metabolomic analysis of CSF identifies disease-relevant groups that correlate with biopsy data.. DISCUSSION As additional biomarkers are added to AD diagnostic panels, our work provides insight into the CNS pathophysiology these markers are tracking.
Collapse
Affiliation(s)
- Ali S. Ropri
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Tiffany G. Lam
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Vrinda Kalia
- Dept. of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY 10032, USA
| | - Heather M. Buchanan
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Anne Marie W. Bartosch
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Elliot H. H. Youth
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Harrison Xiao
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Sophie K. Ross
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Anu Jain
- Quantitative Proteomics and Metabolomics Center, Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Jayanta K. Chakrabarty
- Quantitative Proteomics and Metabolomics Center, Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Min Suk Kang
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Neurology, Columbia University, New York, NY 10032, USA
| | - Deborah Boyett
- Department of Neurosurgery, Columbia University, New York, NY 10032, USA
| | | | - Gail Iodice
- Ankyra Therapeutics, Cambridge, MA 02142, USA
| | - Robert A. McGovern
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA
| | - Lawrence S. Honig
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Neurology, Columbia University, New York, NY 10032, USA
| | - Lewis M. Brown
- Quantitative Proteomics and Metabolomics Center, Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Gary W. Miller
- Dept. of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY 10032, USA
| | - Guy M. McKhann
- Department of Neurosurgery, Columbia University, New York, NY 10032, USA
| | - Andrew F. Teich
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Neurology, Columbia University, New York, NY 10032, USA
| |
Collapse
|
30
|
Peng T, Yang Y, Ma J, Xu P, Xie X, Hu N, Yan Y. Dementia and metabolic syndrome: a bibliometric analysis. Front Aging Neurosci 2024; 16:1400589. [PMID: 38934020 PMCID: PMC11199533 DOI: 10.3389/fnagi.2024.1400589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Background Dementia is a progressive neurodegenerative condition, while metabolic syndrome (MetS) is characterized by a combination of metabolic abnormalities such as hypertension, high blood sugar, and obesity. There exists a connection and overlap between the two conditions in certain aspects, and both are influenced to varying degrees by the process of aging. This study presents an overview of the current research landscape regarding dementia and MetS through bibliometric analysis. Methods A systematic search was conducted to retrieve relevant literature on dementia and MetS published between 1 January 2000, and 30 November 2023, from the Web of Science Core Collection database. Various bibliometric tools, including VOSviewer, CiteSpace, and the R software package "bibliometrix," were utilized for analysis. Results A total of 717 articles were identified, showing an upward trend in annual publications. Leading contributors included the United States, Italy, and China, with institutions such as the University of California System at the forefront. The Journal of Alzheimer's Disease emerged as the top publisher, while research published in Neurology garnered significant citations. Noteworthy authors encompassed Panza, Francesco; Frisardi, Vincenza; and Feldman, Eva L, with Kristine Yaffe being the most cited author (280 citations). Recent studies have focused on themes like "gut microbiota," "neuroinflammation," "fatty acids," and "microglia." Conclusion This bibliometric analysis summarizes the foundational knowledge structure in the realm of dementia and MetS from 2000 to 2023. By highlighting current research frontiers and trending topics, this analysis serves as a valuable reference for researchers in the field.
Collapse
Affiliation(s)
- Tao Peng
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Yifan Yang
- Department of Encephalopathy, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Jingying Ma
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Peili Xu
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Xinchun Xie
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Nan Hu
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Yongmei Yan
- Department of Encephalopathy, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| |
Collapse
|
31
|
Hristovska I, Binette AP, Kumar A, Gaiteri C, Karlsson L, Strandberg O, Janelidze S, van Westen D, Stomrud E, Palmqvist S, Ossenkoppele R, Mattsson-Carlgren N, Vogel JW, Hansson O. Identification of distinct and shared biomarker panels in different manifestations of cerebral small vessel disease through proteomic profiling. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.10.24308599. [PMID: 38947084 PMCID: PMC11213103 DOI: 10.1101/2024.06.10.24308599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The pathophysiology underlying various manifestations of cerebral small vessel disease (cSVD) remains obscure. Using cerebrospinal fluid proximity extension assays and co-expression network analysis of 2,943 proteins, we found common and distinct proteomic signatures between white matter lesions (WML), microbleeds and infarcts measured in 856 living patients, and validated WML-associated proteins in three additional datasets. Proteins indicative of extracellular matrix dysregulation and vascular remodeling, including ELN, POSTN, CCN2 and MMP12 were elevated across all cSVD manifestations, with MMP12 emerging as an early cSVD indicator. cSVD-associated proteins formed a co-abundance network linked to metabolism and enriched in endothelial and arterial smooth muscle cells, showing elevated levels at early disease manifestations. Later disease stages involved changes in microglial proteins, associated with longitudinal WML progression, and changes in neuronal proteins mediating WML-associated cognitive decline. These findings provide an atlas of novel cSVD biomarkers and a promising roadmap for the next generation of cSVD therapeutics.
Collapse
Affiliation(s)
- Ines Hristovska
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Atul Kumar
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Chris Gaiteri
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA
- Rush University Alzheimer's Disease Center, Rush University, Chicago IL, USA
| | - Linda Karlsson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Danielle van Westen
- Diagnostic Radiology, Department of Clinical Sciences Lund, Lund University
- Imaging and Function, Skåne University Hospital, Lund, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Jacob W Vogel
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Department of Clinical Sciences, Malmö, SciLifeLab, Lund University, Lund, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
32
|
Hidese S. Search for cerebrospinal fluid biomarkers in patients with major psychiatric disorders: Multiplex immunoassay findings and proximity extension assay prospects. Neuropsychopharmacol Rep 2024; 44:314-320. [PMID: 38686540 PMCID: PMC11144604 DOI: 10.1002/npr2.12439] [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: 01/08/2024] [Revised: 03/05/2024] [Accepted: 03/24/2024] [Indexed: 05/02/2024] Open
Abstract
Multiplex immunoassays have been developed to detect multiple proteins simultaneously and are used to search for biomarkers, including those present in major psychiatric disorders. This study aimed to review multiplex immunoassay studies on cerebrospinal fluid (CSF) biomarkers in patients with schizophrenia, bipolar disorder (BD), and major depressive disorder (MDD) and examine future research directions using improved proteomic techniques. According to the results of previous multiplex immunoassay studies, increased CSF IFN-β, IL-8, MCP-2, MMP-2, PAI-1, sICAM-1, and sVCAM-1 and decreased CSF ACE, APP, fibrinogen, and GDNF were observed in patients with schizophrenia, while CSF HGF and S100B were positively correlated with psychotic symptom and CSF IL-11, IL-29/IFN-λ1, and TSLP were negatively correlated. Increased CSF IFN-β and IL-1β and decreased CSF Aβ42, APP, IL-6, and NCAM-1 were observed, while CSF S100B was positively correlated with manic symptom in patients with BD. Increased CSF IL-4, MCP-1, MIP-1β, and MMP-2 were observed in patients with MDD, while CSF HGF and MMP-2 were positively correlated with depressive symptom and CSF IL-15 and MCP-1 were negatively correlated. However, signal cross-talk and cross-reactivity problems have been observed in previous studies using multiplex immunoassay. The proximity extension assay can be used to overcome cross-reactivity and enable ultrasensitive multiplexed detection and quantification of more than 1000 target proteins. However, proteomic studies using proximity extension assay technology in patients with schizophrenia, BD, or MDD are still scarce. Therefore, future high-quality proteomic studies are required to identify CSF biomarkers for larger sets of target proteins in patients with major psychiatric disorders.
Collapse
Affiliation(s)
- Shinsuke Hidese
- Department of PsychiatryTeikyo University School of MedicineTokyoJapan
- Department of Mental Disorder Research, National Center of Neurology and PsychiatryNational Institute of NeuroscienceKodaira, TokyoJapan
| |
Collapse
|
33
|
Seitz-Holland J, Alemán-Gómez Y, Cho KIK, Pasternak O, Cleusix M, Jenni R, Baumann PS, Klauser P, Conus P, Hagmann P, Do KQ, Kubicki M, Dwir D. Matrix metalloproteinase 9 (MMP-9) activity, hippocampal extracellular free water, and cognitive deficits are associated with each other in early phase psychosis. Neuropsychopharmacology 2024; 49:1140-1150. [PMID: 38431757 PMCID: PMC11109110 DOI: 10.1038/s41386-024-01814-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 12/18/2023] [Accepted: 01/29/2024] [Indexed: 03/05/2024]
Abstract
Increasing evidence points toward the role of the extracellular matrix, specifically matrix metalloproteinase 9 (MMP-9), in the pathophysiology of psychosis. MMP-9 is a critical regulator of the crosstalk between peripheral and central inflammation, extracellular matrix remodeling, hippocampal development, synaptic pruning, and neuroplasticity. Here, we aim to characterize the relationship between plasma MMP-9 activity, hippocampal microstructure, and cognition in healthy individuals and individuals with early phase psychosis. We collected clinical, blood, and structural and diffusion-weighted magnetic resonance imaging data from 39 individuals with early phase psychosis and 44 age and sex-matched healthy individuals. We measured MMP-9 plasma activity, hippocampal extracellular free water (FW) levels, and hippocampal volumes. We used regression analyses to compare MMP-9 activity, hippocampal FW, and volumes between groups. We then examined associations between MMP-9 activity, FW levels, hippocampal volumes, and cognitive performance assessed with the MATRICS battery. All analyses were controlled for age, sex, body mass index, cigarette smoking, and years of education. Individuals with early phase psychosis demonstrated higher MMP-9 activity (p < 0.0002), higher left (p < 0.05) and right (p < 0.05) hippocampal FW levels, and lower left (p < 0.05) and right (p < 0.05) hippocampal volume than healthy individuals. MMP-9 activity correlated positively with hippocampal FW levels (all participants and individuals with early phase psychosis) and negatively with hippocampal volumes (all participants and healthy individuals). Higher MMP-9 activity and higher hippocampal FW levels were associated with slower processing speed and worse working memory performance in all participants. Our findings show an association between MMP-9 activity and hippocampal microstructural alterations in psychosis and an association between MMP-9 activity and cognitive performance. Further, more extensive longitudinal studies should examine the therapeutic potential of MMP-9 modulators in psychosis.
Collapse
Affiliation(s)
- Johanna Seitz-Holland
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Yasser Alemán-Gómez
- Connectomics Lab, Department of Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Medical Image Analysis Laboratory, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Kang Ik K Cho
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ofer Pasternak
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Martine Cleusix
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Raoul Jenni
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Philipp S Baumann
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Paul Klauser
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Service of Child and Adolescent Psychiatry, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Philippe Conus
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Patric Hagmann
- Connectomics Lab, Department of Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Kim Q Do
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Marek Kubicki
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniella Dwir
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
34
|
Walker RM, Chong M, Perrot N, Pigeyre M, Gadd DA, Stolicyn A, Shi L, Campbell A, Shen X, Whalley HC, Nevado-Holgado A, McIntosh AM, Heitmeier S, Rangarajan S, O'Donnell M, Smith EE, Yusuf S, Whiteley WN, Paré G. The circulating proteome and brain health: Mendelian randomisation and cross-sectional analyses. Transl Psychiatry 2024; 14:204. [PMID: 38762535 PMCID: PMC11102511 DOI: 10.1038/s41398-024-02915-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 04/17/2024] [Accepted: 04/23/2024] [Indexed: 05/20/2024] Open
Abstract
Decline in cognitive function is the most feared aspect of ageing. Poorer midlife cognitive function is associated with increased dementia and stroke risk. The mechanisms underlying variation in cognitive function are uncertain. Here, we assessed associations between 1160 proteins' plasma levels and two measures of cognitive function, the digit symbol substitution test (DSST) and the Montreal Cognitive Assessment in 1198 PURE-MIND participants. We identified five DSST performance-associated proteins (NCAN, BCAN, CA14, MOG, CDCP1), with NCAN and CDCP1 showing replicated association in an independent cohort, GS (N = 1053). MRI-assessed structural brain phenotypes partially mediated (8-19%) associations between NCAN, BCAN, and MOG, and DSST performance. Mendelian randomisation analyses suggested higher CA14 levels might cause larger hippocampal volume and increased stroke risk, whilst higher CDCP1 levels might increase intracranial aneurysm risk. Our findings highlight candidates for further study and the potential for drug repurposing to reduce the risk of stroke and cognitive decline.
Collapse
Affiliation(s)
- Rosie M Walker
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, ON, Canada.
- School of Psychology, University of Exeter, Perry Road, Exeter, UK.
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK.
| | - Michael Chong
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, ON, Canada
- Department of Pathology and Molecular Medicine, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Nicolas Perrot
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, ON, Canada
| | - Marie Pigeyre
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, ON, Canada
- Department of Medicine, Michael G DeGroote School of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Danni A Gadd
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Aleks Stolicyn
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | - Liu Shi
- Department of Psychiatry, University of Oxford, Oxford, UK
- Nxera Pharma UK Limited, Cambridge, UK
| | - Archie Campbell
- Generation Scotland, Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Xueyi Shen
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | - Heather C Whalley
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
- Generation Scotland, Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | | | - Andrew M McIntosh
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | | | - Sumathy Rangarajan
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, ON, Canada
| | - Martin O'Donnell
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, ON, Canada
- Health Research Board Clinical Research Facility, University of Galway, Galway, Ireland
| | - Eric E Smith
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, ON, Canada
- Department of Clinical Neurosciences and Hotchkiss Brain Institute, Cumming School of Medicine, Calgary, AB, Canada
- University of Calgary, Calgary, AB, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| | - Salim Yusuf
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, ON, Canada
- Department of Medicine, Michael G DeGroote School of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - William N Whiteley
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, ON, Canada
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
- MRC Centre for Population Health, University of Oxford, Oxford, UK
| | - Guillaume Paré
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, ON, Canada.
- Department of Pathology and Molecular Medicine, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada.
- Thrombosis and Atherosclerosis Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
35
|
Paes Leme AF, Yokoo S, Normando AGC, Ormonde JVS, Domingues RR, Cruz FF, Silva PL, Souza BSF, Dos Santos CC, Castro-Faria-Neto H, Martins CM, Lopes-Pacheco M, Rocco PRM. Proteomics of serum-derived extracellular vesicles are associated with the severity and different clinical profiles of patients with COVID-19: An exploratory secondary analysis. Cytotherapy 2024; 26:444-455. [PMID: 38363248 DOI: 10.1016/j.jcyt.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 02/01/2024] [Accepted: 02/01/2024] [Indexed: 02/17/2024]
Abstract
BACKGROUND AIMS Coronavirus disease 2019 (COVID-19) is characterized by a broad spectrum of clinical manifestations with the potential to progress to multiple organ dysfunction in severe cases. Extracellular vesicles (EVs) carry a range of biological cargoes, which may be used as biomarkers of disease state. METHODS An exploratory secondary analysis of the SARITA-2 and SARITA-1 datasets (randomized clinical trials on patients with mild and moderate/severe COVID-19) was performed. Serum-derived EVs were used for proteomic analysis to identify enriched biological processes and key proteins, thus providing insights into differences in disease severity. Serum-derived EVs were separated from patients with COVID-19 by size exclusion chromatography and nanoparticle tracking analysis was used to determine particle concentration and diameter. Liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) was performed to identify and quantify protein signatures. Bioinformatics and multivariate statistical analysis were applied to distinguish candidate proteins associated with disease severity (mild versus moderate/severe COVID-19). RESULTS No differences were observed in terms of the concentration and diameter of enriched EVs between mild (n = 14) and moderate/severe (n = 30) COVID-19. A total of 414 proteins were found to be present in EVs, of which 360 were shared while 48 were uniquely present in severe/moderate compared to mild COVID-19. The main biological signatures in moderate/severe COVID-19 were associated with platelet degranulation, exocytosis, complement activation, immune effector activation, and humoral immune response. Von Willebrand factor, serum amyloid A-2 protein, histone H4 and H2A type 2-C, and fibrinogen β-chain were the most differentially expressed proteins between severity groups. CONCLUSION Exploratory proteomic analysis of serum-derived EVs from patients with COVID-19 detected key proteins related to immune response and activation of coagulation and complement pathways, which are associated with disease severity. Our data suggest that EV proteins may be relevant biomarkers of disease state and prognosis.
Collapse
Affiliation(s)
- Adriana F Paes Leme
- Laboratório Nacional de Biociências - LNBio, Centro Nacional de Pesquisa em Energia e Materiais - CNPEM, Campinas, São Paulo, Brazil
| | - Sami Yokoo
- Laboratório Nacional de Biociências - LNBio, Centro Nacional de Pesquisa em Energia e Materiais - CNPEM, Campinas, São Paulo, Brazil
| | - Ana Gabriela C Normando
- Laboratório Nacional de Biociências - LNBio, Centro Nacional de Pesquisa em Energia e Materiais - CNPEM, Campinas, São Paulo, Brazil
| | - João Vitor S Ormonde
- Laboratório Nacional de Biociências - LNBio, Centro Nacional de Pesquisa em Energia e Materiais - CNPEM, Campinas, São Paulo, Brazil
| | - Romenia Ramos Domingues
- Laboratório Nacional de Biociências - LNBio, Centro Nacional de Pesquisa em Energia e Materiais - CNPEM, Campinas, São Paulo, Brazil
| | - Fernanda F Cruz
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSaúde, Research Support Foundation of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro L Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSaúde, Research Support Foundation of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bruno S F Souza
- Goncalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Bahia, Brazil; D'Or Institute for Research and Education (IDOR), Salvador, Bahia, Brazil; Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Bahia, Brazil
| | - Claudia C Dos Santos
- The Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada; Institute of Medical Sciences and Interdepartmental Division of Critical Care, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | | | - Miquéias Lopes-Pacheco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSaúde, Research Support Foundation of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
36
|
Jiang Y, Uhm H, Ip FC, Ouyang L, Lo RMN, Cheng EYL, Cao X, Tan CMC, Law BCH, Ortiz‐Romero P, Puig‐Pijoan A, Fernández‐Lebrero A, Contador J, Mok KY, Hardy J, Kwok TCY, Mok VCT, Suárez‐Calvet M, Zetterberg H, Fu AKY, Ip NY. A blood-based multi-pathway biomarker assay for early detection and staging of Alzheimer's disease across ethnic groups. Alzheimers Dement 2024; 20:2000-2015. [PMID: 38183344 PMCID: PMC10984431 DOI: 10.1002/alz.13676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 01/08/2024]
Abstract
INTRODUCTION Existing blood-based biomarkers for Alzheimer's disease (AD) mainly focus on its pathological features. However, studies on blood-based biomarkers associated with other biological processes for a comprehensive evaluation of AD status are limited. METHODS We developed a blood-based, multiplex biomarker assay for AD that measures the levels of 21 proteins involved in multiple biological pathways. We evaluated the assay's performance for classifying AD and indicating AD-related endophenotypes in three independent cohorts from Chinese or European-descent populations. RESULTS The 21-protein assay accurately classified AD (area under the receiver operating characteristic curve [AUC] = 0.9407 to 0.9867) and mild cognitive impairment (MCI; AUC = 0.8434 to 0.8945) while also indicating brain amyloid pathology. Moreover, the assay simultaneously evaluated the changes of five biological processes in individuals and revealed the ethnic-specific dysregulations of biological processes upon AD progression. DISCUSSION This study demonstrated the utility of a blood-based, multi-pathway biomarker assay for early screening and staging of AD, providing insights for patient stratification and precision medicine. HIGHLIGHTS The authors developed a blood-based biomarker assay for Alzheimer's disease. The 21-protein assay classifies AD/MCI and indicates brain amyloid pathology. The 21-protein assay can simultaneously assess activities of five biological processes. Ethnic-specific dysregulations of biological processes in AD were revealed.
Collapse
Affiliation(s)
- Yuanbing Jiang
- Division of Life ScienceState Key Laboratory of Molecular Neuroscience, Molecular Neuroscience CenterThe Hong Kong University of Science and Technology, Clear Water Bay, KowloonHKSARChina
| | - Hyebin Uhm
- Division of Life ScienceState Key Laboratory of Molecular Neuroscience, Molecular Neuroscience CenterThe Hong Kong University of Science and Technology, Clear Water Bay, KowloonHKSARChina
| | - Fanny C. Ip
- Hong Kong Center for Neurodegenerative Diseases, InnoHKHKSARChina
- Guangdong Provincial Key Laboratory of Brain ScienceDisease and Drug DevelopmentHKUST Shenzhen Research InstituteShenzhenGuangdongChina
| | - Li Ouyang
- Hong Kong Center for Neurodegenerative Diseases, InnoHKHKSARChina
| | - Ronnie M. N. Lo
- Division of Life ScienceState Key Laboratory of Molecular Neuroscience, Molecular Neuroscience CenterThe Hong Kong University of Science and Technology, Clear Water Bay, KowloonHKSARChina
- Hong Kong Center for Neurodegenerative Diseases, InnoHKHKSARChina
| | - Elaine Y. L. Cheng
- Division of Life ScienceState Key Laboratory of Molecular Neuroscience, Molecular Neuroscience CenterThe Hong Kong University of Science and Technology, Clear Water Bay, KowloonHKSARChina
- Hong Kong Center for Neurodegenerative Diseases, InnoHKHKSARChina
| | - Xiaoyun Cao
- Hong Kong Center for Neurodegenerative Diseases, InnoHKHKSARChina
| | - Clara M. C. Tan
- Hong Kong Center for Neurodegenerative Diseases, InnoHKHKSARChina
| | - Brian C. H. Law
- Hong Kong Center for Neurodegenerative Diseases, InnoHKHKSARChina
| | - Paula Ortiz‐Romero
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Research InstituteBarcelonaSpain
| | - Albert Puig‐Pijoan
- Hospital del Mar Research InstituteBarcelonaSpain
- Cognitive Decline Unit, Department of NeurologyHospital Del MarBarcelonaSpain
- Medicine DepartmentUniversitat Autònoma de BarcelonaBarcelonaSpain
- ERA‐Net on Cardiovascular Diseases (ERA‐CVD) ConsortiumBarcelonaSpain
| | - Aida Fernández‐Lebrero
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Research InstituteBarcelonaSpain
- Cognitive Decline Unit, Department of NeurologyHospital Del MarBarcelonaSpain
- ERA‐Net on Cardiovascular Diseases (ERA‐CVD) ConsortiumBarcelonaSpain
- Department of Medicine and Life SciencesUniversitat Pompeu FabraBarcelonaSpain
| | - José Contador
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Research InstituteBarcelonaSpain
- Cognitive Decline Unit, Department of NeurologyHospital Del MarBarcelonaSpain
| | - Kin Y. Mok
- Division of Life ScienceState Key Laboratory of Molecular Neuroscience, Molecular Neuroscience CenterThe Hong Kong University of Science and Technology, Clear Water Bay, KowloonHKSARChina
- Hong Kong Center for Neurodegenerative Diseases, InnoHKHKSARChina
- Department of Neurodegenerative DiseaseQueen Square Institute of NeurologyUniversity College LondonLondonUK
| | - John Hardy
- Hong Kong Center for Neurodegenerative Diseases, InnoHKHKSARChina
- Department of Neurodegenerative DiseaseQueen Square Institute of NeurologyUniversity College LondonLondonUK
- UK Dementia Research InstituteUniversity College LondonLondonUK
| | - Timothy C. Y. Kwok
- Therese Pei Fong Chow Research Centre for Prevention of DementiaDivision of GeriatricsDepartment of Medicine and TherapeuticsThe Chinese University of Hong Kong, ShatinHKSARChina
| | - Vincent C. T. Mok
- Lau Tat‐chuen Research Centre of Brain Degenerative Diseases in ChineseGerald Choa Neuroscience InstituteLui Che Woo Institute of Innovative MedicineLi Ka Shing Institute of Health SciencesDivision of NeurologyDepartment of Medicine and TherapeuticsThe Chinese University of Hong Kong, ShatinHKSARChina
| | - Marc Suárez‐Calvet
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Research InstituteBarcelonaSpain
- Cognitive Decline Unit, Department of NeurologyHospital Del MarBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
| | - Henrik Zetterberg
- Hong Kong Center for Neurodegenerative Diseases, InnoHKHKSARChina
- Department of Neurodegenerative DiseaseQueen Square Institute of NeurologyUniversity College LondonLondonUK
- UK Dementia Research InstituteUniversity College LondonLondonUK
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Amy K. Y. Fu
- Division of Life ScienceState Key Laboratory of Molecular Neuroscience, Molecular Neuroscience CenterThe Hong Kong University of Science and Technology, Clear Water Bay, KowloonHKSARChina
- Hong Kong Center for Neurodegenerative Diseases, InnoHKHKSARChina
- Guangdong Provincial Key Laboratory of Brain ScienceDisease and Drug DevelopmentHKUST Shenzhen Research InstituteShenzhenGuangdongChina
| | - Nancy Y. Ip
- Division of Life ScienceState Key Laboratory of Molecular Neuroscience, Molecular Neuroscience CenterThe Hong Kong University of Science and Technology, Clear Water Bay, KowloonHKSARChina
- Hong Kong Center for Neurodegenerative Diseases, InnoHKHKSARChina
- Guangdong Provincial Key Laboratory of Brain ScienceDisease and Drug DevelopmentHKUST Shenzhen Research InstituteShenzhenGuangdongChina
| |
Collapse
|
37
|
Abiose O, Rutledge J, Moran‐Losada P, Belloy ME, Wilson EN, He Z, Trelle AN, Channappa D, Romero A, Park J, Yutsis MV, Sha SJ, Andreasson KI, Poston KL, Henderson VW, Wagner AD, Wyss‐Coray T, Mormino EC. Post-translational modifications linked to preclinical Alzheimer's disease-related pathological and cognitive changes. Alzheimers Dement 2024; 20:1851-1867. [PMID: 38146099 PMCID: PMC10984434 DOI: 10.1002/alz.13576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 11/08/2023] [Accepted: 11/13/2023] [Indexed: 12/27/2023]
Abstract
INTRODUCTION In this study, we leverage proteomic techniques to identify communities of proteins underlying Alzheimer's disease (AD) risk among clinically unimpaired (CU) older adults. METHODS We constructed a protein co-expression network using 3869 cerebrospinal fluid (CSF) proteins quantified by SomaLogic, Inc., in a cohort of participants along the AD clinical spectrum. We then replicated this network in an independent cohort of CU older adults and related these modules to clinically-relevant outcomes. RESULTS We discovered modules enriched for phosphorylation and ubiquitination that were associated with abnormal amyloid status, as well as p-tau181 (M4: β = 2.44, p < 0.001, M7: β = 2.57, p < 0.001) and executive function performance (M4: β = -2.00, p = 0.005, M7: β = -2.39, p < 0.001). DISCUSSION In leveraging CSF proteomic data from individuals spanning the clinical spectrum of AD, we highlight the importance of post-translational modifications for early cognitive and pathological changes.
Collapse
Affiliation(s)
- Olamide Abiose
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Wu Tsai Neurosciences InstituteStanford University School of MedicineStanfordCaliforniaUSA
| | - Jarod Rutledge
- The Phil and Penny Knight Initiative for Brain ResilienceStanford UniversityStanfordCaliforniaUSA
- Department of GeneticsStanford UniversityStanfordCaliforniaUSA
| | - Patricia Moran‐Losada
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Wu Tsai Neurosciences InstituteStanford University School of MedicineStanfordCaliforniaUSA
- The Phil and Penny Knight Initiative for Brain ResilienceStanford UniversityStanfordCaliforniaUSA
| | - Michael E. Belloy
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Edward N. Wilson
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Wu Tsai Neurosciences InstituteStanford University School of MedicineStanfordCaliforniaUSA
| | - Zihuai He
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Center for Biomedical Informatics ResearchStanford University School of MedicineStanfordCaliforniaUSA
| | - Alexandra N. Trelle
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Divya Channappa
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - America Romero
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Jennifer Park
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Maya V. Yutsis
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Sharon J. Sha
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Katrin I. Andreasson
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Wu Tsai Neurosciences InstituteStanford University School of MedicineStanfordCaliforniaUSA
- Chan Zuckerberg BiohubSan FranciscoCaliforniaUSA
| | - Kathleen L. Poston
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Wu Tsai Neurosciences InstituteStanford University School of MedicineStanfordCaliforniaUSA
- The Phil and Penny Knight Initiative for Brain ResilienceStanford UniversityStanfordCaliforniaUSA
| | - Victor W. Henderson
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Department of Epidemiology & Population HealthStanford University School of MedicineStanfordCaliforniaUSA
| | - Anthony D. Wagner
- Wu Tsai Neurosciences InstituteStanford University School of MedicineStanfordCaliforniaUSA
- Department of PsychologyStanford UniversityStanfordCaliforniaUSA
| | - Tony Wyss‐Coray
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Wu Tsai Neurosciences InstituteStanford University School of MedicineStanfordCaliforniaUSA
- The Phil and Penny Knight Initiative for Brain ResilienceStanford UniversityStanfordCaliforniaUSA
| | - Elizabeth C. Mormino
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Wu Tsai Neurosciences InstituteStanford University School of MedicineStanfordCaliforniaUSA
| |
Collapse
|
38
|
Wang YL, Zhu MY, Yuan ZF, Ren XY, Guo XT, Hua Y, Xu L, Zhao CY, Jiang LH, Zhang X, Sheng GX, Jiang PF, Zhao ZY, Gao F. Proteomic profiling of cerebrospinal fluid in pediatric myelin oligodendrocyte glycoprotein antibody-associated disease. World J Pediatr 2024; 20:259-271. [PMID: 36507981 PMCID: PMC10957615 DOI: 10.1007/s12519-022-00661-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 11/15/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Myelin oligodendrocyte glycoprotein (MOG) antibody-associated disease (MOGAD) is an autoimmune demyelinating disorder of the central nervous system. METHODS Extracted proteins from 34 cerebrospinal fluid (CSF) samples [patients with MOGAD (MOG group, n = 12); healthy controls (HC group, n = 12); patients with MOG seronegative and metagenomics next-generation sequencing-negative inflammatory neurological diseases (IND group, n = 10)] were processed and subjected to label-free quantitative proteomics. Supervised partial least squares-discriminant analysis (PLS-DA) and orthogonal PLS-DA (O-PLS-DA) models were also performed based on proteomics data. Functional analysis of differentially expressed proteins (DEPs) was performed using Gene Ontology, InterPro, and Kyoto Encyclopedia Genes and Genomes. An enzyme-linked immunosorbent assay was used to determine the complement levels in serum from patients with MOGAD. RESULTS Four hundred and twenty-nine DEPs (149 upregulated and 280 downregulated proteins) were identified in the MOG group compared to the HC group according to the P value and fold change (FC). Using the O-PLS-DA model, 872 differentially abundant proteins were identified with variable importance projection (VIP) scores > 1. Five proteins (gamma-glutamyl hydrolase, cathepsin F, interalpha-trypsin inhibitor heavy chain 5, latent transforming growth factor beta-binding protein 4 and leukocyte-associated immunoglobulin-like receptor 1) overlapping between the top 30 DEPs with top-ranked P value and FC and top 30 proteins in PLS-DA VIP lists were acquired. Functional analysis revealed that the dysregulated proteins in the MOG group were primarily involved in complement and coagulation cascades, cell adhesion, axon guidance, and glycosphingolipid biosynthesis compared to the HC group. CONCLUSION The proteomic alterations in CSF samples from children with MOGAD identified in the current study might provide opportunities for developing novel biomarker candidates.
Collapse
Affiliation(s)
- Yi-Long Wang
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
- Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou 310052, China
| | - Meng-Ying Zhu
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
- Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou 310052, China
| | - Zhe-Feng Yuan
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
- Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou 310052, China
| | - Xiao-Yan Ren
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
| | - Xiao-Tong Guo
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
- Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou 310052, China
| | - Yi Hua
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
- Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou 310052, China
| | - Lu Xu
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
- Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou 310052, China
| | - Cong-Ying Zhao
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
- Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou 310052, China
| | - Li-Hua Jiang
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
- Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou 310052, China
| | - Xin Zhang
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
- Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou 310052, China
| | - Guo-Xia Sheng
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
- Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou 310052, China
| | - Pei-Fang Jiang
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
- Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou 310052, China
| | - Zheng-Yan Zhao
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China.
- Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou 310052, China.
| | - Feng Gao
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China.
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China.
- Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou 310052, China.
| |
Collapse
|
39
|
Guo Q, Ping L, Dammer EB, Yin L, Xu K, Shantaraman A, Fox EJ, Golde TE, Johnson ECB, Roberts BR, Lah JJ, Levey AI, Seyfried NT. Heparin-enriched plasma proteome is significantly altered in Alzheimer's Disease. RESEARCH SQUARE 2024:rs.3.rs-3933136. [PMID: 38464223 PMCID: PMC10925398 DOI: 10.21203/rs.3.rs-3933136/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Introduction Heparin binding proteins (HBPs) with roles in extracellular matrix assembly are strongly correlated to β-amyloid (Aβ) and tau pathology in Alzheimer's disease (AD) brain and cerebrospinal fluid (CSF). However, it remains challenging to detect these proteins in plasma using standard mass spectrometry-based proteomic approaches. Methods We employed heparin affinity chromatography, followed by off-line fractionation and tandem mass tag mass spectrometry (TMT-MS), to capture and enrich HBPs in plasma obtained from AD (n=62) and control (n=47) samples. These profiles were then correlated to a consensus AD brain proteome, as well as with Aβ, tau and phosphorylated tau (pTau) CSF biomarkers from the same individuals. We then leveraged published human postmortem brain proteome datasets to assess the overlap with the heparin-enriched plasma proteome. Results Heparin-enrichment from plasma was highly reproducible, enriched well-known HBPs like APOE and thrombin, and depleted high-abundance proteins such as albumin. A total of 2865 proteins, spanning 10 orders of magnitude were detectable. Utilizing a consensus AD brain protein co-expression network, we observed that specific plasma HBPs exhibited consistent direction of change in both brain and plasma, whereas others displayed divergent changes highlighting the complex interplay between the two compartments. Elevated HBPs in AD plasma, when compared to controls, included members of the matrisome module in brain that accumulate within Aβ deposits, such as SMOC1, SMOC2, SPON1, MDK, OLFML3, FRZB, GPNMB, and APOE. Additionally, heparin enriched plasma proteins demonstrated significant correlations with conventional AD CSF biomarkers, including Aβ, total tau, pTau, and plasma pTau from the same individuals. Conclusion These findings support the utility of a heparin-affinity approach for enriching amyloid-associated proteins, as well as a wide spectrum of plasma biomarkers that reflect pathological changes in the AD brain.
Collapse
Affiliation(s)
- Qi Guo
- Emory University School of Medicine
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Hu Q, Shi M, Li Y, Zhao X. Elevated plasma neurofilament light was associated with multi-modal neuroimaging features in Alzheimer's Disease signature regions and predicted future tau deposition. RESEARCH SQUARE 2024:rs.3.rs-3946421. [PMID: 38464117 PMCID: PMC10925409 DOI: 10.21203/rs.3.rs-3946421/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Background Neurofilament Light (NfL) is a biomarker for early neurodegeneration in Alzheimer's disease (AD). This study aims to examine the association between plasma NfL and multi-modal neuroimaging features across the AD spectrum and whether NfL predicts future tau deposition. Methods The present study recruited 517 participants comprising Aβ negative cognitively normal (CN-) participants (n = 135), CN + participants (n = 64), individuals with mild cognitive impairment (MCI) (n = 212), and those diagnosed with AD dementia (n = 106). All the participants underwent multi-modal neuroimaging examinations. Cross-sectional and longitudinal associations between plasma NfL and multi-modal neuro-imaging features were evaluated using partial correlation analysis and linear mixed effects models. We also used linear regression analysis to investigate the association of baseline plasma NfL with future PET tau load. Mediation analysis was used to explore whether the effect of NfL on cognition was mediated by these MRI markers. Results The results showed that baseline NfL levels and the rate of change were associated with Aβ deposition, brain atrophy, brain connectome, glucose metabolism, and brain perfusion in AD signature regions. In both Aβ positive CN and MCI participants, baseline NfL showed a significant predictive value of elevating tau burden in the left medial orbitofrontal cortex and para-hippocampus. Lastly, the multi-modal neuroimaging features mediated the association between plasma NfL and cognitive performance. Conclusions The study supports the association between plasma NfL and multi-modal neuroimaging features in AD-vulnerable regions and its predictive value for future tau deposition.
Collapse
Affiliation(s)
- Qili Hu
- The Fifth People's Hospital of Shanghai, Fudan University
| | - Mengqiu Shi
- The Fifth People's Hospital of Shanghai, Fudan University
| | - Yunfei Li
- The Fifth People's Hospital of Shanghai, Fudan University
| | - Xiaohu Zhao
- The Fifth People's Hospital of Shanghai, Fudan University
| |
Collapse
|
41
|
Li R, Wang J, Xiong W, Luo Y, Feng H, Zhou H, Peng Y, He Y, Ye Q. The oral-brain axis: can periodontal pathogens trigger the onset and progression of Alzheimer's disease? Front Microbiol 2024; 15:1358179. [PMID: 38362505 PMCID: PMC10868393 DOI: 10.3389/fmicb.2024.1358179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 01/18/2024] [Indexed: 02/17/2024] Open
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia, characterized by a progressive cognitive decline. Sporadic AD, accounting for more than 95% of cases, may arise due to the influence of environmental factors. It was reported that periodontitis, a common oral ailment, shares several risk factors with AD, including advanced age, smoking, diabetes, and hypertension, among others. Periodontitis is an inflammatory disease triggered by dysbiosis of oral microorganisms, whereas Alzheimer's disease is characterized by neuroinflammation. Many studies have indicated that chronic inflammation can instigate brain AD-related pathologies, including amyloid-β plaques, Tau protein hyperphosphorylation, neuroinflammation, and neurodegeneration. The potential involvement of periodontal pathogens and/or their virulence factors in the onset and progression of AD by the oral-brain axis has garnered significant attention among researchers with ongoing investigations. This review has updated the periodontal pathogens potentially associated with AD, elucidating their impact on the central nervous system, immune response, and related pathological processes in the brain to provide valuable insights for future research on the oral-brain axis.
Collapse
Affiliation(s)
- Ruohan Li
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Junnan Wang
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Wei Xiong
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yu Luo
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
- Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Huixian Feng
- Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Heng Zhou
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Youjian Peng
- Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yan He
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Qingsong Ye
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
- Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| |
Collapse
|
42
|
Guo Y, You J, Zhang Y, Liu WS, Huang YY, Zhang YR, Zhang W, Dong Q, Feng JF, Cheng W, Yu JT. Plasma proteomic profiles predict future dementia in healthy adults. NATURE AGING 2024; 4:247-260. [PMID: 38347190 DOI: 10.1038/s43587-023-00565-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 12/22/2023] [Indexed: 02/22/2024]
Abstract
The advent of proteomics offers an unprecedented opportunity to predict dementia onset. We examined this in data from 52,645 adults without dementia in the UK Biobank, with 1,417 incident cases and a follow-up time of 14.1 years. Of 1,463 plasma proteins, GFAP, NEFL, GDF15 and LTBP2 consistently associated most with incident all-cause dementia (ACD), Alzheimer's disease (AD) and vascular dementia (VaD), and ranked high in protein importance ordering. Combining GFAP (or GDF15) with demographics produced desirable predictions for ACD (area under the curve (AUC) = 0.891) and AD (AUC = 0.872) (or VaD (AUC = 0.912)). This was also true when predicting over 10-year ACD, AD and VaD. Individuals with higher GFAP levels were 2.32 times more likely to develop dementia. Notably, GFAP and LTBP2 were highly specific for dementia prediction. GFAP and NEFL began to change at least 10 years before dementia diagnosis. Our findings strongly highlight GFAP as an optimal biomarker for dementia prediction, even more than 10 years before the diagnosis, with implications for screening people at high risk for dementia and for early intervention.
Collapse
Affiliation(s)
- Yu Guo
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jia You
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
| | - Yi Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei-Shi Liu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu-Yuan Huang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ya-Ru Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Zhang
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China.
| | - Wei Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China.
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
43
|
Han Y, Zeng X, Hua L, Quan X, Chen Y, Zhou M, Chuang Y, Li Y, Wang S, Shen X, Wei L, Yuan Z, Zhao Y. The fusion of multi-omics profile and multimodal EEG data contributes to the personalized diagnostic strategy for neurocognitive disorders. MICROBIOME 2024; 12:12. [PMID: 38243335 PMCID: PMC10797890 DOI: 10.1186/s40168-023-01717-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 11/07/2023] [Indexed: 01/21/2024]
Abstract
BACKGROUND The increasing prevalence of neurocognitive disorders (NCDs) in the aging population worldwide has become a significant concern due to subjectivity of evaluations and the lack of precise diagnostic methods and specific indicators. Developing personalized diagnostic strategies for NCDs has therefore become a priority. RESULTS Multimodal electroencephalography (EEG) data of a matched cohort of normal aging (NA) and NCDs seniors were recorded, and their faecal samples and urine exosomes were collected to identify multi-omics signatures and metabolic pathways in NCDs by integrating metagenomics, proteomics, and metabolomics analysis. Additionally, experimental verification of multi-omics signatures was carried out in aged mice using faecal microbiota transplantation (FMT). We found that NCDs seniors had low EEG power spectral density and identified specific microbiota, including Ruminococcus gnavus, Enterocloster bolteae, Lachnoclostridium sp. YL 32, and metabolites, including L-tryptophan, L-glutamic acid, gamma-aminobutyric acid (GABA), and fatty acid esters of hydroxy fatty acids (FAHFAs), as well as disturbed biosynthesis of aromatic amino acids and TCA cycle dysfunction, validated in aged mice. Finally, we employed a support vector machine (SVM) algorithm to construct a machine learning model to classify NA and NCDs groups based on the fusion of EEG data and multi-omics profiles and the model demonstrated 92.69% accuracy in classifying NA and NCDs groups. CONCLUSIONS Our study highlights the potential of multi-omics profiling and EEG data fusion in personalized diagnosis of NCDs, with the potential to improve diagnostic precision and provide insights into the underlying mechanisms of NCDs. Video Abstract.
Collapse
Affiliation(s)
- Yan Han
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, 999078, Macau SAR, China
| | - Xinglin Zeng
- Centre for Cognitive and Brain Sciences, University of Macau, Avenida da Universidade, Taipa, 999078, Macau SAR, China
| | - Lin Hua
- Centre for Cognitive and Brain Sciences, University of Macau, Avenida da Universidade, Taipa, 999078, Macau SAR, China
| | - Xingping Quan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, 999078, Macau SAR, China
| | - Ying Chen
- School of Health Economics and Management, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Manfei Zhou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, 999078, Macau SAR, China
| | | | - Yang Li
- Department of Gastrointestinal Surgery, Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, 518020, China
| | - Shengpeng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, 999078, Macau SAR, China
| | - Xu Shen
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lai Wei
- School of Pharmaceutical Science, Southern Medical University, Guangzhou, 510515, China
| | - Zhen Yuan
- Centre for Cognitive and Brain Sciences, University of Macau, Avenida da Universidade, Taipa, 999078, Macau SAR, China.
| | - Yonghua Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, 999078, Macau SAR, China.
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR 999078, China.
| |
Collapse
|
44
|
Bhalala OG, Watson R, Yassi N. Multi-Omic Blood Biomarkers as Dynamic Risk Predictors in Late-Onset Alzheimer's Disease. Int J Mol Sci 2024; 25:1231. [PMID: 38279230 PMCID: PMC10816901 DOI: 10.3390/ijms25021231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024] Open
Abstract
Late-onset Alzheimer's disease is the leading cause of dementia worldwide, accounting for a growing burden of morbidity and mortality. Diagnosing Alzheimer's disease before symptoms are established is clinically challenging, but would provide therapeutic windows for disease-modifying interventions. Blood biomarkers, including genetics, proteins and metabolites, are emerging as powerful predictors of Alzheimer's disease at various timepoints within the disease course, including at the preclinical stage. In this review, we discuss recent advances in such blood biomarkers for determining disease risk. We highlight how leveraging polygenic risk scores, based on genome-wide association studies, can help stratify individuals along their risk profile. We summarize studies analyzing protein biomarkers, as well as report on recent proteomic- and metabolomic-based prediction models. Finally, we discuss how a combination of multi-omic blood biomarkers can potentially be used in memory clinics for diagnosis and to assess the dynamic risk an individual has for developing Alzheimer's disease dementia.
Collapse
Affiliation(s)
- Oneil G. Bhalala
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; (R.W.); (N.Y.)
- Department of Neurology, Melbourne Brain Centre at The Royal Melbourne Hospital, University of Melbourne, Parkville 3050, Australia
| | - Rosie Watson
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; (R.W.); (N.Y.)
- Department of Medicine, The Royal Melbourne Hospital, University of Melbourne, Parkville 3050, Australia
| | - Nawaf Yassi
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; (R.W.); (N.Y.)
- Department of Neurology, Melbourne Brain Centre at The Royal Melbourne Hospital, University of Melbourne, Parkville 3050, Australia
- Department of Medicine, The Royal Melbourne Hospital, University of Melbourne, Parkville 3050, Australia
| |
Collapse
|
45
|
Tao QQ, Cai X, Xue YY, Ge W, Yue L, Li XY, Lin RR, Peng GP, Jiang W, Li S, Zheng KM, Jiang B, Jia JP, Guo T, Wu ZY. Alzheimer's disease early diagnostic and staging biomarkers revealed by large-scale cerebrospinal fluid and serum proteomic profiling. Innovation (N Y) 2024; 5:100544. [PMID: 38235188 PMCID: PMC10794110 DOI: 10.1016/j.xinn.2023.100544] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 11/19/2023] [Indexed: 01/19/2024] Open
Abstract
Amyloid-β, tau pathology, and biomarkers of neurodegeneration make up the core diagnostic biomarkers of Alzheimer disease (AD). However, these proteins represent only a fraction of the complex biological processes underlying AD, and individuals with other brain diseases in which AD pathology is a comorbidity also test positive for these diagnostic biomarkers. More AD-specific early diagnostic and disease staging biomarkers are needed. In this study, we performed tandem mass tag proteomic analysis of paired cerebrospinal fluid (CSF) and serum samples in a discovery cohort comprising 98 participants. Candidate biomarkers were validated by parallel reaction monitoring-based targeted proteomic assays in an independent multicenter cohort comprising 288 participants. We quantified 3,238 CSF and 1,702 serum proteins in the discovery cohort, identifying 171 and 860 CSF proteins and 37 and 323 serum proteins as potential early diagnostic and staging biomarkers, respectively. In the validation cohort, 58 and 21 CSF proteins, as well as 12 and 18 serum proteins, were verified as early diagnostic and staging biomarkers, respectively. Separate 19-protein CSF and an 8-protein serum biomarker panels were built by machine learning to accurately classify mild cognitive impairment (MCI) due to AD from normal cognition with areas under the curve of 0.984 and 0.881, respectively. The 19-protein CSF biomarker panel also effectively discriminated patients with MCI due to AD from patients with other neurodegenerative diseases. Moreover, we identified 21 CSF and 18 serum stage-associated proteins reflecting AD stages. Our findings provide a foundation for developing blood-based tests for AD screening and staging in clinical practice.
Collapse
Affiliation(s)
- Qing-Qing Tao
- Department of Neurology and Research Center of Neurology in the Second Affiliated Hospital and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310009, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311100, China
| | - Xue Cai
- Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzho 310024, China
| | - Yan-Yan Xue
- Department of Neurology and Research Center of Neurology in the Second Affiliated Hospital and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Weigang Ge
- Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzho 310024, China
| | - Liang Yue
- Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzho 310024, China
| | - Xiao-Yan Li
- Department of Neurology and Research Center of Neurology in the Second Affiliated Hospital and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Rong-Rong Lin
- Department of Neurology and Research Center of Neurology in the Second Affiliated Hospital and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Guo-Ping Peng
- Department of Neurology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Wenhao Jiang
- Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzho 310024, China
| | - Sainan Li
- Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzho 310024, China
| | - Kun-Mu Zheng
- Department of Neurology, First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen 361009, China
| | - Bin Jiang
- Department of Neurology, First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen 361009, China
| | - Jian-Ping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, China
| | - Tiannan Guo
- Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzho 310024, China
| | - Zhi-Ying Wu
- Department of Neurology and Research Center of Neurology in the Second Affiliated Hospital and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310009, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311100, China
- MOE Frontier Science Center for Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai 200031, China
| |
Collapse
|
46
|
Kojima R, Paslawski W, Lyu G, Arenas E, Zhang X, Svenningsson P. Secretome Analyses Identify FKBP4 as a GBA1-Associated Protein in CSF and iPS Cells from Parkinson's Disease Patients with GBA1 Mutations. Int J Mol Sci 2024; 25:683. [PMID: 38203854 PMCID: PMC10779269 DOI: 10.3390/ijms25010683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 12/31/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024] Open
Abstract
Mutations in the GBA1 gene increase the risk of developing Parkinson's disease (PD). However, most carriers of GBA1 mutations do not develop PD throughout their lives. The mechanisms of how GBA1 mutations contribute to PD pathogenesis remain unclear. Cerebrospinal fluid (CSF) is used for detecting pathological conditions of diseases, providing insights into the molecular mechanisms underlying neurodegenerative disorders. In this study, we utilized the proximity extension assay to examine the levels of metabolism-linked protein in the CSF from 17 PD patients carrying GBA1 mutations (GBA1-PD) and 17 idiopathic PD (iPD). The analysis of CSF secretome in GBA1-PD identified 11 significantly altered proteins, namely FKBP4, THOP1, GLRX, TXNDC5, GAL, SEMA3F, CRKL, APLP1, LRP11, CD164, and NPTXR. To investigate GBA1-associated CSF changes attributed to specific neuronal subtypes responsible for PD, we analyzed the cell culture supernatant from GBA1-PD-induced pluripotent stem cell (iPSC)-derived midbrain dopaminergic (mDA) neurons. The secretome analysis of GBA1-PD iPSC-derived mDA neurons revealed that five differently regulated proteins overlapped with those identified in the CSF analysis: FKBP4, THOP1, GLRX, GAL, and CRKL. Reduced intracellular level of the top hit, FKPB4, was confirmed via Western Blot. In conclusion, our findings identify significantly altered CSF GBA1-PD-associated proteins with FKPB4 being firmly attributed to mDA neurons.
Collapse
Affiliation(s)
- Rika Kojima
- Department of Clinical Neuroscience, Karolinska Institutet, 171 76 Stockholm, Sweden; (R.K.)
| | - Wojciech Paslawski
- Department of Clinical Neuroscience, Karolinska Institutet, 171 76 Stockholm, Sweden; (R.K.)
| | - Guochang Lyu
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Ernest Arenas
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Xiaoqun Zhang
- Department of Clinical Neuroscience, Karolinska Institutet, 171 76 Stockholm, Sweden; (R.K.)
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institutet, 171 76 Stockholm, Sweden; (R.K.)
| |
Collapse
|
47
|
van Eijck CWF, Strijk G, Vietsch EE, van der Sijde F, Verheij M, Mustafa DAM, Vink M, Aerts JGJV, van Eijck CHJ, Willemsen M. FOLFIRINOX chemotherapy modulates the peripheral immune landscape in pancreatic cancer: Implications for combination therapies and early response prediction. Eur J Cancer 2024; 196:113440. [PMID: 37988843 DOI: 10.1016/j.ejca.2023.113440] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 11/01/2023] [Indexed: 11/23/2023]
Abstract
BACKGROUND FOLFIRINOX chemotherapy has improved outcomes for pancreatic cancer patients, but poor long-term survival outcomes and high toxicity remain challenges. This study investigates the impact of FOLFIRINOX on plasma proteins and peripheral immune cells to guide immune-based combination therapies and, ideally, to identify a potential biomarker to predict early disease progression during FOLFIRINOX. METHODS Blood samples were collected from 86 pancreatic cancer patients before and two weeks after the first FOLFIRINOX cycle and subjected to comprehensive immune cell and proteome profiling. Principal Component Analysis and Linear Mixed Effect Regression models were used for data analysis. FOLFIRINOX efficacy was radiologically evaluated after the fourth cycle. RESULTS One cycle of FOLFIRINOX diminished tumour-cell-related pathways and enhanced pathways related to immune activation, illustrated by an increase in pro-inflammatory IL-18, IL-15, and TNFRSF4. Similarly, FOLFIRINOX promoted the activation of CD4 + and CD8 + T cells, the proliferation of NK(T), and the activation of antigen-presenting cells. Furthermore, high pre-treatment levels of VEGFA and PRDX3 and an elevation in FCRL3 levels after one cycle predicted early progression under FOLFIRINOX. Finally, patients with progressive disease exhibited high levels of inhibitory markers on B cells and CD8 + T cells, while responding patients exhibited high levels of activation markers on CD4 + and CD8 + T cell subsets. CONCLUSION FOLFIRINOX has immunomodulatory effects, providing a foundation for clinical trials exploring immune-based combination therapies that harness the immune system to treat pancreatic cancer. In addition, several plasma proteins hold potential as circulating predictive biomarkers for early prediction of FOLFIRINOX response in patients with pancreatic cancer.
Collapse
Affiliation(s)
- Casper W F van Eijck
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, the Netherlands; Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre, Madrid, Spain
| | - Gaby Strijk
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Eveline E Vietsch
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, the Netherlands; Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Fleur van der Sijde
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Maaike Verheij
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Dana A M Mustafa
- Department of Pathology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Madelief Vink
- Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, the Netherlands; Department of Pulmonary Medicine, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Joachim G J V Aerts
- Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, the Netherlands; Department of Pulmonary Medicine, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Casper H J van Eijck
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, the Netherlands; Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre, Madrid, Spain
| | - Marcella Willemsen
- Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, the Netherlands; Department of Pulmonary Medicine, Erasmus University Medical Centre, Rotterdam, the Netherlands.
| |
Collapse
|
48
|
Beydoun MA, Beydoun HA, Noren Hooten N, Meirelles O, Li Z, El-Hajj ZW, Weiss J, Maino Vieytes CA, Launer LJ, Evans MK, Zonderman AB. Hospital-treated prevalent infections, the plasma proteome and incident dementia among UK older adults. iScience 2023; 26:108526. [PMID: 38162022 PMCID: PMC10755048 DOI: 10.1016/j.isci.2023.108526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/18/2023] [Accepted: 11/20/2023] [Indexed: 01/03/2024] Open
Abstract
The plasma proteome can mediate the association of hospital-treated infections with dementia incidence. We screened up to 37,269 UK Biobank participants aged 50-74 years for the presence of a prevalent hospital-treated infection, subsequently tested as a predictor for ≤1,463 plasma proteins and dementia incidence. Four-way decomposition models decomposed infection-dementia total effect into pure mediation, pure interaction, neither or both through the plasma proteome. Hospital-treated infections increased dementia two-fold. The strongest mediation effect was through the growth differentiation factor 15 (GDF15) protein. Top 17 proteomic mediators explained collectively 5% of the total effect, while pathway analysis of all mediators (k = 221 plasma proteins) revealed top pathways including the immune system, signal transduction, metabolism, disease and metabolism of proteins, with the GDF15 cluster reflecting most strongly the "transmembrane receptor protein tyrosine kinase signaling pathway". The association of hospital-treated infections with dementia was partially mediated through GDF15 and other plasma proteomic markers.
Collapse
Affiliation(s)
- May A. Beydoun
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Intramural Research Program, NIA/NIH/IRP, Baltimore, MD 21224, USA
| | - Hind A. Beydoun
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Intramural Research Program, NIA/NIH/IRP, Baltimore, MD 21224, USA
- AT Augusta Military Medical Center, Fort Belvoir, VA 22060, USA
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Intramural Research Program, NIA/NIH/IRP, Baltimore, MD 21224, USA
| | - Osorio Meirelles
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Intramural Research Program, NIA/NIH/IRP, Baltimore, MD 21224, USA
| | - Zhiguang Li
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Intramural Research Program, NIA/NIH/IRP, Baltimore, MD 21224, USA
| | - Ziad W. El-Hajj
- Department of Biology, McGill University, Montreal, QC, Canada
| | - Jordan Weiss
- Stanford Center on Longevity, Stanford University, Palo Alto, CA 94305, USA
| | - Christian A. Maino Vieytes
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Intramural Research Program, NIA/NIH/IRP, Baltimore, MD 21224, USA
| | - Lenore J. Launer
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Intramural Research Program, NIA/NIH/IRP, Baltimore, MD 21224, USA
| | - Michele K. Evans
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Intramural Research Program, NIA/NIH/IRP, Baltimore, MD 21224, USA
| | - Alan B. Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Intramural Research Program, NIA/NIH/IRP, Baltimore, MD 21224, USA
| |
Collapse
|
49
|
Collu R, Giunti E, Daley S, Chen M, Xia W. Angiotensin-converting enzyme inhibitors and statins therapies-induced changes in omics profiles in humans and transgenic tau mice. Biomed Pharmacother 2023; 168:115756. [PMID: 37865996 DOI: 10.1016/j.biopha.2023.115756] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023] Open
Abstract
BACKGROUND Hypertension and hyperlipidemia are considered risk factors for Alzheimer's disease (AD) and other related dementias. Clinically approved medications typically prescribed to manage these conditions have shown an association with reduced risk of developing AD and could be explored as potential repurposed therapeutics. OBJECTIVE We aimed to explore the effects of the pharmacological treatment with angiotensin-converting enzyme inhibitors (ACEI) and statins (STAT) on AD-related neuropathology and the potential benefits of their concurrent use. METHODS We investigated the effect of ACEI, STAT or combination of both by exploring the transcriptomic, proteomic and tau pathology profiles after treatment in both human patients and in P301S transgenic mice (PS19) modeling tauopathies and AD. We performed bioinformatic analysis of enriched pathways after treatment. RESULTS Proteomics and transcriptomics analysis revealed proteins and genes whose expression is significantly changed in subjects receiving treatment with ACEI, STAT or combined drugs. In mice, treatment with the ACEI lisinopril significantly decreased brain levels of total tau (Tau) and phosphorylated tau (pTau)-181, while the STAT atorvastatin significantly reduced the levels of pTau-396. The combined therapy with lisinopril and atorvastatin significantly decreased Tau. Moreover, brain levels of lisinopril were negatively correlated with Tau. Among the others, CD200, ADAM22, BCAN and NCAM1 were significantly affected by treatments in both human subjects and transgenic mice. CONCLUSIONS Our findings provide significant information that may guide future investigation of the potential use of ACEI, STAT, or the combination of the two drug classes as repurposed therapies or preventive strategies for AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Roberto Collu
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, United States; Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
| | - Elisa Giunti
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, United States; Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
| | - Sarah Daley
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, United States; Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
| | - Mei Chen
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, United States
| | - Weiming Xia
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, United States; Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States; Department of Biological Sciences, University of Massachusetts Kennedy College of Science, Lowell, MA, United States.
| |
Collapse
|
50
|
Cederroth CR, Hong MG, Freydin MB, Edvall NK, Trpchevska N, Jarach C, Schlee W, Schwenk JM, Lopez-Escamez JA, Gallus S, Canlon B, Bulla J, Williams FMK. Screening for Circulating Inflammatory Proteins Does Not Reveal Plasma Biomarkers of Constant Tinnitus. J Assoc Res Otolaryngol 2023; 24:593-606. [PMID: 38079022 PMCID: PMC10752855 DOI: 10.1007/s10162-023-00920-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/22/2023] [Indexed: 12/29/2023] Open
Abstract
BACKGROUND AND OBJECTIVE Tinnitus would benefit from an objective biomarker. The goal of this study is to identify plasma biomarkers of constant and chronic tinnitus among selected circulating inflammatory proteins. METHODS A case-control retrospective study on 548 cases with constant tinnitus and 548 matched controls from the Swedish Tinnitus Outreach Project (STOP), whose plasma samples were examined using Olink's Inflammatory panel. Replication and meta-analysis were performed using the same method on samples from the TwinsUK cohort. Participants from LifeGene, whose blood was collected in Stockholm and Umeå, were recruited to STOP for a tinnitus subtyping study. An age and sex matching was performed at the individual level. TwinsUK participants (n = 928) were selected based on self-reported tinnitus status over 2 to 10 years. Primary outcomes include normalized levels for 96 circulating proteins, which were used as an index test. No reference standard was available in this study. RESULTS After adjustment for age, sex, BMI, smoking, hearing loss, and laboratory site, the top proteins identified were FGF-21, MCP4, GDNF, CXCL9, and MCP-1; however, these were no longer statistically significant after correction for multiple testing. Stratification by sex did not yield any significant associations. Similarly, associations with hearing loss or other tinnitus-related comorbidities such as stress, anxiety, depression, hyperacusis, temporomandibular joint disorders, and headache did not yield any significant associations. Analysis in the TwinsUK failed in replicating the top candidates. Meta-analysis of STOP and TwinsUK did not reveal any significant association. Using elastic net regularization, models exhibited poor predictive capacity tinnitus based on inflammatory markers [sensitivity = 0.52 (95% CI 0.47-0.57), specificity = 0.53 (0.48-0.58), positive predictive value = 0.52 (0.47-0.56), negative predictive values = 0.53 (0.49-0.58), and AUC = 0.53 (0.49-0.56)]. DISCUSSION Our results did not identify significant associations of the selected inflammatory proteins with constant tinnitus. Future studies examining longitudinal relations among those with more severe tinnitus and using more recent expanded proteomics platforms and sampling of cerebrospinal fluid could increase the likelihood of identifying relevant molecular biomarkers.
Collapse
Affiliation(s)
- Christopher R Cederroth
- Section of Experimental Audiology, Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden.
- National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust, Ropewalk House, Nottingham, UK.
- Department of Otolaryngology, Head and Neck Surgery, Translational Hearing Research, Tübingen Hearing Research Center, University of Tübingen, Tubingen, Germany.
| | - Mun-Gwan Hong
- Affinity Proteomics, Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
- Science for Life Laboratory, Department of Biochemistry and Biophysics, National Bioinformatics Infrastructure Sweden, Stockholm University, Stockholm, Sweden
| | - Maxim B Freydin
- Department of Twin Research and Genetic Epidemiology, School of Life Course Sciences, King's College London, London, UK
| | - Niklas K Edvall
- Section of Experimental Audiology, Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Natalia Trpchevska
- Section of Experimental Audiology, Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Carlotta Jarach
- Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Winfried Schlee
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Jochen M Schwenk
- Science for Life Laboratory, Department of Biochemistry and Biophysics, National Bioinformatics Infrastructure Sweden, Stockholm University, Stockholm, Sweden
| | - Jose-Antonio Lopez-Escamez
- Faculty of Medicine & Health, School of Medical Sciences, Meniere's Disease Neuroscience Research Program, The Kolling Institute, University of Sydney, Sydney, NSW, Australia
- Otology and Neurotology Group CTS495, Department of Genomic Medicine, GENYO - Centre for Genomics and Oncological Research - Pfizer, University of Granada, PTS, Junta de Andalucía, Granada, Spain
- Division of Otolaryngology, Department of Surgery, Instituto de Investigación Biosanitaria, ibs.GRANADA, Universidad de Granada, GranadaGranada, Spain
| | - Silvano Gallus
- Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Barbara Canlon
- Section of Experimental Audiology, Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Jan Bulla
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
- Department of Mathematics, University of Bergen, Bergen, Norway
| | - Frances M K Williams
- Department of Twin Research and Genetic Epidemiology, School of Life Course Sciences, King's College London, London, UK
| |
Collapse
|