1
|
Matlyuba Jakhonkulovna S, Bahodirova Kamolovna G, Zokirov M, Umida Tajimuratovna B, Yumashev A, Shichiyakh R, Safarova NI, Nargiza Nusratovna A, Esanmuradova N, Muyassar Karimbaevna T, Lazizakhon A, Ishankulov A. Electrochemical biosensors for early detection of Alzheimer's disease. Clin Chim Acta 2025; 572:120278. [PMID: 40185381 DOI: 10.1016/j.cca.2025.120278] [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: 03/15/2025] [Revised: 03/29/2025] [Accepted: 03/30/2025] [Indexed: 04/07/2025]
Abstract
In recent years, electrochemical biosensors have shown great promise as innovative tools for the early identification of Alzheimer's disease (AD), a neurodegenerative disorder that severely affects cognitive ability and overall quality of life. This comprehensive review aims to consolidate the latest research on the creation and implementation of electrochemical biosensors designed to detect AD-related biomarkers. We examine cutting-edge approaches to surface modification that enhance the attachment of biorecognition molecules, thus enabling the simultaneous identification of multiple biomarkers. This review emphasizes the crucial role that electrochemical biosensors play in the early diagnosis of Alzheimer's disease, highlighting their potential to revolutionize clinical practices by facilitating timely interventions. In the future, research efforts should concentrate on refining these technologies for widespread clinical adoption, ensuring that they meet the needs of both healthcare professionals and patients.
Collapse
Affiliation(s)
| | - Gulnoz Bahodirova Kamolovna
- Department of Scientific Research, Innovations and Scientific and Pedagogical Personnel Training International School of Finance Technology and Science (ISFT Institute), Uzbekistan
| | | | | | - Alexey Yumashev
- Department of Prosthetic Dentistry, Doctor of Medicine, Professor Sechenov First Moscow State Medical University, Russia
| | - Rustem Shichiyakh
- Department of Management, Candidate of Economic Sciences, Associate Professor. Kuban State Agrarian University named after I.T. Trubilin, Krasnodar, Russia
| | - Nasiba I Safarova
- Department of Otorhinolaryngology, Faculty of Postgraduate Education, Samarkand State Medical University, Samarkand, Uzbekistan
| | | | - Nilufar Esanmuradova
- "Tashkent Institute of Irrigation and Agricultural Mechanization Engineers" National Research University, Tashkent, Uzbekistan; Western Caspian University, Scientific Researcher, Baku, Azerbaijan
| | - Tadjibaeva Muyassar Karimbaevna
- Department of Zoology, Human Morphophysiology and Nutrition (PhD), Nukus State Pedagogical Institute Named After Ajiniyaz, Uzbekistan
| | - Alidjanova Lazizakhon
- International Islamic Academy of Uzbekistan, Senior Lecturer of "UNESCO Chair on Religious Studies and the Comparative Study of World Religions", Kadiri st. 11, Tashkent, Uzbekistan
| | - Alisher Ishankulov
- Samarkand State University named after Sharof Rashidov, Uzbekistan; Kimyo International University in Tashkent, Branch Samarkand, Uzbekistan
| |
Collapse
|
2
|
Nemati R, Sohrabi-Ashlaghi A, Saberian P, Sadeghi M, Mardani S, Abadi ASJH, Yaghoobpoor A, Heydari A, Khoshroo N, Rahnama Y, Mayeli M, Nasiri H. Growth associated protein 43 (GAP-43) predicts brain amyloidosis in Alzheimer's dementia continuum: an [ 18 F] AV-45 study. BMC Neurol 2025; 25:134. [PMID: 40170060 PMCID: PMC11959801 DOI: 10.1186/s12883-025-04140-5] [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: 08/30/2024] [Accepted: 03/17/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND Growth-associated protein 43 (GAP-43) is a key protein involved in neuronal growth and synaptic plasticity. Alterations in GAP-43 levels have been associated with Alzheimer's Disease (AD), potentially reflecting synaptic dysfunction. We evaluated the potential of GAP-43 as a biomarker for AD and explored its association with amyloid-beta (Aβ) levels, as well as its correlation with Aβ plaque burden in the brain. METHODS We screened 1,639 participants from the Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort. A total of 226 individuals met the eligibility criteria and were enrolled. Participants were classified into three groups: 77 cognitively normal (CN) individuals, 111 with mild cognitive impairment (MCI), and 38 with a diagnosis of AD. The associations between cerebrospinal fluid (CSF) GAP-43 levels with other biomarkers as well as [¹⁸F] AV-45 (Florbetapir) PET Standardized Uptake Value Ratios (SUVR) were investigated. RESULTS Our findings revealed significantly elevated CSF GAP-43 levels in individuals with AD compared to CN and MCI groups. Furthermore, GAP-43 levels showed a significant positive correlation with tau pathology. Notably, we observed a significant association between GAP-43 and [¹⁸F] Florbetapir PET SUVR in the MCI group, suggesting that GAP-43 may serve as a reliable biomarker in the early stages of AD. CONCLUSION This study provides evidence supporting the role of GAP-43 as a potential biomarker for AD, particularly in relation to predicting the amyloid pathology pattern in the brain in the MCI stage.
Collapse
Affiliation(s)
- Rezvan Nemati
- Department of Psychology, Islamic Azad University Arak branch, Arak, Iran
| | - Ahmadreza Sohrabi-Ashlaghi
- Department of Physiology, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Parsa Saberian
- Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Hormozgan Province, 7916969573, Iran.
| | - Mohammad Sadeghi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- School of Rehabilitation, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sajjad Mardani
- School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | | | - Ali Yaghoobpoor
- Student's Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Niloofar Khoshroo
- School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Yassin Rahnama
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Mayeli
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamide Nasiri
- Student Research Committee, School of Medicine, Zanjan University of Medical Science, Zanjan, Iran.
| |
Collapse
|
3
|
Smallwood MJ, Alghayth MA, Knight AR, Tveen-Jensen K, Pitt AR, Spickett CM, Llewellyn D, Pula G, Wearn A, Vanhatalo A, Jones AM, Francis P, Coulthard E, Kehoe PG, Winyard PG. Hemoglobin in the brain frontal lobe tissue of patients with Alzheimer's disease is susceptible to reactive nitrogen species-mediated oxidative damage. Redox Biol 2025; 82:103612. [PMID: 40184643 DOI: 10.1016/j.redox.2025.103612] [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: 02/19/2025] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/07/2025] Open
Abstract
Brain inflammation in Alzheimer's disease (AD) involves reactive nitrogen species (RNS) generation. Protein contents of 3-nitrotyrosine, a product of RNS generation, were assessed in frontal lobe brain homogenates from patients with AD, patients with vascular dementia (VaD) and non-dementia (ND) controls. Western blotting revealed a dominant 15 kDa nitrated protein band in both dementia (AD/VaD) and ND frontal lobe brain tissue. Surprisingly, this protein band was identified by mass spectrometry as hemoglobin, an erythrocytic protein. The same band stained positively when western blotted using an anti-hemoglobin antibody. On western blots, the median (IQR) normalized staining intensity for 3-nitrotyrosine in hemoglobin was increased in both AD [1.71 (1.20-3.05) AU] and VaD [1.50 (0.59-3.04) AU] brain tissue compared to ND controls [0.41 (0.09-0.75) AU] (Mann-Whitney U test: AD v ND, P < 0.0005; VaD v ND, P < 0.05; n = 11). The median normalized staining of the nitrated hemoglobin band was higher in advanced AD patients compared with early-stage AD (P < 0.005). The median brain tissue NO2- levels (nmol/mg protein) were significantly higher in AD samples than in ND controls (P < 0.05). Image analysis of western blots of lysates from peripheral blood erythrocytes suggested that hemoglobin nitration was increased in AD compared to ND (P < 0.05; n = 4 in each group). Total protein-associated 3-nitrotyrosine was measured by an electrochemiluminescence-based immunosorbent assay, but showed no statistically significant differences between AD, VaD and ND. Females showed larger increases in hemoglobin nitration and NO2- levels between disease and control groups compared to males, although the group sizes in these sub-analyses were small. In conclusion, the extent of hemoglobin nitration was increased in AD and VaD brain frontal lobe tissue compared with ND. We propose that reactive nitrogen species-mediated damage to hemoglobin may be involved in the pathogenesis of AD.
Collapse
Affiliation(s)
- M J Smallwood
- University of Exeter Medical School, Exeter, EX1 2LU, UK
| | - M Abu Alghayth
- University of Exeter Medical School, Exeter, EX1 2LU, UK; Current Address: Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha, P.O. Box 255, 67714, Saudi Arabia
| | - A R Knight
- University of Exeter Medical School, Exeter, EX1 2LU, UK
| | - K Tveen-Jensen
- College of Health & Life Sciences, Aston University, Birmingham, B4 7ET, UK
| | - A R Pitt
- College of Health & Life Sciences, Aston University, Birmingham, B4 7ET, UK
| | - C M Spickett
- College of Health & Life Sciences, Aston University, Birmingham, B4 7ET, UK
| | - D Llewellyn
- University of Exeter Medical School, Exeter, EX1 2LU, UK
| | - G Pula
- University of Exeter Medical School, Exeter, EX1 2LU, UK; Centre for Biomedicine, Hull York Medical School, Hull, HU6 7RX, UK
| | - A Wearn
- Translational Health Sciences, Bristol Medical School, University of Bristol, Southmead Hospital, Bristol, BS10 5NB, UK
| | - A Vanhatalo
- University of Exeter Medical School, Exeter, EX1 2LU, UK
| | - A M Jones
- University of Exeter Medical School, Exeter, EX1 2LU, UK
| | - P Francis
- University of Exeter Medical School, Exeter, EX1 2LU, UK; Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, WC2R 2LS, UK
| | - E Coulthard
- Translational Health Sciences, Bristol Medical School, University of Bristol, Southmead Hospital, Bristol, BS10 5NB, UK
| | - P G Kehoe
- Translational Health Sciences, Bristol Medical School, University of Bristol, Southmead Hospital, Bristol, BS10 5NB, UK
| | - P G Winyard
- University of Exeter Medical School, Exeter, EX1 2LU, UK.
| |
Collapse
|
4
|
Liu Y, Liu X, Che P, Wang Y, Piao Z, Wang Y, Cai L, Xing M, Xu Y, Sun W, Wang Y, Zhang N. A cytometric bead array for the measurement of plasma biomarker levels in patients with Alzheimer's disease. Sci Rep 2025; 15:9767. [PMID: 40118895 PMCID: PMC11928653 DOI: 10.1038/s41598-024-83919-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: 09/10/2024] [Accepted: 12/18/2024] [Indexed: 03/24/2025] Open
Abstract
Identifying plasma biomarkers for Alzheimer's disease (AD) has garnered strong interest. In this study, a cytometric bead array (CBA) method for measuring the levels of amyloid-β (Aβ) peptides and phosphorylated tau (P-tau) was evaluated. Fifty patients with cognitive impairment (CI) and 22 cognitively unimpaired (CU) controls were recruited. CI patients were classified into Aβ + and Aβ - groups according to amyloid positron emission tomography (PET) scan results. Biomarker levels in the plasma of all participants and in the cerebrospinal fluid (CSF) of 28 CI patients were measured via CBA. Plasma P-tau181 levels were greatest in the Aβ + CI group and showed excellent performance in differentiating Aβ + CI patients from CU controls. The plasma and CSF levels of P-tau181 were correlated with each other and had similar diagnostic performance for distinguishing between Aβ + CI patients and Aβ- CI patients. Overall, CBA is a potential cost-effective method for measuring plasma biomarkers, particularly P-tau181, in AD patients.
Collapse
Affiliation(s)
- Ye Liu
- Department of Neurology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Tianjin Neurological Institute, Ministry of Education, Variations and Regeneration of Nervous System, Tianjin, China
| | - Xin Liu
- Department of Neurology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Department of Neurology, Affiliated Hospital of Hebei University, Hebei, China
| | - Ping Che
- Department of Neurology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Yu Wang
- Department of Neurology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Zhiyan Piao
- Department of Neurology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Ying Wang
- PET/CT Center, Tianjin Medical University General Hospital, Tianjin, China
| | - Li Cai
- PET/CT Center, Tianjin Medical University General Hospital, Tianjin, China
| | - Mengya Xing
- Department of Neurology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Yanwei Xu
- Department of Neurology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Wenhao Sun
- Department of Neurology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Yue Wang
- Department of Neurology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Nan Zhang
- Department of Neurology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
- Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Tianjin Neurological Institute, Ministry of Education, Variations and Regeneration of Nervous System, Tianjin, China.
| |
Collapse
|
5
|
Parker CS, Oxtoby NP, Young AL. Parsimonious EBM: Generalising the event-based model of disease progression for simultaneous events. Neuroimage 2025; 311:121162. [PMID: 40118234 DOI: 10.1016/j.neuroimage.2025.121162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 02/09/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025] Open
Abstract
The event-based model of disease progression (EBM) infers a temporal ordering of biomarker abnormalities, defining different disease stages, from cross-sectional data. A key modelling choice of the EBM is that biomarker abnormalities, termed events, are serially ordered. However, this choice enforces a strict equality between the number of input biomarkers and the number of modelled disease stages, limiting the EBM's ability to infer simple staging systems and identify latent disease processes driving multiple biomarker changes. To overcome this, we introduce the parsimonious event-based model of disease progression (P-EBM). The P-EBM generalises the EBM to allow multiple new biomarker abnormalities, termed "simultaneous events", at each model stage. We evaluate the P-EBM performance in simulated data and demonstrate its ability to reconstruct event orderings with arbitrary arrangements under realistic experimental conditions. In sporadic AD data from the Alzheimer's Disease Neuroimaging Initiative, the P-EBM estimated a sequence with 7 model stages from a dataset of 12 biomarkers that more closely fitted the data than the EBM. The inferred sets of simultaneous events, such as decreased cerebrospinal fluid total tau and p-tau181, correspond closely to known underlying disease processes. P-EBM patient stages were strongly associated with clinical diagnosis at baseline and future conversion and could be accurately estimated from a smaller number of biomarkers than the EBM. The P-EBM enables the data-driven discovery of simple disease staging systems which could highlight new latent disease processes and suggest practical strategies for patient staging.
Collapse
Affiliation(s)
- C S Parker
- UCL Hawkes Institute, Department of Computer Science, UCL, London, UK.
| | - N P Oxtoby
- UCL Hawkes Institute, Department of Computer Science, UCL, London, UK
| | - A L Young
- UCL Hawkes Institute, Department of Computer Science, UCL, London, UK
| |
Collapse
|
6
|
Thal DR, Poesen K, Vandenberghe R, De Meyer S. Alzheimer's disease neuropathology and its estimation with fluid and imaging biomarkers. Mol Neurodegener 2025; 20:33. [PMID: 40087672 PMCID: PMC11907863 DOI: 10.1186/s13024-025-00819-y] [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: 07/10/2024] [Accepted: 02/26/2025] [Indexed: 03/17/2025] Open
Abstract
Alzheimer's disease (AD) is neuropathologically characterized by the extracellular deposition of the amyloid-β peptide (Aβ) and the intraneuronal accumulation of abnormal phosphorylated tau (τ)-protein (p-τ). Most frequently, these hallmark lesions are accompanied by other co-pathologies in the brain that may contribute to cognitive impairment, such as vascular lesions, intraneuronal accumulation of phosphorylated transactive-response DNA-binding protein 43 (TDP-43), and/or α-synuclein (αSyn) aggregates. To estimate the extent of these AD and co-pathologies in patients, several biomarkers have been developed. Specific tracers target and visualize Aβ plaques, p-τ and αSyn pathology or inflammation by positron emission tomography. In addition to these imaging biomarkers, cerebrospinal fluid, and blood-based biomarker assays reflecting AD-specific or non-specific processes are either already in clinical use or in development. In this review, we will introduce the pathological lesions of the AD brain, the related biomarkers, and discuss to what extent the respective biomarkers estimate the pathology determined at post-mortem histopathological analysis. It became evident that initial stages of Aβ plaque and p-τ pathology are not detected with the currently available biomarkers. Interestingly, p-τ pathology precedes Aβ deposition, especially in the beginning of the disease when biomarkers are unable to detect it. Later, Aβ takes the lead and accelerates p-τ pathology, fitting well with the known evolution of biomarker measures over time. Some co-pathologies still lack clinically established biomarkers today, such as TDP-43 pathology or cortical microinfarcts. In summary, specific biomarkers for AD-related pathologies allow accurate clinical diagnosis of AD based on pathobiological parameters. Although current biomarkers are excellent measures for the respective pathologies, they fail to detect initial stages of the disease for which post-mortem analysis of the brain is still required. Accordingly, neuropathological studies remain essential to understand disease development especially in early stages. Moreover, there is an urgent need for biomarkers reflecting co-pathologies, such as limbic predominant, age-related TDP-43 encephalopathy-related pathology, which is known to modify the disease by interacting with p-τ. Novel biomarker approaches such as extracellular vesicle-based assays and cryptic RNA/peptides may help to better detect these co-pathologies in the future.
Collapse
Affiliation(s)
- Dietmar Rudolf Thal
- Department of Imaging and Pathology, Laboratory for Neuropathology, Leuven Brain Institute, KU Leuven, Herestraat 49, Leuven, 3000, Belgium.
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium.
| | - Koen Poesen
- Department of Neurosciences, Laboratory for Molecular Neurobiomarker Research, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Rik Vandenberghe
- Department of Neurosciences, Laboratory for Cognitive Neurology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Steffi De Meyer
- Department of Neurosciences, Laboratory for Molecular Neurobiomarker Research, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Neurosciences, Laboratory for Cognitive Neurology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| |
Collapse
|
7
|
Schneeberger S, Kim SJ, Geesdorf MN, Friebel E, Eede P, Jendrach M, Boltengagen A, Braeuning C, Ruhwedel T, Hülsmeier AJ, Gimber N, Foerster M, Obst J, Andreadou M, Mundt S, Schmoranzer J, Prokop S, Kessler W, Kuhlmann T, Möbius W, Nave KA, Hornemann T, Becher B, Edgar JM, Karaiskos N, Kocks C, Rajewsky N, Heppner FL. Interleukin-12 signaling drives Alzheimer's disease pathology through disrupting neuronal and oligodendrocyte homeostasis. NATURE AGING 2025:10.1038/s43587-025-00816-2. [PMID: 40082619 DOI: 10.1038/s43587-025-00816-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 01/23/2025] [Indexed: 03/16/2025]
Abstract
Neuroinflammation including interleukin (IL)-12/IL-23-signaling is central to Alzheimer's disease (AD) pathology. Inhibition of p40, a subunit of IL-12/IL-23, attenuates pathology in AD-like mice; however, its signaling mechanism and expression pattern remained elusive. Here we show that IL-12 receptors are predominantly expressed in neurons and oligodendrocytes in AD-like APPPS1 mice and in patients with AD, whereas IL-23 receptor transcripts are barely detectable. Consistently, deletion of the IL-12 receptor in neuroectodermal cells ameliorated AD pathology in APPPS1 mice, whereas removal of IL-23 receptors had no effect. Genetic ablation of IL-12 signaling alone reverted the loss of mature oligodendrocytes, restored myelin homeostasis, rescued the amyloid-β-dependent reduction of parvalbumin-positive interneurons and restored phagocytosis-related changes in microglia of APPPS1 mice. Furthermore, IL-12 protein expression was increased in human AD brains compared to healthy age-matched controls, and human oligodendrocyte-like cells responded profoundly to IL-12 stimulation. We conclude that oligodendroglial and neuronal IL-12 signaling, but not IL-23 signaling, are key in orchestrating AD-related neuroimmune crosstalk and that IL-12 represents an attractive therapeutic target in AD.
Collapse
Affiliation(s)
- Shirin Schneeberger
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Seung Joon Kim
- Systems Biology of Gene Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Maria N Geesdorf
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ekaterina Friebel
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Pascale Eede
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Marina Jendrach
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Anastasiya Boltengagen
- Systems Biology of Gene Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Caroline Braeuning
- Genomics Platform, Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Torben Ruhwedel
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Department of Neurogenetics, Electron Microscopy Unit City Campus, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | | | - Niclas Gimber
- AMBIO Advanced Medical Bioimaging Core Facility, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marlene Foerster
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Juliane Obst
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Myrto Andreadou
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Sarah Mundt
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Jan Schmoranzer
- AMBIO Advanced Medical Bioimaging Core Facility, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Stefan Prokop
- Department of Pathology, College of Medicine, University of Florida, Gainesville, FL, USA
- Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA
- Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
| | - Wiebke Kessler
- Institute of Neuropathology, University Hospital Münster, Münster, Germany
| | - Tanja Kuhlmann
- Institute of Neuropathology, University Hospital Münster, Münster, Germany
| | - Wiebke Möbius
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Department of Neurogenetics, Electron Microscopy Unit City Campus, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Thorsten Hornemann
- Institute of Clinical Chemistry, University of Zürich, Zürich, Switzerland
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Julia M Edgar
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Nikos Karaiskos
- Systems Biology of Gene Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Christine Kocks
- Systems Biology of Gene Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Nikolaus Rajewsky
- Systems Biology of Gene Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.
- Cluster of Excellence, NeuroCure, Berlin, Germany.
- German Cancer Consortium (DKTK), Heidelberg, Germany.
- German Center for Cardiovascular Research (DZHK), Berlin, Germany.
- National Center for Tumor Diseases (NCT), Berlin, Germany.
- Charité - Universitätsmedizin, Berlin, Germany.
| | - Frank L Heppner
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
- Cluster of Excellence, NeuroCure, Berlin, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany.
| |
Collapse
|
8
|
Wang Z, Chen Y, Gong K, Zhao B, Ning Y, Chen M, Li Y, Ali M, Timsina J, Liu M, Cruchaga C, Jia J. Cerebrospinal fluid proteomics identification of biomarkers for amyloid and tau PET stages. Cell Rep Med 2025:102031. [PMID: 40118053 DOI: 10.1016/j.xcrm.2025.102031] [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: 09/24/2024] [Revised: 01/15/2025] [Accepted: 02/24/2025] [Indexed: 03/23/2025]
Abstract
Accurate staging of Alzheimer's disease (AD) pathology is crucial for therapeutic trials and prognosis, but existing fluid biomarkers lack specificity, especially for assessing tau deposition severity, in amyloid-beta (Aβ)-positive patients. We analyze cerebrospinal fluid (CSF) samples from 136 participants in the Alzheimer's Disease Neuroimaging Initiative using more than 6,000 proteins. We apply machine learning to predict AD pathological stages defined by amyloid and tau positron emission tomography (PET). We identify two distinct protein panels: 16 proteins, including neurofilament heavy chain (NEFH) and SPARC-related modular calcium-binding protein 1 (SMOC1), that distinguished Aβ-negative/tau-negative (A-T-) from A+ individuals and nine proteins, such as HCLS1-associated protein X-1 (HAX1) and glucose-6-phosphate isomerase (GPI), that differentiated A+T+ from A+T- stages. These signatures outperform the established CSF biomarkers (area under the curve [AUC]: 0.92 versus 0.67-0.70) and accurately predicted disease progression over a decade. The findings are validated in both internal and external cohorts. These results underscore the potential of proteomic-based signatures to refine AD diagnostic criteria and improve patient stratification in clinical trials.
Collapse
Affiliation(s)
- Zhibo Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, P.R.China
| | - Yuhan Chen
- The First Clinical Medical School, Hebei North University, Zhangjiakou 075000, China
| | - Katherine Gong
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, USA
| | - Bote Zhao
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, P.R.China
| | - Yuye Ning
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, P.R.China
| | - Meilin Chen
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, P.R.China
| | - Yan Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, P.R.China
| | - Muhammad Ali
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, USA
| | - Jigyasha Timsina
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, USA
| | - Menghan Liu
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Hope Center for Neurologic Diseases, Washington University, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO, USA.
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, P.R.China; Beijing Key Laboratory of Geriatric Cognitive Disorders, Beijing 100053, P.R.China; Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, Beijing 100053, P.R.China; Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100053, P.R.China; Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, P.R.China.
| |
Collapse
|
9
|
Liakakis G, Vakrakou AG, Boufidou F, Constantinides V, Velonakis G, Paraskevas GP, Stefanis L, Kapaki E. Exploratory Analysis of Cerebrospinal Fluid IL-6 and IL-17A Levels in Subcortical Small-Vessel Disease Compared to Alzheimer's Disease: A Pilot Study. Diagnostics (Basel) 2025; 15:669. [PMID: 40150012 PMCID: PMC11941723 DOI: 10.3390/diagnostics15060669] [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: 12/17/2024] [Revised: 01/12/2025] [Accepted: 03/04/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Low-grade inflammation in the form of microglial activation may be involved in neurodegenerative and vascular dementias. Subcortical small-vessel disease (SSVD) is the main form of vascular dementia, associated with brain barrier dysfunction and endothelial and monocyte activation. IL-6 and IL-17A are known proinflammatory cytokines that contribute to the disruption of blood-brain barrier integrity and microvascular dysfunction, features that are central to SSVD pathophysiological pathways. We herein compared cerebrospinal fluid (CSF) IL-6 and IL-17A concentrations in SSVD and AD patients as well as control subjects and examined the potential associations among IL-6 and IL-17A levels with cognitive and ΜRΙ changes. The albumin quotient (Qalb) was also calculated. Methods: CSF IL-6 and IL-17A (18 SSVD, 17 AD, and 12 healthy controls) were measured with solid-phase sandwich ELISAs, while albumin levels were measured by immunonephelometry. MMSE, FAB, and the CLOX tests were used for cognitive assessment and MRI was used for atrophy and white matter hyperintensities. Results: Significantly elevated CSF levels of Qalb and IL-6 were found in SSVD patients compared to both AD (p = 0.02) and controls (p = 0.002), respectively. Moreover, CSF IL-6 levels displayed a significant inverse correlation with CLOX2 scores (r = -0.641, p = 0.02), as well as a positive correlation with the total normalized CSF volume (r = 0.7, p = 0.01). CSF IL-17A levels were found to be reduced in SSVD patients, compared to controls and AD patients (p < 0.0001 and p = 0.002, respectively). The IL-6/IL-17A ratio with a cut-off value > 1.004 displayed a sensitivity of 83.33% (95%CI; 60.78% to 94.16%) and a specificity of 68.97% (95%CI; 50.77% to 82.72%) for the discrimination of SSVD from AD patients and controls. Conclusions: In the present pilot single-center study, we found increased CSF IL-6 and IL-6/IL-17A ratio levels in SSVD patients that correlated with reduced scores in the CLOX2 test and increased CSF volume. These preliminary findings deserve further evaluation in larger cohorts in order to elucidate their potential as surrogate biomarkers for the discrimination of SSVD from AD pathology.
Collapse
Affiliation(s)
- Georgios Liakakis
- Neurochemistry and Biological Markers Unit, 1st Department of Neurology, School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens, Vass. Sophias Ave. 74, 11528 Athens, Greece; (G.L.); (F.B.); (V.C.)
| | - Aigli G. Vakrakou
- Laboratory of Neuroimmunology, First Department of Neurology, Aeginition Hospital, National and Kapodistrian, University of Athens, 21 Papadiamantopoulou, Ilisia, 11528 Athens, Greece;
| | - Fotini Boufidou
- Neurochemistry and Biological Markers Unit, 1st Department of Neurology, School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens, Vass. Sophias Ave. 74, 11528 Athens, Greece; (G.L.); (F.B.); (V.C.)
| | - Vasilios Constantinides
- Neurochemistry and Biological Markers Unit, 1st Department of Neurology, School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens, Vass. Sophias Ave. 74, 11528 Athens, Greece; (G.L.); (F.B.); (V.C.)
| | - Georgios Velonakis
- Research Unit of Radiology, 2nd Department of Radiology, Medical School, “Attikon” University General Hospital, National and Kapodistrian University of Athens, Rimini 1, Chaidari, 12462 Athens, Greece
| | - George P. Paraskevas
- 2nd Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, Rimini 1, 12462 Athens, Greece;
| | - Leonidas Stefanis
- 1st Department of Neurology, School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens, 74 Vass. Sophias Ave., 11528 Athens, Greece;
| | - Elisabeth Kapaki
- Neurochemistry and Biological Markers Unit, 1st Department of Neurology, School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens, Vass. Sophias Ave. 74, 11528 Athens, Greece; (G.L.); (F.B.); (V.C.)
| |
Collapse
|
10
|
Xu X, Kwon J, Yan R, Apio C, Song S, Heo G, Yang Q, Timsina J, Liu M, Budde J, Blennow K, Zetterberg H, Lleó A, Ruiz A, Molinuevo JL, Lee VMY, Deming Y, Heslegrave AJ, Hohman TJ, Pastor P, Peskind ER, Albert MS, Morris JC, Park T, Cruchaga C, Sung YJ. Sex Differences in Apolipoprotein E and Alzheimer Disease Pathology Across Ancestries. JAMA Netw Open 2025; 8:e250562. [PMID: 40067298 PMCID: PMC11897841 DOI: 10.1001/jamanetworkopen.2025.0562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 01/05/2025] [Indexed: 03/15/2025] Open
Abstract
Importance Age, sex, and apolipoprotein E (APOE) are the strongest risk factors for late-onset Alzheimer disease (AD). The role of APOE in AD varies with sex and ancestry. While the association of APOE with AD biomarkers also varies across sex and ancestry, no study has systematically investigated both sex-specific and ancestry differences of APOE on cerebrospinal fluid (CSF) biomarkers together, resulting in limited insights and generalizability. Objective To systematically investigate the association of sex and APOE-ε4 with 3 core CSF biomarkers across ancestries. Design, Setting, and Participants This cohort study examined 3 CSF biomarkers (amyloid β1-42 [Aβ42], phosphorylated tau 181 [p-tau], and total tau, in participants from 20 cohorts from July 1, 1985, to March 31, 2020. These individuals were grouped into African, Asian, and European ancestries based on genetic data. Data analyses were conducted from June 1, 2023, to November 10, 2024. Exposure Sex (male or female) and APOE-ε4. Main Outcomes and Measures The associations of sex and APOE-ε4 with biomarker levels were assessed within each ancestry group, adjusting for age. Meta-analyses were performed to identify these associations across ancestries. Sensitivity analyses were conducted to exclude the potential influence of the APOE-ε2 allele. Results This cohort study included 4592 individuals (mean [SD] age, 70.8 [10.2] years; 2425 [52.8%] female; 119 [2.6%] African, 52 [1.1%] Asian, and 4421 [96.3%] European). Higher APOE-ε4 dosage scores were associated with lower Aβ42 values (β [SE], -0.58 [0.02], P < .001), indicating more severe pathology; these associations were seen in men and women separately and jointly. The association with APOE-ε4 was statistically greater in men (β [SE], -0.63 [0.03]; P < .001) vs women (β [SE], -0.52 [0.03]; P < .001) of European ancestry (P = .01 for interaction). Women had higher levels of p-tau, indicating more severe neurofibrillary pathology. The association between APOE-ε4 dosage and p-tau was in the expected direction (higher APOE-ε4 dosage for higher p-tau values) in both sexes, but the difference between sexes was significant only in those of African ancestry (β [SE], 0.10 [0.18]; P = .57 for men; β [SE], 0.66 [0.17]; P < .001 for women; P = .03 for interaction). Women also had higher levels of total tau, indicating more neuronal damage. The association between APOE-ε4 dosage and total tau was stronger in women than in men in the African cohort (β [SE], 0.20 [0.22]; P = .36 for men and β [SE], 0.65 [0.22], P = .004 for women [P = .16 for interaction]) and European cohort (β [SE], 0.36 [0.03]; P < .001 in women and β [SE], 0.27 [0.03], P < .001 in men [P = .053 for interaction]); no significant associations were found in the Asian cohort. Sensitivity analysis excluding APOE-ε2 carriers yielded similar results. Conclusions and Relevance In this cohort study, the association of the APOE-ε4 risk allele with tau accumulation was higher in women than in men. These findings underscore the importance of considering sex differences in APOE-ε4's association with AD biomarkers and tau pathology mechanisms in AD. Although this study provides robust evidence of complex interplay between sex and APOE-ε4 for European ancestry, further research is needed to fully understand other ancestry differences.
Collapse
Affiliation(s)
- Xiaoyi Xu
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, Missouri
- Division of Biostatistics, Washington University School of Medicine, St Louis, Missouri
| | - Jiseon Kwon
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, Missouri
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri
| | - Ruiqi Yan
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, Missouri
- Division of Biostatistics, Washington University School of Medicine, St Louis, Missouri
| | - Catherine Apio
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, Missouri
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, Korea
| | - Soomin Song
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, Missouri
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri
| | - Gyujin Heo
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, Missouri
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri
| | - Qijun Yang
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, Missouri
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio
| | - Jigyasha Timsina
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, Missouri
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri
| | - Menghan Liu
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, Missouri
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri
| | - John Budde
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, Missouri
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Alberto Lleó
- Sant Pau Memory Unit, Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Agustin Ruiz
- Research Center and Memory Clinic, ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
| | - José Luis Molinuevo
- BarcelonaBeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
| | - Virginia Man-Yee Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Yuetiva Deming
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison
| | - Amanda J. Heslegrave
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Tim J. Hohman
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Pau Pastor
- University Hospital Germans Trias i Pujol and The Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
| | - Elaine R. Peskind
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle
| | - Marilyn S. Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - John C. Morris
- Department of Neurology, Washington University, St Louis, Missouri
- Knight Alzheimer’s Disease Research Center, Washington University, St Louis, Missouri
| | - Taesung Park
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, Korea
- Department of Statistics, Seoul National University, Seoul, Korea
| | - Carlos Cruchaga
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, Missouri
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri
- Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, Missouri
| | - Yun Ju Sung
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, Missouri
- Division of Biostatistics, Washington University School of Medicine, St Louis, Missouri
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri
| |
Collapse
|
11
|
Ivarsson Orrelid C, Rosberg O, Weiner S, Johansson FD, Gobom J, Zetterberg H, Mwai N, Stempfle L. Applying machine learning to high-dimensional proteomics datasets for the identification of Alzheimer's disease biomarkers. Fluids Barriers CNS 2025; 22:23. [PMID: 40033432 DOI: 10.1186/s12987-025-00634-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 02/11/2025] [Indexed: 03/05/2025] Open
Abstract
PURPOSE This study explores the application of machine learning to high-dimensional proteomics datasets for identifying Alzheimer's disease (AD) biomarkers. AD, a neurodegenerative disorder affecting millions worldwide, necessitates early and accurate diagnosis for effective management. METHODS We leverage Tandem Mass Tag (TMT) proteomics data from the cerebrospinal fluid (CSF) samples from the frontal cortex of patients with idiopathic normal pressure hydrocephalus (iNPH), a condition often comorbid with AD, with rare access to both lumbar and ventricular samples. Our methodology includes extensive data preprocessing to address batch effects and missing values, followed by the use of the Synthetic Minority Over-sampling Technique (SMOTE) for data augmentation to overcome the small sample size. We apply linear, and non-linear machine learning models, and ensemble methods, to compare iNPH patients with and without biomarker evidence of AD pathology ( A β - T - or A β + T + ) in a classification task. RESULTS We present a machine learning workflow for working with high-dimensional TMT proteomics data that addresses their inherent data characteristics. Our results demonstrate that batch effect correction has no or minor impact on the models' performance and robust feature selection is critical for model stability and performance, especially in the high-dimensional proteomics data setting for AD diagnostics. The results further indicated that removing features with missing values produced stronger models than imputing them, and the batch effect had minimal impact on the models Our best-performing disease-progression detection model, a random forest, achieves an AUC of 0.84 (± 0.03). CONCLUSION We identify several novel protein biomarkers candidates, such as FABP3 and GOT1, with potential diagnostic value for AD pathology detection, suggesting the necessity of different biomarkers for AD diagnoses for patients with iNPH, and considering different biomarkers for ventricular and lumbar CSF samples. This work underscores the importance of a meticulous machine learning process in enhancing biomarker discovery. Our study also provides insights in translating biomarkers from other central nervous system diseases like iNPH, and both ventricular and lumbar CSF samples for biomarker discovery, providing a foundation for future research and clinical applications.
Collapse
Affiliation(s)
- Christoffer Ivarsson Orrelid
- Computer Science and Engineering, Chalmers University of Technology and University of Gothenburg, Rännvägen 6b, 41296, Gothenburg, Västra Götalandsregionen, Sweden.
| | - Oscar Rosberg
- Computer Science and Engineering, Chalmers University of Technology and University of Gothenburg, Rännvägen 6b, 41296, Gothenburg, Västra Götalandsregionen, Sweden
| | - Sophia Weiner
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Wallinsgatan 6, 43141, Möndal, Västra Götalandsregionen, Sweden
| | - Fredrik D Johansson
- Computer Science and Engineering, Chalmers University of Technology and University of Gothenburg, Rännvägen 6b, 41296, Gothenburg, Västra Götalandsregionen, Sweden
| | - Johan Gobom
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Wallinsgatan 6, 43141, Möndal, Västra Götalandsregionen, Sweden
- Clinical Neurochemistry Lab, Clinical Neurochemistry Lab, Institute of Neuroscience and Physiology, Sahlgrenska University Hospital, Wallinsgatan 6, 43141, Möndal, Västra Götalandsregionen, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Wallinsgatan 6, 43141, Möndal, Västra Götalandsregionen, Sweden
- Clinical Neurochemistry Lab, Clinical Neurochemistry Lab, Institute of Neuroscience and Physiology, Sahlgrenska University Hospital, Wallinsgatan 6, 43141, Möndal, Västra Götalandsregionen, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, UCL Institute of Neurology, Queen Square, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Newton Mwai
- Computer Science and Engineering, Chalmers University of Technology and University of Gothenburg, Rännvägen 6b, 41296, Gothenburg, Västra Götalandsregionen, Sweden
| | - Lena Stempfle
- Computer Science and Engineering, Chalmers University of Technology and University of Gothenburg, Rännvägen 6b, 41296, Gothenburg, Västra Götalandsregionen, Sweden
| |
Collapse
|
12
|
Rehman H, Ang TFA, Tao Q, Au R, Farrer LA, Qiu WQ, Zhang X. Plasma protein risk scores for mild cognitive impairment and Alzheimer's disease in the Framingham heart study. Alzheimers Dement 2025; 21:e70066. [PMID: 40156298 PMCID: PMC11953566 DOI: 10.1002/alz.70066] [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: 08/09/2024] [Revised: 02/03/2025] [Accepted: 02/08/2025] [Indexed: 04/01/2025]
Abstract
INTRODUCTION It is unclear whether aggregated plasma protein risk scores (PPRSs) could be useful in predicting the risks of mild cognitive impairment (MCI) and Alzheimer's disease (AD). METHODS The Cox proportional hazard model with the Least Absolute Shrinkage and Selection Operator penalty was used to build the PPRSs for MCI and AD in 1515 Framingham Heart Study Generation 2 with 1128 proteins measured in plasma at exam 5 (cognitively normal [CN] = 1258, MCI = 129, AD = 128). RESULTS MCI PPRS had a hazard ratio (HR) of 6.97 [5.34, 9.12], with a discriminating power (C-index = 82.52%). AD PPRS had a HR of 5.74 [4.67, 7.05] (C-index = 88.15%). Both PPRSs were also significantly associated with cognitive changes, brain atrophy, and plasma AD biomarkers. Proteins in the MCI and AD PPRSs were involved in several pathways related to leukocyte, chemotaxis, immunity, inflammation, and cellular migration. DISCUSSION This study suggests that PPRSs serve well to predict the risk of developing MCI and AD as well as cognitive changes and AD-related pathogenesis in the brain. HIGHLIGHTS PPRSs were developed for the risk of AD and AD preclinical stage, MCI. PPRSs were developed for MCI and AD associated with cognitive changes, loss of brain volume, and increasing level of plasma AD biomarkers. Leukocyte, chemotaxis, immunity, inflammation, and cellular migration enriched in proteins were identified as being involved in MCI and AD PPRSs.
Collapse
Grants
- Alzheimer's Disease Neuroimaging Initiative
- U01 AG024904 NIH HHS
- W81XWH-12-2-0012 (DOD) ADNI
- National Institute on Aging (NIA)
- the National Institute of Biomedical Imaging and Bioengineering
- AbbVie
- Alzheimer's Association
- Alzheimer's Drug Discovery Foundation
- Araclon Biotech
- BioClinica, Inc.
- Biogen
- Bristol-Myers Squibb Company
- CereSpir, Inc.
- Cogstate
- Eisai Inc.
- Elan Pharmaceuticals, Inc.
- Eli Lilly and Co.
- EuroImmun
- F. Hoffmann-La Roche Ltd and its affiliated company Genentech, Inc.
- Fujirebio
- GE Healthcare
- IXICO Ltd.
- Janssen Alzheimer Immunotherapy Research & Development, LLC
- Johnson & Johnson Pharmaceutical Research & Development LLC
- Lumosity
- Merck & Co., Inc.
- Meso Scale Diagnostics, LLC
- NeuroRx Research
- Neurotrack Technologies
- Novartis Pharmaceuticals Corp.
- Pfizer Inc.
- Piramal Imaging
- Servier
- Takeda Pharmaceutical Company
- Transition Therapeutics
- CIHR
- N01-HC-25195 Framingham Heart Study's National Heart, Lung, and Blood Institute
- U19-AG068753 NIA NIH HHS
- RF1AG075832-01A1 NIA NIH HHS
- U01-AG072577 NIA NIH HHS
- R01-AG080810 NIA NIH HHS
- U19-AG068753 Framingham Heart Study Brain Aging Program (FHS-BAP) pilot
- NSF DMS/NIGMS-2347698 National Science Foundation
- Alzheimer's Disease Neuroimaging Initiative
- National Institutes of Health
- AbbVie
- Alzheimer's Association
- Alzheimer's Drug Discovery Foundation
- BioClinica, Inc.
- Biogen
- Bristol‐Myers Squibb Company
- Fujirebio
- GE Healthcare
- Merck & Co., Inc.
- Pfizer Inc.
- Servier
- Takeda Pharmaceutical Company
- Canadian Institutes of Health Research
- NIA
- National Science Foundation
Collapse
Affiliation(s)
- Habbiburr Rehman
- Departments of Medicine (Biomedical Genetics)Boston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Ting Fang Alvin Ang
- Departments of Anatomy & NeurobiologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Departments of NeurologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Qiushan Tao
- Departments of Pharmacology & Experimental TherapeuticsBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Rhoda Au
- Departments of Medicine (Biomedical Genetics)Boston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Departments of Anatomy & NeurobiologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Departments of NeurologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Departments of EpidemiologyBoston University School of Public HealthBostonMassachusettsUSA
- Framingham Heart StudyBoston University School of MedicineFraminghamMassachusettsUSA
- Alzheimer's Disease Research CenterBoston University School of MedicineBostonMassachusettsUSA
| | - Lindsay A. Farrer
- Departments of Medicine (Biomedical Genetics)Boston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Departments of NeurologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Departments of EpidemiologyBoston University School of Public HealthBostonMassachusettsUSA
- Framingham Heart StudyBoston University School of MedicineFraminghamMassachusettsUSA
- Alzheimer's Disease Research CenterBoston University School of MedicineBostonMassachusettsUSA
- Departments of OphthalmologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Departments of BiostatisticsBoston University School of Public HealthBostonMassachusettsUSA
| | - Wei Qiao Qiu
- Departments of Pharmacology & Experimental TherapeuticsBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Alzheimer's Disease Research CenterBoston University School of MedicineBostonMassachusettsUSA
- Departments of PsychiatryBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Xiaoling Zhang
- Departments of Medicine (Biomedical Genetics)Boston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Framingham Heart StudyBoston University School of MedicineFraminghamMassachusettsUSA
- Departments of BiostatisticsBoston University School of Public HealthBostonMassachusettsUSA
| | | |
Collapse
|
13
|
Baek S, Lee JC, Byun BH, Park SY, Ha JH, Lee KC, Yang SH, Lee JS, Hong S, Han G, Lim SM, Kim Y, Kim HY. Combination of Aβ40, Aβ42, and Tau Plasma Levels to Distinguish Amyloid-PET Positive Alzheimer Patients from Normal Controls. Exp Neurobiol 2025; 34:1-8. [PMID: 40091634 PMCID: PMC11919640 DOI: 10.5607/en25008] [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: 02/20/2025] [Revised: 02/28/2025] [Accepted: 02/28/2025] [Indexed: 03/19/2025] Open
Abstract
Alzheimer disease (AD) diagnosis is confirmed using a medley of modalities, such as the detection of amyloid-β (Aβ) neuritic plaques and neurofibrillary tangles with positron electron tomography (PET) or the appraisal of irregularities in cognitive function with examinations. Although these methods have been efficient in confirming AD pathology, the rising demand for earlier intervention during pathogenesis has led researchers to explore the diagnostic potential of fluid biomarkers in cerebrospinal fluid (CSF) and plasma. Since CSF sample collection is invasive and limited in quantity, biomarker detection in plasma has become more attractive and modern advancements in technology has permitted more efficient and accurate analysis of plasma biomolecules. In this study, we found that a composite of standard factors, Aβ40 and total tau levels in plasma, divided by the variation factor, plasma Aβ42 level, provide better correlation with amyloid neuroimaging and neuropsychological test results than a level comparison between total tau and Aβ42 in plasma. We collected EDTA-treated blood plasma samples of 53 subjects, of randomly selected 27 AD patients and 26 normal cognition (NC) individuals, who received amyloid-PET scans for plaque quantification, and measured plasma levels of Aβ40, Aβ42, and total tau with digital enzyme-linked immunosorbent assay (ELISA) in a blinded manner. There was difficulty distinguishing AD patients from controls when analyzing biomarkers independently. However, significant differentiation was observed between the two groups when comparing individual ratios of total-tau×Aβ40/Aβ42. Our results indicate that collectively comparing fluctuations of these fluid biomarkers could aid in monitoring AD pathogenesis.
Collapse
Affiliation(s)
- Seungyeop Baek
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon 21983, Korea
| | - Jinny Claire Lee
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon 21983, Korea
| | - Byung Hyun Byun
- Department of Nuclear Medicine, Korea Cancer Center Hospital, Seoul 01812, Korea
| | - Su Yeon Park
- Department of Neurology, Korea Cancer Center Hospital, Seoul 01812, Korea
| | - Jeong Ho Ha
- Department of Neurology, Korea Cancer Center Hospital, Seoul 01812, Korea
| | - Kyo Chul Lee
- Division of RI-Convergence Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea
| | - Seung-Hoon Yang
- Department of Medical Biotechnology, Dongguk University, Seoul 04620, Korea
| | - Jun-Seok Lee
- Department of Pharmacology, Korea University, Seoul 02841, Korea
| | - Seungpyo Hong
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon 21983, Korea
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano), School of Pharmacy, University of Wisconsin, Madison, WI 53705, USA
| | - Gyoonhee Han
- Department of Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Sang Moo Lim
- Department of Nuclear Medicine, Korea Cancer Center Hospital, Seoul 01812, Korea
| | - YoungSoo Kim
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon 21983, Korea
- Amyloid Solution Inc., Seongnam 13486, Korea
| | - Hye Yun Kim
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon 21983, Korea
| |
Collapse
|
14
|
Niknam N, Khaefi S, Heidarpour H, Sadeghi M, Jafari NA, Mohammadi S, Ahmadi Z, Ahangar-Sirous R, Mayeli M, Seyedmirzaei H. Associations between diffusion tensor imaging patterns and cerebrospinal fluid markers in mild cognitive impairment. J Clin Neurosci 2025; 135:111141. [PMID: 40010169 DOI: 10.1016/j.jocn.2025.111141] [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: 11/17/2024] [Revised: 02/17/2025] [Accepted: 02/17/2025] [Indexed: 02/28/2025]
Abstract
Diffusion tensor imaging (DTI) can be used to detect early signs of increased water diffusivity in white matter tracts in patients with mild cognitive impairment (MCI). This study examined how DTI, alongside cerebrospinal fluid (CSF) biomarkers (like tau proteins and amyloid-β), can help identify early brain changes in MCI. We included 159 individuals (92 with MCI and 67 healthy controls) from the Alzheimer's Disease Neuroimaging Initiative (ADNI) and extracted their demographics, CSF biomarkers, and DTI metrics. We compared the biomarkers (CSF biomarkers and DTI markers in 57 white matter tracts) between the two study groups using a general linear model, adjusting for age, sex, and handedness. CSF biomarker levels showed a statistically significant difference between the two study groups. Also, diffusion properties of left Cingulum and left Uncinate fasciculus in both groups were statistically different. Additionally, we explored possible associations between CSF and DTI markers in the MCI group. Our results indicated several statistically significant associations between DTI metrics and CSF biomarkers within specific white matter tracts. These findings underscore the complexity of imaging and molecular markers associated with MCI.
Collapse
Affiliation(s)
| | - Sara Khaefi
- Department of Electrical Engineering, Shahed University, Tehran, Iran
| | - Hadise Heidarpour
- Neuroscience Research Center, Golestan University of Medical Sciences, Gorgan, Iran; Gastroenterology and Hepatology Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mohammad Sadeghi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; School of Rehabilitation, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Sheida Mohammadi
- Department of Psychology, South Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Zeinab Ahmadi
- Neuroscience Graduate Program, Medical School, University of Crete, Greece
| | - Ramin Ahangar-Sirous
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahsa Mayeli
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Department of Radiology and Biomedical Imaging, Yale School of Medicine, CT, USA
| | - Homa Seyedmirzaei
- Occupational Sleep Research Center, Baharloo Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
Delaby C, Alcolea D, Busto G, Gabelle A, Ayrignac X, Bennys K, Muiño E, Villatoro P, Fernández-Cadenas I, Pradeilles N, Bounasri SE, Torres S, Hirtz C, Zetterberg H, Lleó A, Fortea J, Lehmann S. Plasma Hepcidin as a potential informative biomarker of Alzheimer disease and vascular dementia. Alzheimers Res Ther 2025; 17:42. [PMID: 39948603 PMCID: PMC11823057 DOI: 10.1186/s13195-025-01696-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/07/2025] [Indexed: 02/16/2025]
Abstract
BACKGROUND Blood-based assays are expected to be integrated into clinical routines across various contexts, including Alzheimer's disease (AD). Vascular dementia (VaD), which is the second most common cause leading to dementia after AD, could also significantly benefit from this advancement. However, no informative fluid biomarker has been identified for VaD. Given the disruption of iron homeostasis in both AD and VaD, this study aims to characterize the potential of the iron-related hormone Hepcidin as a biomarker for these two conditions. We will compare its added value to established AT(N) blood biomarkers. METHODS Blood biomarkers (amyloid-composite, p-Tau181, Neurofilament Light Chain [NfL] and Hepcidin) levels in blood were analyzed in two independent cohorts and compared between AD patients and non-AD individuals. Additionally, blood Hepcidin and NfL were evaluated in the contexts of VaD and CADASIL, with their relative diagnostic value assessed. RESULTS Blood Hepcidin and NfL do not significantly increase the AUC obtained with both p-Tau181 and amyloid composite in the context of AD. In contrast, AUC for VaD diagnosis is significantly higher when combining these two blood biomarkers compared to NfL alone. Hepcidin was not significantly modified in CADASIL patients compared to control subjects. CONCLUSION Blood Hepcidin and NfL have limited interest in the clinical context of AD but determination of these biomarkers shows to be highly informative for the diagnosis of VaD. This result could have important implications for diagnosis and implementation of clinical trials.
Collapse
Affiliation(s)
- Constance Delaby
- Université de Montpellier, INM INSERM LBPC-PPC, IRMB CHU de Montpellier, Montpellier, France.
- Department of Neurology, Sant Pau Memory Unit, Hospital de La Santa Creu I Sant Pau - IIB Sant Pau, Barcelona, Spain.
| | - Daniel Alcolea
- Department of Neurology, Sant Pau Memory Unit, Hospital de La Santa Creu I Sant Pau - IIB Sant Pau, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, Ciberned, Spain
| | - Germain Busto
- Department of Neurology, Université de Montpellier, Inserm INM NeuroPEPs Team, 34000, Montpellier, France
| | - Audrey Gabelle
- Department of Neurology, Université de Montpellier, Inserm INM NeuroPEPs Team, 34000, Montpellier, France
| | - Xavier Ayrignac
- Department of Neurology, Université de Montpellier, Inserm INM NeuroPEPs Team, 34000, Montpellier, France
| | - Karim Bennys
- Department of Neurology, Université de Montpellier, Inserm INM NeuroPEPs Team, 34000, Montpellier, France
| | - Elena Muiño
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Farmacogenómica y Genética Neurovascular, Sant Quintí 77-79, Barcelona, 08041, Spain
- Unidad de Epilepsia, Hospital de la Santa Creu i Sant Pau, Sant Antoni Maria Claret 167, Spain, 08025, Barcelona
| | - Paula Villatoro
- Farmacogenómica y Genética Neurovascular. Institut d'Investigació Biomèdica Sant Pau (IIB, SANT PAU), Sant Quintí 77-79, 08041, Barcelona, Spain
| | - Israel Fernández-Cadenas
- Farmacogenómica y Genética Neurovascular. Institut d'Investigació Biomèdica Sant Pau (IIB, SANT PAU), Sant Quintí 77-79, 08041, Barcelona, Spain
| | - Nicolas Pradeilles
- Université de Montpellier, INM INSERM LBPC-PPC, IRMB CHU de Montpellier, Montpellier, France
| | - Shaima El Bounasri
- Department of Neurology, Sant Pau Memory Unit, Hospital de La Santa Creu I Sant Pau - IIB Sant Pau, Barcelona, Spain
| | - Soraya Torres
- Department of Neurology, Sant Pau Memory Unit, Hospital de La Santa Creu I Sant Pau - IIB Sant Pau, Barcelona, Spain
| | - Christophe Hirtz
- Université de Montpellier, INM INSERM LBPC-PPC, IRMB CHU de Montpellier, Montpellier, France
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the , Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- Dementia Research Institute at UCL, London, UK
- Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, University of Wisconsin-Madison, Madison, WI, USA
| | - Alberto Lleó
- Department of Neurology, Sant Pau Memory Unit, Hospital de La Santa Creu I Sant Pau - IIB Sant Pau, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, Ciberned, Spain
| | - Juan Fortea
- Department of Neurology, Sant Pau Memory Unit, Hospital de La Santa Creu I Sant Pau - IIB Sant Pau, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, Ciberned, Spain
| | - Sylvain Lehmann
- Université de Montpellier, INM INSERM LBPC-PPC, IRMB CHU de Montpellier, Montpellier, France
| |
Collapse
|
16
|
Yang M, Zhang X, Zhang D, Zhang Y, Wang J, Zhang Y, Gu C, Zhang X, Wei L. Body Fluid Biomarkers of Neurological Injury in HIV-1-Associated Neurocognitive Disorder. AIDS Res Hum Retroviruses 2025. [PMID: 39938886 DOI: 10.1089/aid.2024.0053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
Since combined antiretroviral therapy for human immunodeficiency virus-associated neurocognitive dysfunction (HAND) only slows the disease's progression, early identification and timely intervention are crucial for effective therapy. In this article, we review the latest evidence in body fluid biomarkers of HAND, providing an overview of research conducted on cerebrospinal fluid and blood samples to draw conclusions on promising biomarkers. Although the significance of biomarkers such as amyloid metabolites, tau proteins, neurofilament light chain, myelin oligodendrocyte glycoprotein, and brain-derived neurotrophic factor in the early detection of HAND may not be immediately clear, they could potentially play a crucial role in evaluating prognosis and tracking the effectiveness of treatment.
Collapse
Affiliation(s)
- Meijuan Yang
- Department of Clinical Laboratory, Gansu Provincial Hospital, Lanzhou, China
| | - Xiaomei Zhang
- Department of Clinical Laboratory, Gansu Provincial Hospital, Lanzhou, China
| | - Dong Zhang
- Department of Anesthesiology, Gansu Provincial Hospital, Lanzhou, China
| | - Yamin Zhang
- Department of Neurology, Gansu Provincial Hospital, Lanzhou, China
| | - Jiamei Wang
- Department of Clinical Laboratory, Gansu Provincial Hospital, Lanzhou, China
| | - Yi Zhang
- Department of Neurology, Gansu Provincial Hospital, Lanzhou, China
| | - Cheng Gu
- Department of Neurology, Gansu Provincial Hospital, Lanzhou, China
| | - Xingwang Zhang
- Department of Clinical Laboratory, Gansu Provincial Hospital, Lanzhou, China
| | - Lianhua Wei
- Department of Clinical Laboratory, Gansu Provincial Hospital, Lanzhou, China
| |
Collapse
|
17
|
Song J, Cho E, Lee H, Lee S, Kim S, Kim J. Development of Neurodegenerative Disease Diagnosis and Monitoring from Traditional to Digital Biomarkers. BIOSENSORS 2025; 15:102. [PMID: 39997004 PMCID: PMC11852611 DOI: 10.3390/bios15020102] [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: 12/31/2024] [Revised: 02/03/2025] [Accepted: 02/10/2025] [Indexed: 02/26/2025]
Abstract
Monitoring and assessing the progression of symptoms in neurodegenerative diseases, including Alzheimer's and Parkinson's disease, are critical for improving patient outcomes. Traditional biomarkers, such as cerebrospinal fluid analysis and brain imaging, are widely used to investigate the underlying mechanisms of disease and enable early diagnosis. In contrast, digital biomarkers derived from phenotypic changes-such as EEG, eye movement, gait, and speech analysis-offer a noninvasive and accessible alternative. Leveraging portable and widely available devices, such as smartphones and wearable sensors, digital biomarkers are emerging as a promising tool for ND diagnosis and monitoring. This review highlights the comprehensive developments in digital biomarkers, emphasizing their unique advantages and integration potential alongside traditional biomarkers.
Collapse
Affiliation(s)
| | | | | | | | | | - Jinsik Kim
- Department of Biomedical Engineering, College of Life Science and Biotechnology, Dongguk University, Seoul 04620, Republic of Korea; (J.S.); (E.C.); (H.L.); (S.L.); (S.K.)
| |
Collapse
|
18
|
Duan S, Cai T, Chen L, Wang X, Zhang S, Han B, Lim EG, Hoettges K, Hu Y, Song P. An integrated paper-based microfluidic platform for screening of early-stage Alzheimer's disease by detecting Aβ42. LAB ON A CHIP 2025; 25:512-523. [PMID: 39803675 DOI: 10.1039/d4lc00748d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2025]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia worldwide, and the development of early screening methods can address its significant health and social consequences. In this paper, we present a rotary-valve assisted paper-based immunoassay device (RAPID) for early screening of AD, featuring a highly integrated on-chip rotary micro-valve that enables fully automated and efficient detection of the AD biomarker (amyloid beta 42, Aβ42) in artificial plasma. The microfluidic paper-based analytical device (μPAD) of the RAPID pre-stores the required assay reagents on a μPAD and automatically controls the liquid flow through a single valve. Once the test sample is added, the test reagents are sequentially transferred to the test area in the order set by the enzyme-linked immunosorbent assay (ELISA) protocol. In addition, the RAPID can remotely control the operation of the μPAD valve via a micro-servomotor, quantify the signals generated, display the results, and wirelessly transmit the data to a smartphone. To calibrate the RAPID, we performed a sandwich ELISA for Aβ42 in artificial plasma, and obtained a low limit of detection (LOD) of 9.6 pg mL-1, a coefficient of determination (COD) of 0.994, and an individual assay time of ∼30 minutes. In addition, we simulated 24 artificial samples to quantify Aβ42 protein concentrations in artificial plasma samples. The results show good consistency between the conventional ELISA and RAPID detection. The experimental results demonstrate that the RAPID is expected to promote further popularization of the screening of early-stage AD.
Collapse
Affiliation(s)
- Sixuan Duan
- School of Advanced Technology, Xi'an Jiaotong-Liverpool University, Suzhou, 215000, China.
- Department of Electrical and Electronic Engineering, University of Liverpool, Liverpool, L69 7ZX, UK
| | - Tianyu Cai
- School of Advanced Technology, Xi'an Jiaotong-Liverpool University, Suzhou, 215000, China.
- Department of Electrical and Electronic Engineering, University of Liverpool, Liverpool, L69 7ZX, UK
| | - Lizhe Chen
- School of Advanced Technology, Xi'an Jiaotong-Liverpool University, Suzhou, 215000, China.
- Department of Electrical and Electronic Engineering, University of Liverpool, Liverpool, L69 7ZX, UK
| | - Xiaoyan Wang
- School of Advanced Technology, Xi'an Jiaotong-Liverpool University, Suzhou, 215000, China.
- Department of Electrical and Electronic Engineering, University of Liverpool, Liverpool, L69 7ZX, UK
| | - Shuailong Zhang
- School of Integrated Circuits and Electronics, Beijing Institute of Technology, Beijing, 100081, China
| | - Bing Han
- School of Advanced Technology, Xi'an Jiaotong-Liverpool University, Suzhou, 215000, China.
- Department of Electrical and Electronic Engineering, University of Liverpool, Liverpool, L69 7ZX, UK
| | - Eng Gee Lim
- School of Advanced Technology, Xi'an Jiaotong-Liverpool University, Suzhou, 215000, China.
- Department of Electrical and Electronic Engineering, University of Liverpool, Liverpool, L69 7ZX, UK
| | - Kai Hoettges
- Department of Electrical and Electronic Engineering, University of Liverpool, Liverpool, L69 7ZX, UK
| | - Yong Hu
- School of Mechanical and Aerospace Engineering, Jilin University, Changchun, 130022, China
| | - Pengfei Song
- School of Advanced Technology, Xi'an Jiaotong-Liverpool University, Suzhou, 215000, China.
- Department of Electrical and Electronic Engineering, University of Liverpool, Liverpool, L69 7ZX, UK
| |
Collapse
|
19
|
Andersson K, Akel S, Asztély F, Larsson D, Zetterberg H, Zelano J. Higher plasma total tau concentrations among patients reporting CNS-related side effects from antiseizure medication. Seizure 2025; 125:99-105. [PMID: 39826304 DOI: 10.1016/j.seizure.2025.01.015] [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: 07/31/2024] [Revised: 12/12/2024] [Accepted: 01/11/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Side effects from antiseizure medication (ASM) are common in epilepsy but biomarkers for detection and monitoring are missing. This study investigated associations between CNS-related side effects from ASM and blood concentrations of the brain injury markers neurofilament-light (NFL), total tau, glial acidic fibrillary protein (GFAP), S100 calcium-binding protein B (S100B) and neuron-specific enolase (NSE). METHODS This is a population-based cohort study of adults with epilepsy recruited from five Swedish outpatient neurology clinics from December 2020 to April 2023. Side effects classified as CNS-related: tiredness, dizziness, headache, concentration, memory, mood, motor/tremor, or sleep. Marker concentrations in the groups CNS side effects/no side effects were analyzed with Mann-Whitney U-test and significant differences were included in multivariable logistic regression models adjusting for age, epilepsy duration, seizure status, acquired structural lesion, and mono-/polytherapy. RESULTS The cohort consisted of 367 patients, 187 (51 %) were females, the median age was 43 years (IQR 30-61), and 123 (34 %) reported CNS side effects. Total tau was higher among participants reporting CNS side effects (median 4.44 (95 %CI 4.12-4.88) pg/ml) compared with participants without side effects (3.84 (95 %CI 3.52-4.07) pg/ml, p < 0.01). The difference remained significant in multivariable regression models. NSE was higher among participants without side effects but did not remain significant in the multivariable regression model. No differences were observed for NFL, GFAP or S100B. CONCLUSIONS Higher total tau plasma concentration could be associated with increased risk of CNS side effects from ASM. Longitudinal studies could determine if this reflects vulnerability or detrimental effects of ASM. TRIAL REGISTRATION PREDICT, clinicaltrials.gov identifier NCT04559919.
Collapse
Affiliation(s)
- Klara Andersson
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden; Department of Neurology, Sahlgrenska University Hospital, member of ERN Epicare, Gothenburg, Sweden; Wallenberg Center of Molecular and Translational Medicine, Gothenburg University, Sweden.
| | - Sarah Akel
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden; Wallenberg Center of Molecular and Translational Medicine, Gothenburg University, Sweden
| | - Fredrik Asztély
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden; Department of Neurology, Angered Hospital, Sweden
| | - David Larsson
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden; Department of Neurology, Sahlgrenska University Hospital, member of ERN Epicare, Gothenburg, Sweden; Wallenberg Center of Molecular and Translational Medicine, Gothenburg University, Sweden
| | - Henrik Zetterberg
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK; UK Dementia Research Institute at UCL, London, UK
| | - Johan Zelano
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden; Department of Neurology, Sahlgrenska University Hospital, member of ERN Epicare, Gothenburg, Sweden; Wallenberg Center of Molecular and Translational Medicine, Gothenburg University, Sweden
| |
Collapse
|
20
|
Abdelhamed HG, Hassan AA, Sakraan AA, Al-Deeb RT, Mousa DM, Aboul Ezz HS, Noor NA, Khadrawy YA, Radwan NM. Brain interleukins and Alzheimer's disease. Metab Brain Dis 2025; 40:116. [PMID: 39891777 PMCID: PMC11787210 DOI: 10.1007/s11011-025-01538-5] [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: 10/15/2024] [Accepted: 01/10/2025] [Indexed: 02/03/2025]
Abstract
The central nervous system (CNS) is immune-privileged by several immuno-modulators as interleukins (ILs). ILs are cytokines secreted by immune cells for cell-cell signaling communications and affect the functions of the CNS. ILs were reported to orchestrate different molecular and cellular mechanisms of both physiological and pathological events, through overproduction or over-expression of their receptors. They interact with numerous receptors mediating pro-inflammatory and/or anti-inflammatory actions. Interleukins have been implicated to participate in neurodegenerative diseases. They play a critical role in Alzheimer's disease (AD) pathology which is characterized by the over-production of pro-inflammatory ILs. These may aggravate neurodegeneration, in addition to their contribution to detrimental mechanisms as oxidative stress, and excitotoxicity. However, recent research on the relation between ILs and AD revealed major discrepancies. Most of the major ILs were shown to play both pro- and anti-inflammatory roles in different experimental settings and models. The interactions between different ILs through shared pathways also add to the difficulty of drawing solid conclusions. In addition, targeting the different ILs has not yielded consistent results. The repeated failures of therapeutic drugs in treating AD necessitate the search for novel agents targeting multiple mechanisms of the disease pathology. In this context, the understanding of interleukins and their roles throughout the disease progression and interaction with other systems in the brain may provide promising therapeutic targets for the prevention or treatment of AD.
Collapse
Affiliation(s)
- Heba G Abdelhamed
- Department of Zoology and Chemistry, Faculty of Science, Cairo University, Giza, Egypt
| | - Arwa A Hassan
- Faculty of Pharmacy & Pharmaceutical Industries, Sinai University, Sinai, Egypt
| | - Alaa A Sakraan
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt
| | | | - Dalia M Mousa
- Department of Biotechnology, Faculty of Science, Cairo University, Giza, Egypt
| | - Heba S Aboul Ezz
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt.
| | - Neveen A Noor
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt
| | - Yasser A Khadrawy
- Medical Physiology Department, Medical Research and Clinical Studies Institute, National Research Center, Giza, Egypt
| | - Nasr M Radwan
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt
| |
Collapse
|
21
|
Philippi SM, BP K, Raj T, Castellano JM. APOE genotype and brain amyloid are associated with changes in the plasma proteome in elderly subjects without dementia. Ann Clin Transl Neurol 2025; 12:366-382. [PMID: 39689057 PMCID: PMC11822792 DOI: 10.1002/acn3.52250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 10/03/2024] [Accepted: 10/24/2024] [Indexed: 12/19/2024] Open
Abstract
OBJECTIVE Recent work has bolstered the possibility that peripheral changes may be relevant to Alzheimer's disease pathogenesis in the brain. While age-associated blood-borne proteins have been targeted to restore function to the aged brain, it remains unclear whether other dysfunctional systemic states can be exploited for similar benefits. Here, we investigate whether APOE allelic variation or presence of brain amyloid are associated with plasma proteomic changes and the molecular processes associated with these changes. METHODS Using the SOMAscan assay, we measured 1305 plasma proteins from 53 homozygous, APOE3 and APOE4 subjects without dementia. We investigated the relationship of either the APOE-ε4 allele or amyloid positivity with plasma proteome changes by linear mixed effects modeling and ontology-based pathway and module-trait correlation analyses. RESULTS APOE4 is associated with plasma protein differences linked to atherosclerosis, tyrosine kinase activity, cholesterol transport, extracellular matrix, and synaptogenesis pathways. Independent of APOE4, we found that subjects likely harboring brain amyloid exhibit plasma proteome signatures associated with AD-linked pathways, including neurovascular dysfunction. INTERPRETATION Our results indicate that APOE4 status or presence of brain amyloid are associated with plasma proteomic shifts prior to the onset of symptoms, suggesting that systemic pathways in certain risk contexts may be plausible targets for disease modification.
Collapse
Affiliation(s)
- Sarah M. Philippi
- Nash Family Department of Neuroscience, Department of Neurology, Friedman Brain InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Ronald M. Loeb Center for Alzheimer's DiseaseIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Graduate School of Biomedical SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Black Family Stem Cell InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Kailash BP
- Nash Family Department of Neuroscience, Department of Neurology, Friedman Brain InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Ronald M. Loeb Center for Alzheimer's DiseaseIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Graduate School of Biomedical SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Genetics and Genomic Sciences, Icahn Institute for Data Science and Genomic TechnologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Towfique Raj
- Nash Family Department of Neuroscience, Department of Neurology, Friedman Brain InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Ronald M. Loeb Center for Alzheimer's DiseaseIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Genetics and Genomic Sciences, Icahn Institute for Data Science and Genomic TechnologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Joseph M. Castellano
- Nash Family Department of Neuroscience, Department of Neurology, Friedman Brain InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Ronald M. Loeb Center for Alzheimer's DiseaseIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Black Family Stem Cell InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| |
Collapse
|
22
|
Wang M, Hua Y, Bai Y. A review of the application of exercise intervention on improving cognition in patients with Alzheimer's disease: mechanisms and clinical studies. Rev Neurosci 2025; 36:1-25. [PMID: 39029521 DOI: 10.1515/revneuro-2024-0046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 07/08/2024] [Indexed: 07/21/2024]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, leading to sustained cognitive decline. An increasing number of studies suggest that exercise is an effective strategy to promote the improvement of cognition in AD. Mechanisms of the benefits of exercise intervention on cognitive function may include modulation of vascular factors by affecting cardiovascular risk factors, regulating cardiorespiratory health, and enhancing cerebral blood flow. Exercise also promotes neurogenesis by stimulating neurotrophic factors, affecting neuroplasticity in the brain. Additionally, regular exercise improves the neuropathological characteristics of AD by improving mitochondrial function, and the brain redox status. More and more attention has been paid to the effect of Aβ and tau pathology as well as sleep disorders on cognitive function in persons diagnosed with AD. Besides, there are various forms of exercise intervention in cognitive improvement in patients with AD, including aerobic exercise, resistance exercise, and multi-component exercise. Consequently, the purpose of this review is to summarize the findings of the mechanisms of exercise intervention on cognitive function in patients with AD, and also discuss the application of different exercise interventions in cognitive impairment in AD to provide a theoretical basis and reference for the selection of exercise intervention in cognitive rehabilitation in AD.
Collapse
Affiliation(s)
- Man Wang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, No. 12 Middle Wulumuqi Road, Jing'an District, Shanghai 200040, China
- Department of Rehabilitation Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yan Hua
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, No. 12 Middle Wulumuqi Road, Jing'an District, Shanghai 200040, China
| | - Yulong Bai
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, No. 12 Middle Wulumuqi Road, Jing'an District, Shanghai 200040, China
| |
Collapse
|
23
|
Sun X, Zhu J, Li R, Peng Y, Gong L. The global research of magnetic resonance imaging in Alzheimer's disease: a bibliometric analysis from 2004 to 2023. Front Neurol 2025; 15:1510522. [PMID: 39882364 PMCID: PMC11774745 DOI: 10.3389/fneur.2024.1510522] [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: 10/13/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025] Open
Abstract
Background Alzheimer's disease (AD) is a common neurodegenerative disorder worldwide and the using of magnetic resonance imaging (MRI) in the management of AD is increasing. The present study aims to summarize MRI in AD researches via bibliometric analysis and predict future research hotspots. Methods We searched for records related to MRI studies in AD patients from 2004 to 2023 in the Web of Science Core Collection (WoSCC) database. CiteSpace was applied to analyze institutions, references and keywords. VOSviewer was used for the analysis of countries, authors and journals. Results A total of 13,659 articles were obtained in this study. The number of published articles showed overall exponential growth from 2004 to 2023. The top country and institution were the United States and the University of California System, accounting for 40.30% and 9.88% of the total studies, respectively. Jack CR from the United States was the most productive author. The most productive journal was the Journal of Alzheimers Disease. Keyword burst analysis revealed that "machine learning" and "deep learning" were the keywords that frequently appeared in the past 6 years. Timeline views of the references revealed that "#0 tau pathology" and "#1 deep learning" are currently the latest research focuses. Conclusion This study provides an in-depth overview of publications on MRI studies in AD. The United States is the leading country in this field with a concentration of highly productive researchers and high-level institutions. The current research hotspot is deep learning, which is being applied to develop noninvasive diagnosis and safer treatment of AD.
Collapse
Affiliation(s)
- Xiaoyu Sun
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Intelligent Medical Imaging, Nanchang, China
| | - Jianghua Zhu
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Intelligent Medical Imaging, Nanchang, China
| | - Ruowei Li
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Intelligent Medical Imaging, Nanchang, China
| | - Yun Peng
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Intelligent Medical Imaging, Nanchang, China
| | - Lianggeng Gong
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Intelligent Medical Imaging, Nanchang, China
| |
Collapse
|
24
|
Jiang H, Qian Y, Zhang L, Jiang T, Tai Y. ReIU: an efficient preliminary framework for Alzheimer patients based on multi-model data. Front Public Health 2025; 12:1449798. [PMID: 39830185 PMCID: PMC11739287 DOI: 10.3389/fpubh.2024.1449798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025] Open
Abstract
The rising incidence of Alzheimer's disease (AD) poses significant challenges to traditional diagnostic methods, which primarily rely on neuropsychological assessments and brain MRIs. The advent of deep learning in medical diagnosis opens new possibilities for early AD detection. In this study, we introduce retinal vessel segmentation methods based on U-Net ad iterative registration Learning (ReIU), which extract retinal vessel maps from OCT angiography (OCT-A) facilities. Our method achieved segmentation accuracies of 79.1% on the DRIVE dataset, 68.3% on the HRF dataset. Utilizing a multimodal dataset comprising both healthy and AD subjects, ReIU extracted vascular density from fundus images, facilitating primary AD screening with a classification accuracy of 79%. These results demonstrate ReIU's substantial accuracy and its potential as an economical, non-invasive screening tool for Alzheimer's disease. This study underscores the importance of integrating multi-modal data and deep learning techniques in advancing the early detection and management of Alzheimer's disease.
Collapse
Affiliation(s)
- Hao Jiang
- Engineering Research Center of Photoelectric Detection and Perception Technology, Yunnan Normal University, Kunming, China
- Yunnan Key Laboratory of Optoelectronic Information Technology, Kunming, China
| | - Yishan Qian
- Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China
- Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Research Center of Ophthalmology and Optometry, Shanghai, China
| | - Liqiang Zhang
- Engineering Research Center of Photoelectric Detection and Perception Technology, Yunnan Normal University, Kunming, China
- Yunnan Key Laboratory of Optoelectronic Information Technology, Kunming, China
| | - Tao Jiang
- Engineering Research Center of Photoelectric Detection and Perception Technology, Yunnan Normal University, Kunming, China
- Yunnan Key Laboratory of Optoelectronic Information Technology, Kunming, China
| | - Yonghang Tai
- Engineering Research Center of Photoelectric Detection and Perception Technology, Yunnan Normal University, Kunming, China
- Yunnan Key Laboratory of Optoelectronic Information Technology, Kunming, China
| |
Collapse
|
25
|
Waury K. Decision Tree for Protein Biomarker Selection for Clinical Applications. Methods Mol Biol 2025; 2884:355-368. [PMID: 39716013 DOI: 10.1007/978-1-0716-4298-6_21] [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: 12/25/2024]
Abstract
Discovery of novel protein biomarkers for clinical applications is an active research field across a manifold of diseases. Despite some successes and progress, the biomarker development pipeline still frequently ends in failure. Biomarker candidates that are discovered by appropriate technologies such as unbiased mass spectrometry cannot be validated or translated to immunoassays in many cases. Selection of strong disease biomarker candidates that further constitute suitable targets for antibody binding in immunoassays is thus important to allow routine clinical use. This essential selection step can be supported and rationalized using bioinformatics tools such as protein databases. Here, we present a workflow in the form of decision trees to computationally investigate biomarker candidates and their available affinity reagents in depth. This analysis can identify the most promising biomarker candidates for assay development, while minimal time and effort are required.
Collapse
Affiliation(s)
- Katharina Waury
- Department of Computer Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
26
|
Murley AG, Bowns L, Camacho M, Williams‐Gray CH, Tsvetanov KA, Rittman T, Barker RA, O'Brien JT, Rowe JB. Caregiver perspectives enable accurate diagnosis of neurodegenerative disease. Alzheimers Dement 2025; 21:e14377. [PMID: 39559925 PMCID: PMC11772714 DOI: 10.1002/alz.14377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/07/2024] [Accepted: 10/10/2024] [Indexed: 11/20/2024]
Abstract
BACKGROUND The history from a relative or caregiver is an important tool for differentiating neurodegenerative disease. We characterized patterns of caregiver questionnaire responses, at diagnosis and follow-up, on the Cambridge Behavioural Inventory (CBI). METHODS Data-driven multivariate analysis (n = 4952 questionnaires) was undertaken for participants (n = 2481) with Alzheimer's disease (typical/amnestic n = 543, language n = 50, and posterior cortical n = 50 presentations), Parkinson's disease (n = 740), dementia with Lewy bodies (n = 55), multiple system atrophy (n = 55), progressive supranuclear palsy (n = 422), corticobasal syndrome (n = 176), behavioral variant frontotemporal dementia (n = 218), semantic (n = 125) and non-fluent variant progressive aphasia (n = 88), and motor neuron disease (n = 12). RESULTS Item-level support vector machine learning gave high diagnostic accuracy between diseases (area under the curve mean 0.83), despite transdiagnostic changes in memory, behavior, and everyday function. There was progression in CBI subscores over time, which varied by diagnosis. DISCUSSION Our results highlight the differential diagnostic information for a wide range of neurodegenerative diseases contained in a simple, structured collateral history. HIGHLIGHTS We analyzed 4952 questionnaires from caregivers of 2481 participants with neurodegenerative disease. Behavioral and neuropsychiatric manifestations of neurodegenerative disease had overlapping diagnostic boundaries. Simple questionnaire response patterns were sufficient for accurate diagnosis of each disease. We reinforce the value of a collateral history to support a diagnosis of dementia. The Cambridge Behavioural Inventory is sensitive to change over time and suitable as an outcome measure in clinical trials.
Collapse
Affiliation(s)
- Alexander G. Murley
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Cambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Lucy Bowns
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | - Marta Camacho
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | - Caroline H. Williams‐Gray
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Cambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Kamen A. Tsvetanov
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Department of PsychologyUniversity of CambridgeCambridgeUK
| | - Timothy Rittman
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Cambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Roger A. Barker
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Cambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - John T. O'Brien
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Cambridge University Hospitals NHS Foundation TrustCambridgeUK
- Department of PsychiatryUniversity of CambridgeCambridgeUK
| | - James B. Rowe
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Cambridge University Hospitals NHS Foundation TrustCambridgeUK
- MRC Cognition and Brain Sciences UnitUniversity of CambridgeCambridgeUK
| |
Collapse
|
27
|
Aydoğan C, Çakan BB, Ali A. A Review on the Analysis of Chiral Molecules as Disease Biomarkers by LC/MS. Biomed Chromatogr 2025; 39:e6044. [PMID: 39605290 DOI: 10.1002/bmc.6044] [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: 08/10/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024]
Abstract
The chiral compounds may be biomarker candidates in human metabolism, which indicates the health status of humans. There are many applications in LC/MS that show that chiral small molecules are promising biomarkers for human diseases. Both clinical and commercial analyses of chiral metabolites are necessary due to the enantiomeric ratios of chiral molecules in biological samples may show both human health status and diseases. This review provides current and advanced LC/MS techniques for the separation and analysis of chiral molecules as disease biomarkers. In particular, sample preparation and chromatographic analysis of potential chiral biomarkers in biological samples are presented. The preparation and applications of several chiral columns used in enantiomeric separation of chiral metabolites/biomarkers by advanced LC/MS techniques are discussed. The improvement of these analyses will enable both the discovery of new chiral biomarkers and the prognosis of human diseases.
Collapse
Affiliation(s)
- Cemil Aydoğan
- Food Analysis and Research Laboratory, Bingol University, Bingöl, Türkiye
- Department of Food Engineering, Bingol University, Bingöl, Türkiye
- Department of Chemistry, Bingol University, Bingöl, Türkiye
| | | | - Ashraf Ali
- School of Chemistry and Chemical Engineering, Henan University of Technology, Zhengzhou, China
| |
Collapse
|
28
|
Jamerlan AM, An SSA, Hulme JP. Current status of fluid biomarkers for early Alzheimer's disease and FDA regulation implications. J Neurol Sci 2024; 467:123325. [PMID: 39615439 DOI: 10.1016/j.jns.2024.123325] [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] [Revised: 11/08/2024] [Accepted: 11/19/2024] [Indexed: 12/14/2024]
Abstract
Many changes can now be seen in the development and use of tests, especially those incorporating fluid biomarkers, to diagnose Alzheimer's disease (AD), a devastating disease caused by the progressive but rapid degeneration of cortical tissue. Some biomarkers we already know have a significant association with AD, such as amyloid beta (Aβ) and tau, as well as the ratio of concentrations of other Aβ isoforms. In addition, several novel biomarkers are emerging that can also be used as diagnostic fluid biomarkers for AD, but many studies are still needed before we can consider them reliable. The U.S. Federal Food and Drug Administration recently announced its final ruling to regulate laboratory-developed tests (LDTs) as medical devices, which can significantly impact LDT development. In this narrative review, we discuss the current status of fluid biomarkers used to diagnose early AD, their potential and limitations, and the impact caused by the FDA's decision and strategies to help developers navigate the complex changes in the regulatory landscape of LDTs.
Collapse
Affiliation(s)
- Angelo M Jamerlan
- Department of Bionanotechnology, Bionano Research Institute, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 13120, Gyeonggi-do, Republic of Korea
| | - Seong Soo A An
- Department of Bionanotechnology, Bionano Research Institute, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 13120, Gyeonggi-do, Republic of Korea.
| | - John P Hulme
- Department of Bionanotechnology, Bionano Research Institute, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 13120, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
29
|
Safransky M, Groh JR, Blennow K, Zetterberg H, Tripodis Y, Martin B, Weller J, Asken BM, Rabinovici GD, Qiu WWQ, McKee AC, Stein TD, Mez J, Henson RL, Long J, Morris JC, Perrin RJ, Schindler SE, Alosco ML. Lumipulse-Measured Cerebrospinal Fluid Biomarkers for the Early Detection of Alzheimer Disease. Neurology 2024; 103:e209866. [PMID: 39496102 PMCID: PMC11540457 DOI: 10.1212/wnl.0000000000209866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 08/20/2024] [Indexed: 11/06/2024] Open
Abstract
BACKGROUND AND OBJECTIVES CSF biomarkers of Aβ42 and phosphorylated tau (p-tau181) are used clinically for the detection of Alzheimer disease (AD) pathology during life. CSF biomarker validation studies have largely used clinical diagnoses and/or amyloid PET imaging as the reference standard. The few existing CSF-to-autopsy studies have been restricted to late-stage AD. This CSF-to-autopsy study investigated associations between CSF biomarkers of AD and AD neuropathologic changes among brain donors who had normal cognition at the time of lumbar puncture (LP). METHODS This was a retrospective study of brain donors from the National Alzheimer's Coordinating Center who had normal cognition at the time of LP and who had measurements of CSF Aβ42 and p-tau181 performed with Lumipulse assays. All brain donors were from Washington University Knight ADRC. Staging of AD neuropathologic change (ADNC) was made based on National Institute on Aging-Alzheimer's Association criteria. For this study, participants were divided into 2 categories: "AD-" (no AD/low ADNC) and "AD+" (intermediate/high ADNC). Accuracy of each biomarker for discriminating AD status was evaluated using area under the curve (AUC) statistics generated using predicted probabilities from binary logistic regressions that controlled for age, sex, APOE ε4, and interval between LP and death. RESULTS The average age at LP was 79.3 years (SD = 5.6), and the average age at death was 87.1 years (SD = 6.5). Of the 49 brain donors, 24 (49%) were male and 47 (95.9%) were White. 20 (40.8%) had autopsy-confirmed AD. The average interval from LP until death was 7.76 years (SD = 4.31). CSF p-tau181/Aβ42 was the optimal predictor of AD, having excellent discrimination accuracy (AUC = 0.97, 95% CI 0.94-1.00, p = 0.003). CSF p-tau181 alone had the second-best discrimination accuracy (AUC = 0.92, 95% CI 0.84-1.00, p = 0.001), followed by CSF Aβ42 alone (AUC = 0.92, 95% CI 0.85-1.00, p = 0.007), while CSF t-tau had the numerically lowest discrimination accuracy (AUC = 0.87, 95% CI 0.76-0.97, p = 0.005). Effects remained after controlling for prevalent comorbid neuropathologies. CSF p-tau181/Aβ42 was strongly associated with CERAD ratings of neuritic amyloid plaque scores and Braak staging of NFTs. DISCUSSION This study supports Lumipulse-measured CSF Aβ42 and p-tau181 and, particularly, the ratio of p-tau181 to Aβ42, for the early detection of AD pathophysiologic processes. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that Lumipulse measures of p-tau181/Aβ42 in the CSF accurately discriminated cognitively normal participants with and without Alzheimer disease neuropathologic change.
Collapse
Affiliation(s)
- Michelle Safransky
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Jenna R Groh
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Kaj Blennow
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Henrik Zetterberg
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Yorghos Tripodis
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Brett Martin
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Jason Weller
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Breton M Asken
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Gil D Rabinovici
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Wendy Wei Qiao Qiu
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Ann C McKee
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Thor D Stein
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Jesse Mez
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Rachel L Henson
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Justin Long
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - John C Morris
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Richard J Perrin
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Suzanne E Schindler
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Michael L Alosco
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| |
Collapse
|
30
|
Shukla R, Singh TR. AlzGenPred - CatBoost-based gene classifier for predicting Alzheimer's disease using high-throughput sequencing data. Sci Rep 2024; 14:30294. [PMID: 39639110 PMCID: PMC11621786 DOI: 10.1038/s41598-024-82208-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: 08/02/2024] [Accepted: 12/03/2024] [Indexed: 12/07/2024] Open
Abstract
AD is a progressive neurodegenerative disorder characterized by memory loss. Due to the advancement in next-generation sequencing, an enormous amount of AD-associated genomics data is available. However, the information about the involvement of these genes in AD association is still a research topic. Therefore, AlzGenPred is developed to identify the AD-associated genes using machine-learning. A total of 13,504 features derived from eight sequence-encoding schemes were generated and evaluated using 16 machine learning algorithms. Network-based features significantly outperformed sequence-based features, effectively distinguishing AD-associated genes. In contrast, sequence-based features failed to classify accurately. To improve performance, we generated 24 fused features (6020 D) from sequence-based encodings, increasing accuracy by 5-7% using a two-step lightGBM-based recursive feature selection method. However, accuracy remained below 70% even after hyperparameter tuning. Therefore, network-based features were used to generate the CatBoost-based ML method AlzGenPred with 96.55% accuracy and 98.99% AUROC. The developed method is tested on the AlzGene dataset where it showed 96.43% accuracy. Then the model was validated using the transcriptomics dataset. AlzGenPred provides a reliable and user-friendly tool for identifying potential AD biomarkers, accelerating biomarker discovery, and advancing our understanding of AD. It is available at https://www.bioinfoindia.org/alzgenpred/ and https://github.com/shuklarohit815/AlzGenPred .
Collapse
Affiliation(s)
- Rohit Shukla
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology (JUIT), Waknaghat, Solan, 173234, H.P., India
- Center of Excellence for Aging and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, 33613, FL, USA
| | - Tiratha Raj Singh
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology (JUIT), Waknaghat, Solan, 173234, H.P., India.
- Centre of Healthcare Technologies and Informatics (CEHTI), Jaypee University of Information Technology (JUIT), Waknaghat, Solan, 173234, H.P., India.
| |
Collapse
|
31
|
Bamford AR, Adams JN, Kim S, McMillan LC, Malhas R, Mapstone M, Hitt BD, Yassa MA, Thomas EA. The amyloid beta 42/38 ratio as a plasma biomarker of early memory deficits in cognitively unimpaired older adults. Neurobiol Aging 2024; 144:12-18. [PMID: 39241563 PMCID: PMC11706698 DOI: 10.1016/j.neurobiolaging.2024.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 08/13/2024] [Accepted: 08/28/2024] [Indexed: 09/09/2024]
Abstract
The amyloid beta (Aβ) 42/40 ratio has been widely studied as a biomarker in Alzheimer's disease (AD); however, other Aβ peptides could also represent relevant biomarkers. We measured levels of Aβ38/40/42 in plasma samples from cognitively-unimpaired older adults and determined the relationships between Aβ levels and amyloid positron-emission-tomography (PET) and performance on a learning and memory task. We found that all Aβ peptides individually and the Aβ42/40 ratio, but not the Aβ42/38 ratio, were significantly correlated with brain amyloid (Aβ-PET). Multiple linear modeling, adjusting for age, sex, education, APOE4 and Aβ-PET showed significant associations between the Aβ42/38 ratio and memory. Further, associations between the Aβ42/38 ratio and learning scores were stronger in males and in Aβ-PET-negative individuals. In contrast, no significant associations were detected between the Aβ42/40 ratio and any learning measure. These studies implicate the Aβ42/38 ratio as a biomarker to assess early memory deficits and underscore the utility of the Aβ38 fragment as an important biomarker in the AD field.
Collapse
Affiliation(s)
- Alison R Bamford
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA; Institute for Interdisciplinary Salivary Bioscience Research, University of California Irvine, Irvine, CA, USA
| | - Jenna N Adams
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA; Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA, USA
| | - Soyun Kim
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA; Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA, USA
| | - Liv C McMillan
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA
| | - Rond Malhas
- Department of Neurology, School of Medicine, University of California Irvine, Irvine, CA, USA
| | - Mark Mapstone
- Department of Neurology, School of Medicine, University of California Irvine, Irvine, CA, USA
| | - Brian D Hitt
- Department of Neurology, School of Medicine, University of California Irvine, Irvine, CA, USA
| | - Michael A Yassa
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA; Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA, USA
| | - Elizabeth A Thomas
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA; Institute for Interdisciplinary Salivary Bioscience Research, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
32
|
Studart-Neto A, Barbosa BJAP, Coutinho AM, de Souza LC, Schilling LP, da Silva MNM, Castilhos RM, Bertolucci PHF, Borelli WV, Gomes HR, Fernandes GBP, Barbosa MT, Balthazar MLF, Frota NAF, Forlenza OV, Smid J, Brucki SMD, Caramelli P, Nitrini R, Engelhardt E, Resende EDPF. Guidelines for the use and interpretation of Alzheimer's disease biomarkers in clinical practice in Brazil: recommendations from the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology. Dement Neuropsychol 2024; 18:e2024C001. [PMID: 39534442 PMCID: PMC11556292 DOI: 10.1590/1980-5764-dn-2024-c001] [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: 08/01/2024] [Accepted: 08/16/2024] [Indexed: 11/16/2024] Open
Abstract
In recent years, the diagnostic accuracy of Alzheimer's disease has been enhanced by the development of different types of biomarkers that indicate the presence of neuropathological processes. In addition to improving patient selection for clinical trials, biomarkers can assess the effects of new treatments on pathological processes. However, there is concern about the indiscriminate and poorly supported use of biomarkers, especially in asymptomatic individuals or those with subjective cognitive decline. Difficulties interpreting these tests, high costs, and unequal access make this scenario even more challenging in healthcare. This article presents the recommendations from the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology (Departamento Científico de Neurologia Cognitiva e Envelhecimento da Academia Brasileira de Neurologia) regarding the rational use and interpretation of Alzheimer's disease biomarkers in clinical practice. The clinical diagnosis of cognitive-behavioral syndrome is recommended as the initial step to guide the request for biomarkers.
Collapse
Affiliation(s)
- Adalberto Studart-Neto
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil
| | - Breno José Alencar Pires Barbosa
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal de Pernambuco, Hospital das Clínicas, Recife, Centro de Ciências Médicas, Recife PE, Brazil
- Universidade Federal de Pernambuco, Empresa Brasileira de Serviços Hospitalares, Hospital das Clínicas, Departamento de Neurologia, Recife PE, Brazil
| | - Artur Martins Coutinho
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Instituto de Radiologia, Centro de Medicina Nuclear, Laboratório de Investigação Médica (LIM 43), São Paulo SP, Brazil
- Hospital Sírio-Libanês, Medicina Nuclear e Serviço de PET-CT, São Paulo SP, Brazil
| | - Leonardo Cruz de Souza
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Unidade de Neurologia Cognitiva e do Comportamento, Belo Horizonte MG, Brazil
| | - Lucas Porcello Schilling
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Pontifícia Universidade do Rio Grande do Sul, Escola de Medicina, Serviço de Neurologia, Porto Alegre RS, Brazil
| | - Mari Nilva Maia da Silva
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Hospital Nina Rodrigues, Serviço de Neuropsiquiatria, São Luís MA, Brazil
| | - Raphael Machado Castilhos
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Hospital de Clínicas de Porto Alegre, Serviço de Neurologia, Centro de Neurologia Cognitiva e Comportamental, Porto Alegre RS, Brazil
| | - Paulo Henrique Ferreira Bertolucci
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal de São Paulo, Escola Paulista de Medicina, Departamento de Neurologia e Neurocirurgia, São Paulo SP, Brazil
| | - Wyllians Vendramini Borelli
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal do Rio Grande do Sul, Instituto de Ciências Básicas da Saúde, Departamento de Ciências Morfológicas, Porto Alegre RS, Brazil
| | - Hélio Rodrigues Gomes
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Laboratório de Líquido Cefalorraquidiano, São Paulo SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina, Laboratório de Investigação Médica (LIM 15), São Paulo SP, Brazil
- Departamento Científico de Líquido Cefalorraquiano, Academia Brasileira de Neurologia, São Paulo SP, Brazil
| | | | - Maira Tonidandel Barbosa
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Unidade de Neurologia Cognitiva e do Comportamento, Belo Horizonte MG, Brazil
| | - Marcio Luiz Figueredo Balthazar
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Estadual de Campinas, Faculdade de Ciências Médicas, Departamento de Neurologia, Campinas SP, Brazil
| | - Norberto Anízio Ferreira Frota
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Hospital Geral de Fortaleza, Serviço de Neurologia, Fortaleza CE, Brazil
- Universidade de Fortaleza, Fortaleza, CE, Brazil
| | - Orestes Vicente Forlenza
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Instituto de Psiquiatria, Laboratório de Neurociências, São Paulo SP, Brazil
| | - Jerusa Smid
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil
| | - Sonia Maria Dozzi Brucki
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil
| | - Paulo Caramelli
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Unidade de Neurologia Cognitiva e do Comportamento, Belo Horizonte MG, Brazil
| | - Ricardo Nitrini
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil
| | - Eliasz Engelhardt
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal do Rio de Janeiro, Instituto de Neurologia Deolindo Couto, Rio de Janeiro RJ, Brazil
- Universidade Federal do Rio de Janeiro, Instituto de Psiquiatria, Rio de Janeiro RJ, Brazil
| | - Elisa de Paula França Resende
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Unidade de Neurologia Cognitiva e do Comportamento, Belo Horizonte MG, Brazil
| |
Collapse
|
33
|
Qi X, Gao L, Qi L. Genetic determinants of cerebrospinal fluid metabolites and risk of Guillain-Barré syndrome: A bidirectional two-sample Mendelian randomization study. Medicine (Baltimore) 2024; 103:e40352. [PMID: 39533579 PMCID: PMC11557065 DOI: 10.1097/md.0000000000040352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024] Open
Abstract
This study aims to investigate the potential causal relationship between cerebrospinal fluid (CSF) metabolites and Guillain-Barré syndrome (GBS) using a bidirectional two-sample Mendelian randomization (MR) approach. Publicly available summary data from genome-wide association studies (GWAS) were utilized for comprehensive analysis. The CSF metabolite GWAS summary data were extracted from a GWAS conducted by Panyard et al encompassing 338 CSF metabolites in European participants (n = 291). GWAS summary statistics for GBS were obtained from the FinnGen consortium (n = 215,931) comprising European populations. The primary method for MR analysis was the inverse variance weighted method. Various sensitivity analyses were conducted to assess the heterogeneity and pleiotropy of the findings. In the forward MR analysis, we identified a causal relationship between 15 CSF metabolites, including ribitol levels (odds ratio = 3.833, 95% confidence interval: 1.949-7.540, P = 9.87E-05), and the risk of developing GBS. In the reverse MR analysis, we found a causal relationship between GBS and 21 CSF metabolites, including gamma-glutamylphenylalanine levels (odds ratio = 0.934, 95% confidence interval: 0.904-0.966, P = 7.10E-05). No evidence of heterogeneity or horizontal pleiotropy was found in the MR analysis. Our findings suggest that the identified CSF metabolites and metabolic pathways can serve as valuable biomarkers for clinical screening and prevention of GBS. They may also be considered as candidate molecules for future research into the underlying mechanisms and for selecting drug targets.
Collapse
Affiliation(s)
- Xiangjia Qi
- Department of Neurology, Liaocheng People’s Hospital, Liaocheng, China
| | - Liqian Gao
- Department of Neurology, Liaocheng People’s Hospital, Liaocheng, China
| | - Lifeng Qi
- Department of Neurology, Liaocheng People’s Hospital, Liaocheng, China
| |
Collapse
|
34
|
Tenchov R, Sasso JM, Zhou QA. Alzheimer's Disease: Exploring the Landscape of Cognitive Decline. ACS Chem Neurosci 2024; 15:3800-3827. [PMID: 39392435 PMCID: PMC11587518 DOI: 10.1021/acschemneuro.4c00339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/26/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline, memory loss, and impaired daily functioning. The pathology of AD is marked by the accumulation of amyloid beta plaques and tau protein tangles in the brain, along with neuroinflammation and synaptic dysfunction. Genetic factors, such as mutations in APP, PSEN1, and PSEN2 genes, as well as the APOE ε4 allele, contribute to increased risk of acquiring AD. Currently available treatments provide symptomatic relief but do not halt disease progression. Research efforts are focused on developing disease-modifying therapies that target the underlying pathological mechanisms of AD. Advances in identification and validation of reliable biomarkers for AD hold great promise for enhancing early diagnosis, monitoring disease progression, and assessing treatment response in clinical practice in effort to alleviate the burden of this devastating disease. In this paper, we analyze data from the CAS Content Collection to summarize the research progress in Alzheimer's disease. We examine the publication landscape in effort to provide insights into current knowledge advances and developments. We also review the most discussed and emerging concepts and assess the strategies to combat the disease. We explore the genetic risk factors, pharmacological targets, and comorbid diseases. Finally, we inspect clinical applications of products against AD with their development pipelines and efforts for drug repurposing. The objective of this review is to provide a broad overview of the evolving landscape of current knowledge regarding AD, to outline challenges, and to evaluate growth opportunities to further efforts in combating the disease.
Collapse
Affiliation(s)
- Rumiana Tenchov
- CAS, a division of the American Chemical
Society, Columbus Ohio 43210, United States
| | - Janet M. Sasso
- CAS, a division of the American Chemical
Society, Columbus Ohio 43210, United States
| | | |
Collapse
|
35
|
Costanzo M, Cutrona C, Leodori G, Malimpensa L, D'antonio F, Conte A, Belvisi D. Exploring easily accessible neurophysiological biomarkers for predicting Alzheimer's disease progression: a systematic review. Alzheimers Res Ther 2024; 16:244. [PMID: 39497149 PMCID: PMC11533378 DOI: 10.1186/s13195-024-01607-4] [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: 01/25/2024] [Accepted: 10/19/2024] [Indexed: 11/06/2024]
Abstract
Alzheimer disease (AD) remains a significant global health concern. The progression from preclinical stages to overt dementia has become a crucial point of interest for researchers. This paper reviews the potential of neurophysiological biomarkers in predicting AD progression, based on a systematic literature search following PRISMA guidelines, including 55 studies. EEG-based techniques have been predominantly employed, whereas TMS studies are less common. Among the investigated neurophysiological measures, spectral power measurements and event-related potentials-based measures, including P300 and N200 latencies, have emerged as the most consistent and reliable biomarkers for predicting the likelihood of conversion to AD. In addition, TMS-based indices of cortical excitability and synaptic plasticity have also shown potential in assessing the risk of conversion to AD. However, concerns persist regarding the methodological discrepancies among studies, the accuracy of these neurophysiological measures in comparison to established AD biomarkers, and their immediate clinical applicability. Further research is needed to validate the predictive capabilities of EEG and TMS measures. Advancements in this area could lead to cost-effective, reliable biomarkers, enhancing diagnostic processes and deepening our understanding of AD pathophysiology.
Collapse
Affiliation(s)
- Matteo Costanzo
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università 30, Rome, 00185, RM, Italy
- Department of Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome, 00161, Italy
| | - Carolina Cutrona
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università 30, Rome, 00185, RM, Italy
| | - Giorgio Leodori
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università 30, Rome, 00185, RM, Italy
- IRCCS Neuromed, Via Atinense 18, Pozzilli, 86077, IS, Italy
| | | | - Fabrizia D'antonio
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università 30, Rome, 00185, RM, Italy
| | - Antonella Conte
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università 30, Rome, 00185, RM, Italy
- IRCCS Neuromed, Via Atinense 18, Pozzilli, 86077, IS, Italy
| | - Daniele Belvisi
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università 30, Rome, 00185, RM, Italy.
- IRCCS Neuromed, Via Atinense 18, Pozzilli, 86077, IS, Italy.
| |
Collapse
|
36
|
Li Y, Wang H, Wang Y, Chen Z, Liu Y, Tian W, Kang X, Pashang A, Kulasiri D, Yang X, Li HW, Zhang Y. Alterations in the axon initial segment plasticity is involved in early pathogenesis in Alzheimer's disease. MedComm (Beijing) 2024; 5:e768. [PMID: 39415847 PMCID: PMC11473794 DOI: 10.1002/mco2.768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/05/2024] [Accepted: 09/08/2024] [Indexed: 10/19/2024] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder, characterized by the early presence of amyloid-β (Aβ) and hyperphosphorylated tau. Identifying the neuropathological changes preceding cognitive decline is crucial for early intervention. Axon initial segment (AIS) maintains the orderly structure of the axon and is responsible for initiating action potentials (APs). To investigate the role of AIS in early stages of AD pathogenesis, we focused on alterations in the AIS of neurons from APP/PS1 mouse models harboring familial AD mutations. AIS length and electrophysiological properties were assessed in neurons using immunostaining and patch-clamp techniques. The expression and function of ankyrin G (AnkG) isoforms were evaluated by western blot and rescue experiments. We observed a significant shortening of AIS in APP/PS1 mice, which correlated with impaired action potential propagation. Furthermore, a decrease in the 480 kDa isoform of AnkG was observed. Rescue of this isoform restored AIS plasticity and improved long-term potentiation in APP/PS1 neurons. Our study implicates AIS plasticity alterations and AnkG dysregulation as early events in AD. The restoration of AIS integrity by the 480 kDa AnkG isoform presents a potential therapeutic strategy for AD, underscoring the importance of targeting AIS stability in neurodegenerative diseases.
Collapse
Affiliation(s)
- Yu Li
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijingChina
| | - Han Wang
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijingChina
| | - Yiming Wang
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijingChina
| | - Zhiya Chen
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijingChina
| | - Yiqiong Liu
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijingChina
| | - Wu Tian
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijingChina
| | - Xinrui Kang
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijingChina
| | - Abolghasem Pashang
- Centre for Advanced Computational Solutions (C‐fACS)AGLS FacultyLincoln UniversityCanterburyNew Zealand
| | - Don Kulasiri
- Centre for Advanced Computational Solutions (C‐fACS)AGLS FacultyLincoln UniversityCanterburyNew Zealand
| | - Xiaoli Yang
- Division of Life Sciences and MedicineDepartment of NeurologyInstitute on Aging and Brain DisordersThe First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
- Neurodegenerative Disorder Research CenterAnhui Province Key Laboratory of Biomedical Aging ResearchDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Hung Wing Li
- Department of ChemistryThe Chinese University of Hong KongHong KongChina
| | - Yan Zhang
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijingChina
| |
Collapse
|
37
|
Zhou RZ, Duell F, Axenhus M, Jönsson L, Winblad B, Tjernberg LO, Schedin-Weiss S. A glycan biomarker predicts cognitive decline in amyloid- and tau-negative patients. Brain Commun 2024; 6:fcae371. [PMID: 39494362 PMCID: PMC11528473 DOI: 10.1093/braincomms/fcae371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/18/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024] Open
Abstract
Early detection of Alzheimer's disease is vital for timely treatment. Existing biomarkers for Alzheimer's disease reflect amyloid- and tau-related pathology, but it is unknown whether the disease can be detected before cerebral amyloidosis is observed. N-glycosylation has been suggested as an upstream regulator of both amyloid and tau pathology, and levels of the N-glycan structure bisecting N-acetylglucosamine (GlcNAc) correlate with tau in blood and CSF already at pre-clinical stages of the disease. Therefore, we aimed to evaluate whether bisecting GlcNAc could predict future cognitive decline in patients from a memory clinic cohort, stratified by amyloid/tau status. We included 251 patients (mean age: 65.6 ± 10.6 years, 60.6% female) in the GEDOC cohort, from the Memory Clinic at Karolinska University Hospital, Stockholm, Sweden. Patients were classified as amyloid/tau positive or negative based on CSF biomarkers. Cognitive decline, measured by longitudinal Mini-Mental State Examination scores, was followed for an average of 10.7 ± 4.1 years and modelled using non-linear mixed effects models. Additionally, bisecting GlcNAc levels were measured in hippocampus and cortex with lectin-based immunohistochemistry in 10 Alzheimer's disease and control brains. We found that CSF bisecting GlcNAc levels were elevated in tau-positive individuals compared with tau-negative individuals, but not in amyloid-positive individuals compared with amyloid-negative individuals. In the whole sample, high levels of CSF bisecting GlcNAc predicted earlier cognitive decline. Strikingly, amyloid/tau stratification showed that high CSF bisecting GlcNAc levels predicted earlier cognitive decline in amyloid-negative patients (β = 2.53 ± 0.85 years, P = 0.003) and tau-negative patients (β = 2.43 ± 1.01 years, P = 0.017), but not in amyloid- or tau-positive patients. Finally, histochemical analysis of bisecting GlcNAc showed increased levels in neurons in hippocampus and cortex of Alzheimer's disease compared with control brain (fold change = 1.44-1.49, P < 0.001). In conclusion, high CSF levels of bisecting GlcNAc reflected neuronal pathology and predicted cognitive decline in amyloid- and tau-negative individuals, suggesting that abnormal glycosylation precedes cerebral amyloidosis and tau hyper-phosphorylation in Alzheimer's disease. Bisecting GlcNAc is a promising novel early biomarker for Alzheimer's disease.
Collapse
Affiliation(s)
- Robin Ziyue Zhou
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna 171 64, Sweden
| | - Frida Duell
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna 171 64, Sweden
| | - Michael Axenhus
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna 171 64, Sweden
| | - Linus Jönsson
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna 171 64, Sweden
| | - Bengt Winblad
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna 171 64, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Huddinge 141 57, Sweden
| | - Lars O Tjernberg
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna 171 64, Sweden
| | - Sophia Schedin-Weiss
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna 171 64, Sweden
| |
Collapse
|
38
|
Alarjani M, Almarri B. fMRI-based Alzheimer's disease detection via functional connectivity analysis: a systematic review. PeerJ Comput Sci 2024; 10:e2302. [PMID: 39650470 PMCID: PMC11622848 DOI: 10.7717/peerj-cs.2302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 08/12/2024] [Indexed: 12/11/2024]
Abstract
Alzheimer's disease is a common brain disorder affecting many people worldwide. It is the primary cause of dementia and memory loss. The early diagnosis of Alzheimer's disease is essential to provide timely care to AD patients and prevent the development of symptoms of this disease. Various non-invasive techniques can be utilized to diagnose Alzheimer's in its early stages. These techniques include functional magnetic resonance imaging, electroencephalography, positron emission tomography, and diffusion tensor imaging. They are mainly used to explore functional and structural connectivity of human brains. Functional connectivity is essential for understanding the co-activation of certain brain regions co-activation. This systematic review scrutinizes various works of Alzheimer's disease detection by analyzing the learning from functional connectivity of fMRI datasets that were published between 2018 and 2024. This work investigates the whole learning pipeline including data analysis, standard preprocessing phases of fMRI, feature computation, extraction and selection, and the various machine learning and deep learning algorithms that are used to predict the occurrence of Alzheimer's disease. Ultimately, the paper analyzed results on AD and highlighted future research directions in medical imaging. There is a need for an efficient and accurate way to detect AD to overcome the problems faced by patients in the early stages.
Collapse
Affiliation(s)
- Maitha Alarjani
- Department of Computer Science, King Faisal University, Alhsa, Saudi Arabia
| | - Badar Almarri
- Department of Computer Science, King Faisal University, Alhsa, Saudi Arabia
| |
Collapse
|
39
|
Alshehri RS, Abuzinadah AR, Alrawaili MS, Alotaibi MK, Alsufyani HA, Alshanketi RM, AlShareef AA. A Review of Biomarkers of Amyotrophic Lateral Sclerosis: A Pathophysiologic Approach. Int J Mol Sci 2024; 25:10900. [PMID: 39456682 PMCID: PMC11507293 DOI: 10.3390/ijms252010900] [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: 09/10/2024] [Revised: 10/03/2024] [Accepted: 10/05/2024] [Indexed: 10/28/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by progressive degeneration of upper and lower motor neurons. The heterogeneous nature of ALS at the clinical, genetic, and pathological levels makes it challenging to develop diagnostic and prognostic tools that fit all disease phenotypes. Limitations associated with the functional scales and the qualitative nature of mainstay electrophysiological testing prompt the investigation of more objective quantitative assessment. Biofluid biomarkers have the potential to fill that gap by providing evidence of a disease process potentially early in the disease, its progression, and its response to therapy. In contrast to other neurodegenerative diseases, no biomarker has yet been validated in clinical use for ALS. Several fluid biomarkers have been investigated in clinical studies in ALS. Biofluid biomarkers reflect the different pathophysiological processes, from protein aggregation to muscle denervation. This review takes a pathophysiologic approach to summarizing the findings of clinical studies utilizing quantitative biofluid biomarkers in ALS, discusses the utility and shortcomings of each biomarker, and highlights the superiority of neurofilaments as biomarkers of neurodegeneration over other candidate biomarkers.
Collapse
Affiliation(s)
- Rawiah S. Alshehri
- Department of Physiology, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia; (R.S.A.); (H.A.A.)
| | - Ahmad R. Abuzinadah
- Department of Neurology, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia; (M.S.A.); (A.A.A.)
- Neuromuscular Medicine Unit, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| | - Moafaq S. Alrawaili
- Department of Neurology, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia; (M.S.A.); (A.A.A.)
- Neuromuscular Medicine Unit, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| | - Muteb K. Alotaibi
- Neurology Department, Prince Sultan Military Medical City, Riyadh 12233, Saudi Arabia;
| | - Hadeel A. Alsufyani
- Department of Physiology, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia; (R.S.A.); (H.A.A.)
| | - Rajaa M. Alshanketi
- Internal Medicine Department, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah 22252, Saudi Arabia;
| | - Aysha A. AlShareef
- Department of Neurology, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia; (M.S.A.); (A.A.A.)
- Neuromuscular Medicine Unit, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| |
Collapse
|
40
|
Khan A, Zubair S, Shuaib M, Sheneamer A, Alam S, Assiri B. Development of a robust parallel and multi-composite machine learning model for improved diagnosis of Alzheimer's disease: correlation with dementia-associated drug usage and AT(N) protein biomarkers. Front Neurosci 2024; 18:1391465. [PMID: 39308946 PMCID: PMC11412962 DOI: 10.3389/fnins.2024.1391465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 08/12/2024] [Indexed: 09/25/2024] Open
Abstract
Introduction Machine learning (ML) algorithms and statistical modeling offer a potential solution to offset the challenge of diagnosing early Alzheimer's disease (AD) by leveraging multiple data sources and combining information on neuropsychological, genetic, and biomarker indicators. Among others, statistical models are a promising tool to enhance the clinical detection of early AD. In the present study, early AD was diagnosed by taking into account characteristics related to whether or not a patient was taking specific drugs and a significant protein as a predictor of Amyloid-Beta (Aβ), tau, and ptau [AT(N)] levels among participants. Methods In this study, the optimization of predictive models for the diagnosis of AD pathologies was carried out using a set of baseline features. The model performance was improved by incorporating additional variables associated with patient drugs and protein biomarkers into the model. The diagnostic group consisted of five categories (cognitively normal, significant subjective memory concern, early mildly cognitively impaired, late mildly cognitively impaired, and AD), resulting in a multinomial classification challenge. In particular, we examined the relationship between AD diagnosis and the use of various drugs (calcium and vitamin D supplements, blood-thinning drugs, cholesterol-lowering drugs, and cognitive drugs). We propose a hybrid-clinical model that runs multiple ML models in parallel and then takes the majority's votes, enhancing the accuracy. We also assessed the significance of three cerebrospinal fluid biomarkers, Aβ, tau, and ptau in the diagnosis of AD. We proposed that a hybrid-clinical model be used to simulate the MRI-based data, with five diagnostic groups of individuals, with further refinement that includes preclinical characteristics of the disorder. The proposed design builds a Meta-Model for four different sets of criteria. The set criteria are as follows: to diagnose from baseline features, baseline and drug features, baseline and protein features, and baseline, drug and protein features. Results We were able to attain a maximum accuracy of 97.60% for baseline and protein data. We observed that the constructed model functioned effectively when all five drugs were included and when any single drug was used to diagnose the response variable. Interestingly, the constructed Meta-Model worked well when all three protein biomarkers were included, as well as when a single protein biomarker was utilized to diagnose the response variable. Discussion It is noteworthy that we aimed to construct a pipeline design that incorporates comprehensive methodologies to detect Alzheimer's over wide-ranging input values and variables in the current study. Thus, the model that we developed could be used by clinicians and medical experts to advance Alzheimer's diagnosis and as a starting point for future research into AD and other neurodegenerative syndromes.
Collapse
Affiliation(s)
- Afreen Khan
- Department of Computer Application, Faculty of Engineering & IT, Integral University, Lucknow, India
| | - Swaleha Zubair
- Department of Computer Science, Faculty of Science, Aligarh Muslim University, Aligarh, India
| | - Mohammed Shuaib
- Department of Computer Science, College of Engineering and Computer Science, Jazan University, Jazan, Saudi Arabia
| | - Abdullah Sheneamer
- Department of Computer Science, College of Engineering and Computer Science, Jazan University, Jazan, Saudi Arabia
| | - Shadab Alam
- Department of Computer Science, College of Engineering and Computer Science, Jazan University, Jazan, Saudi Arabia
| | - Basem Assiri
- Department of Computer Science, College of Engineering and Computer Science, Jazan University, Jazan, Saudi Arabia
| |
Collapse
|
41
|
Gebre RK, Graff-Radford J, Ramanan VK, Raghavan S, Hofrenning EI, Przybelski SA, Nguyen AT, Lesnick TG, Gunter JL, Algeciras-Schimnich A, Knopman DS, Machulda MM, Vassilaki M, Lowe VJ, Jack CR, Petersen RC, Vemuri P. Can integration of Alzheimer's plasma biomarkers with MRI, cardiovascular, genetics, and lifestyle measures improve cognition prediction? Brain Commun 2024; 6:fcae300. [PMID: 39291164 PMCID: PMC11406552 DOI: 10.1093/braincomms/fcae300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/13/2024] [Accepted: 09/03/2024] [Indexed: 09/19/2024] Open
Abstract
There is increasing interest in Alzheimer's disease related plasma biomarkers due to their accessibility and scalability. We hypothesized that integrating plasma biomarkers with other commonly used and available participant data (MRI, cardiovascular factors, lifestyle, genetics) using machine learning (ML) models can improve individual prediction of cognitive outcomes. Further, our goal was to evaluate the heterogeneity of these predictors across different age strata. This longitudinal study included 1185 participants from the Mayo Clinic Study of Aging who had complete plasma analyte work-up at baseline. We used the Quanterix Simoa immunoassay to measure neurofilament light, Aβ1-42 and Aβ1-40 (used as Aβ42/Aβ40 ratio), glial fibrillary acidic protein, and phosphorylated tau 181 (p-tau181). Participants' brain health was evaluated through gray and white matter structural MRIs. The study also considered cardiovascular factors (hyperlipidemia, hypertension, stroke, diabetes, chronic kidney disease), lifestyle factors (area deprivation index, body mass index, cognitive and physical activities), and genetic factors (APOE, single nucleotide polymorphisms, and polygenic risk scores). An ML model was developed to predict cognitive outcomes at baseline and decline (slope). Three models were created: a base model with groups of risk factors as predictors, an enhanced model included socio-demographics, and a final enhanced model by incorporating plasma and socio-demographics into the base models. Models were explained for three age strata: younger than 65 years, 65-80 years, and older than 80 years, and further divided based on amyloid positivity status. Regardless of amyloid status the plasma biomarkers showed comparable performance (R² = 0.15) to MRI (R² = 0.18) and cardiovascular measures (R² = 0.10) when predicting cognitive decline. Inclusion of cardiovascular or MRI measures with plasma in the presence of socio-demographic improved cognitive decline prediction (R² = 0.26 and 0.27). For amyloid positive individuals Aβ42/Aβ40, glial fibrillary acidic protein and p-tau181 were the top predictors of cognitive decline while Aβ42/Aβ40 was prominent for amyloid negative participants across all age groups. Socio-demographics explained a large portion of the variance in the amyloid negative individuals while the plasma biomarkers predominantly explained the variance in amyloid positive individuals (21% to 37% from the younger to the older age group). Plasma biomarkers performed similarly to MRI and cardiovascular measures when predicting cognitive outcomes and combining them with either measure resulted in better performance. Top predictors were heterogeneous between cross-sectional and longitudinal cognition models, across age groups, and amyloid status. Multimodal approaches will enhance the usefulness of plasma biomarkers through careful considerations of a study population's socio-demographics, brain and cardiovascular health.
Collapse
Affiliation(s)
- Robel K Gebre
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Vijay K Ramanan
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | - Scott A Przybelski
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Aivi T Nguyen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Timothy G Lesnick
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | - David S Knopman
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Mary M Machulda
- Department of Psychology, Mayo Clinic, Rochester, MN 55905, USA
| | - Maria Vassilaki
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Clifford R Jack
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | |
Collapse
|
42
|
Azargoonjahromi A. Immunotherapy in Alzheimer's disease: focusing on the efficacy of gantenerumab on amyloid-β clearance and cognitive decline. J Pharm Pharmacol 2024; 76:1115-1131. [PMID: 38767981 DOI: 10.1093/jpp/rgae066] [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: 01/18/2024] [Accepted: 05/08/2024] [Indexed: 05/22/2024]
Abstract
Gantenerumab, a human monoclonal antibody (mAb), has been thought of as a potential agent to treat Alzheimer's disease (AD) by specifically targeting regions of the amyloid-β (Aβ) peptide sequence. Aβ protein accumulation in the brain leads to amyloid plaques, causing neuroinflammation, oxidative stress, neuronal damage, and neurotransmitter dysfunction, thereby causing cognitive decline in AD. Gantenerumab involves disrupting Aβ aggregation and promoting the breakdown of larger Aβ aggregates into smaller fragments, which facilitates the action of Aβ-degrading enzymes in the brain, thus slowing down the progression of AD. Moreover, Gantenerumab acts as an opsonin, coating Aβ plaques and enhancing their recognition by immune cells, which, combined with its ability to improve the activity of microglia, makes it an intriguing candidate for promoting Aβ plaque clearance. Indeed, the multifaceted effects of Gantenerumab, including Aβ disaggregation, enhanced immune recognition, and improved microglia activity, may position it as a promising therapeutic approach for AD. Of note, reports suggest that Gantenerumab, albeit its capacity to reduce or eliminate Aβ, has not demonstrated effectiveness in reducing cognitive decline. This review, after providing an overview of immunotherapy approaches that target Aβ in AD, explores the efficacy of Gantenerumab in reducing Aβ levels and cognitive decline.
Collapse
|
43
|
Kale MB, Bhondge HM, Wankhede NL, Shende PV, Thanekaer RP, Aglawe MM, Rahangdale SR, Taksande BG, Pandit SB, Upaganlawar AB, Umekar MJ, Kopalli SR, Koppula S. Navigating the intersection: Diabetes and Alzheimer's intertwined relationship. Ageing Res Rev 2024; 100:102415. [PMID: 39002642 DOI: 10.1016/j.arr.2024.102415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/06/2024] [Accepted: 07/06/2024] [Indexed: 07/15/2024]
Abstract
Alzheimer's disease (AD) and Diabetes mellitus (DM) exhibit comparable pathophysiological pathways. Genetic abnormalities in APP, PS-1, and PS-2 are linked to AD, with diagnostic aid from CSF and blood biomarkers. Insulin dysfunction, termed "type 3 diabetes mellitus" in AD, involves altered insulin signalling and neuronal shrinkage. Insulin influences beta-amyloid metabolism, exacerbating neurotoxicity in AD and amyloid production in DM. Both disorders display impaired glucose transporter expression, hastening cognitive decline. Mitochondrial dysfunction and Toll-like receptor 4-mediated inflammation worsen neurodegeneration in both diseases. ApoE4 raises disease risk, especially when coupled with dyslipidemia common in DM. Targeting shared pathways like insulin-degrading enzyme activation and HSP60 holds promise for therapeutic intervention. Recognizing these interconnected mechanisms underscores the imperative for developing tailored treatments addressing the overlapping pathophysiology of AD and DM, offering potential avenues for more effective management of both conditions.
Collapse
Affiliation(s)
- Mayur B Kale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | | | - Nitu L Wankhede
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Prajwali V Shende
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Rushikesh P Thanekaer
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Manish M Aglawe
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Sandip R Rahangdale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Brijesh G Taksande
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Sunil B Pandit
- SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra, India
| | - Aman B Upaganlawar
- SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra, India
| | - Milind J Umekar
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Chungcheongbuk Do 27478, Republic of Korea.
| |
Collapse
|
44
|
Molina-Aguirre G, Chakraborty S, Košmrlj J, Vuković L, Pinter B. Photophysics of Molecular Probes for Amyloid-β Detection: Computational Insights into the Roles of Probe Linker and Functional Groups. J Phys Chem A 2024; 128:7055-7067. [PMID: 39146450 DOI: 10.1021/acs.jpca.4c01547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
In this computational study, density functional theory (DFT) and time-dependent DFT methods (TD-DFT) were employed to study the optical properties of six families of molecules with donor (D), bridge (B), and acceptor (A) fragments that have potential for use as fluorescent molecular probes for the early detection of Alzheimer's disease. After validating our computational method against experimental data, using X-ray and absorption data, the equilibrium geometries and wave functions of the ground and first singlet excited states were systematically studied. Our simulations demonstrate that the S1 states of these rod-like D-B-A fluorescent probes are twisted intramolecular charge transfer states with a predominant highest occupied molecular orbital-least unoccupied molecular orbital (HOMO-LUMO) character, the former localized primarily at the donor, whereas the latter at the acceptor site. Moreover, the influence of the bridge, donor, and acceptor fragments on molecules' absorption energies is explored, highlighting the influence of double and triple bonds and some specific modifications on the acceptor side, including the addition of electronegative atoms, pyranone derivatives, and their functionalization. By having the absorption energies of 324 probes in hand, machine learning models were trained to predict the absorption energies of molecules. The models were found to be predictive, which suggests a potential that predictive models for other crucial properties, such as emission and quantum yield, can also be trained if suitable training data sets are made available.
Collapse
Affiliation(s)
- Gabriela Molina-Aguirre
- Department of Chemistry and Biochemistry, The University of Texas at El Paso, El Paso, Texas 79968, United States
| | - Sayantani Chakraborty
- Department of Chemistry and Biochemistry, The University of Texas at El Paso, El Paso, Texas 79968, United States
| | - Janez Košmrlj
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia
| | - Lela Vuković
- Department of Chemistry and Biochemistry, The University of Texas at El Paso, El Paso, Texas 79968, United States
| | - Balazs Pinter
- Department of Chemistry and Biochemistry, The University of Texas at El Paso, El Paso, Texas 79968, United States
| |
Collapse
|
45
|
Hirakawa H, Terao T. The genetic association between bipolar disorder and dementia: a qualitative review. Front Psychiatry 2024; 15:1414776. [PMID: 39228919 PMCID: PMC11368786 DOI: 10.3389/fpsyt.2024.1414776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 08/05/2024] [Indexed: 09/05/2024] Open
Abstract
Bipolar disorder is a chronic disorder characterized by fluctuations in mood state and energy and recurrent episodes of mania/hypomania and depression. Bipolar disorder may be regarded as a neuro-progressive disorder in which repeated mood episodes may lead to cognitive decline and dementia development. In the current review, we employed genome-wide association studies to comprehensively investigate the genetic variants associated with bipolar disorder and dementia. Thirty-nine published manuscripts were identified: 20 on bipolar disorder and 19 on dementia. The results showed that the genes CACNA1C, GABBR2, SCN2A, CTSH, MSRA, and SH3PXD2A were overlapping between patients with bipolar disorder and dementia. In conclusion, the genes CACNA1C, GABBR2, SCN2A, CTSH, MSRA, and SH3PXD2A may be associated with the neuro-progression of bipolar disorder to dementia. Further genetic studies are needed to comprehensively clarify the role of genes in cognitive decline and the development of dementia in patients with bipolar disorder.
Collapse
Affiliation(s)
- Hirofumi Hirakawa
- Department of Neuropsychiatry, Oita University Faculty of Medicine, Yufu, Oita, Japan
| | | |
Collapse
|
46
|
Noroozi M, Gholami M, Sadeghsalehi H, Behzadi S, Habibzadeh A, Erabi G, Sadatmadani SF, Diyanati M, Rezaee A, Dianati M, Rasoulian P, Khani Siyah Rood Y, Ilati F, Hadavi SM, Arbab Mojeni F, Roostaie M, Deravi N. Machine and deep learning algorithms for classifying different types of dementia: A literature review. APPLIED NEUROPSYCHOLOGY. ADULT 2024:1-15. [PMID: 39087520 DOI: 10.1080/23279095.2024.2382823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
The cognitive impairment known as dementia affects millions of individuals throughout the globe. The use of machine learning (ML) and deep learning (DL) algorithms has shown great promise as a means of early identification and treatment of dementia. Dementias such as Alzheimer's Dementia, frontotemporal dementia, Lewy body dementia, and vascular dementia are all discussed in this article, along with a literature review on using ML algorithms in their diagnosis. Different ML algorithms, such as support vector machines, artificial neural networks, decision trees, and random forests, are compared and contrasted, along with their benefits and drawbacks. As discussed in this article, accurate ML models may be achieved by carefully considering feature selection and data preparation. We also discuss how ML algorithms can predict disease progression and patient responses to therapy. However, overreliance on ML and DL technologies should be avoided without further proof. It's important to note that these technologies are meant to assist in diagnosis but should not be used as the sole criteria for a final diagnosis. The research implies that ML algorithms may help increase the precision with which dementia is diagnosed, especially in its early stages. The efficacy of ML and DL algorithms in clinical contexts must be verified, and ethical issues around the use of personal data must be addressed, but this requires more study.
Collapse
Affiliation(s)
- Masoud Noroozi
- Department of Biomedical Engineering, Faculty of Engineering, University of Isfahan, Isfahan, Iran
| | - Mohammadreza Gholami
- Department of Electrical and Computer Engineering, Tarbiat Modares Univeristy, Tehran, Iran
| | - Hamidreza Sadeghsalehi
- Department of Artificial Intelligence in Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Saleh Behzadi
- Student Research Committee, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Adrina Habibzadeh
- Student Research Committee, Fasa University of Medical Sciences, Fasa, Iran
- USERN Office, Fasa University of Medical Sciences, Fasa, Iran
| | - Gisou Erabi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | | | - Mitra Diyanati
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, USA
| | - Aryan Rezaee
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Dianati
- Student Research Committee, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Pegah Rasoulian
- Sports Medicine Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Yashar Khani Siyah Rood
- Faculty of Engineering, Computer Engineering, Islamic Azad University of Bandar Abbas, Bandar Abbas, Iran
| | - Fatemeh Ilati
- Student Research Committee, Faculty of Medicine, Islamic Azad University of Mashhad, Mashhad, Iran
| | | | - Fariba Arbab Mojeni
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Minoo Roostaie
- School of Medicine, Islamic Azad University Tehran Medical Branch, Tehran, Iran
| | - Niloofar Deravi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
47
|
Chen L, Liu X, Zheng J, Li G, Yang B, He A, Liu H, Liang Y, Wang WA, Du J. A randomized, double-blind, placebo-controlled study of Cistanche tubulosa and Ginkgo biloba extracts for the improvement of cognitive function in middle-aged and elderly people. Phytother Res 2024; 38:4272-4285. [PMID: 38972848 DOI: 10.1002/ptr.8275] [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: 11/19/2023] [Revised: 02/28/2024] [Accepted: 06/07/2024] [Indexed: 07/09/2024]
Abstract
Mild cognitive impairment poses an increasing challenge to middle-aged and elderly populations. Traditional Chinese medicinal herbs like Cistanche tubulosa and Ginkgo biloba (CG) have been proposed as potential agents to improve cognitive and memory functions. A randomized controlled trial involving 100 Chinese middle-aged and elderly participants was conducted to investigate the potential synergistic effects of CG on cognitive function in individuals at risk of neurodegenerative diseases. Over 90 days, both CG group and placebo group received two tablets daily, with each pair of CG tablets containing 72 mg echinacoside and 27 mg flavonol glycosides. Cognitive functions were assessed using multiple scales and blood biomarkers were determined at baseline, Day 45, and Day 90. The CG group exhibited significant improvements in the scores of Mini-Mental State Examination (26.5 at baseline vs. 27.1 at Day 90, p < 0.001), Montreal Cognitive Assessment (23.4 at baseline vs. 25.3 at Day 90, p < 0.001), and World Health Organization Quality of Life (81.6 at baseline vs. 84.2 at Day 90, p < 0.001), all surpassing scores in placebo group. Notably, both the Cognitrax matrix test and the Wechsler Memory Scale-Revised demonstrated enhanced memory functions, including long-term and delayed memory, after CG intervention. Moreover, cognitive-related blood biomarkers, including total tau, pT181, pS199, pT231, pS396, and thyroid-stimulating hormone, significantly decreased, whereas triiodothyronine and free triiodothyronine significantly increased. No treatment-related adverse events were reported, and routine blood and urine tests remained stable. These findings indicated that CG supplementation could potentially serve as an effective supplementary solution for enhancing cognitive and memory functions.
Collapse
Affiliation(s)
- Liang Chen
- Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai, China
| | - Xin Liu
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianheng Zheng
- Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai, China
| | - Gang Li
- Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai, China
| | - Binrui Yang
- Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai, China
| | - Anli He
- Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai, China
| | - Hongyue Liu
- Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai, China
| | | | - Wen' An Wang
- Department of Neurology, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Du
- Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai, China
| |
Collapse
|
48
|
Emeršič A, Ashton NJ, Vrillon A, Lantero‐Rodriguez J, Mlakar J, Gregorič Kramberger M, Gonzalez‐Ortiz F, Kac PR, Dulewicz M, Hanrieder J, Vanmechelen E, Rot U, Zetterberg H, Karikari TK, Čučnik S, Blennow K. Cerebrospinal fluid p-tau181, 217, and 231 in definite Creutzfeldt-Jakob disease with and without concomitant pathologies. Alzheimers Dement 2024; 20:5324-5337. [PMID: 38924651 PMCID: PMC11350132 DOI: 10.1002/alz.13907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 06/28/2024]
Abstract
INTRODUCTION The established cerebrospinal fluid (CSF) phosphorylated tau181 (p-tau181) may not reliably reflect concomitant Alzheimer's disease (AD) and primary age-related tauopathy (PART) found in Creutzfeldt-Jakob disease (CJD) at autopsy. METHODS We investigated CSF N-terminal p-tau181, p-tau217, and p-tau231 with in-house Simoa assays in definite CJD (n = 29), AD dementia (n = 75), mild cognitive impairment (MCI) due to AD (n = 65), and subjective cognitive decline (SCD, n = 28). Post-mortem examination performed in patients with CJD 1.3 (0.3-14.3) months after CSF collection revealed no co-pathology in 10, concomitant AD in 8, PART in 8, and other co-pathologies in 3 patients. RESULTS N-terminal p-tau was increased in CJD versus SCD (p < 0.0001) and correlated with total tau (t-tau) in the presence of AD and PART co-pathology (rho = 0.758-0.952, p ≤ 001). Concentrations in CJD+AD were indistinguishable from AD dementia, with the largest fold-change in p-tau217 (11.6), followed by p-tau231 and p-tau181 (3.2-4.5). DISCUSSION Variable fold-changes and correlation with t-tau suggest that p-tau closely associates with neurodegeneration and concomitant AD in CJD. HIGHLIGHTS N-terminal phosphorylated tau (p-tau) biomarkers are increased in Creutzfeldt-Jakob disease (CJD) with and without concomitant AD. P-tau217, p-tau231, and p-tau181 correlate with total tau (t-tau) and increase in the presence of amyloid beta (Aβ) co-pathology. N-terminal p-tau181 and p-tau231 in Aβ-negative CJD show variation among PRNP genotypes. Compared to mid-region-targeting p-tau181, cerebrospinal fluid (CSF) N-terminal p-tau has greater potential to reflect post-mortem neuropathology in the CJD brain.
Collapse
Affiliation(s)
- Andreja Emeršič
- Department of NeurologyUniversity Medical Centre LjubljanaLjubljanaSlovenia
- Faculty of PharmacyUniversity of LjubljanaLjubljanaSlovenia
| | - Nicholas J. Ashton
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, The Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Wallenberg Centre for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
- King's College LondonInstitute of Psychiatry, Psychology & NeuroscienceMaurice Wohl Clinical Neuroscience InstituteLondonUK
- NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS FoundationLondonUK
| | - Agathe Vrillon
- Université de Paris Cognitive Neurology CenterGHU Nord APHP Hospital Lariboisière Fernand WidalParisFrance
- Université de Paris Inserm UMR S11‐44 Therapeutic Optimization in NeuropsychopharmacologyParisFrance
| | - Juan Lantero‐Rodriguez
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, The Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Jernej Mlakar
- Institute of PathologyFaculty of MedicineUniversity of LjubljanaLjubljanaSlovenia
| | - Milica Gregorič Kramberger
- Department of NeurologyUniversity Medical Centre LjubljanaLjubljanaSlovenia
- Faculty of MedicineUniversity of LjubljanaLjubljanaSlovenia
- Department of Neurobiology, Care Sciences and Society, Division of Clinical GeriatricsKarolinska InstitutetHuddingeSweden
| | - Fernando Gonzalez‐Ortiz
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, The Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - Przemysław R. Kac
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, The Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Maciej Dulewicz
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, The Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Jörg Hanrieder
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, The Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of Neurodegenerative DiseaseUCL Institute of Neurology, Queen SquareLondonUK
| | | | - Uroš Rot
- Department of NeurologyUniversity Medical Centre LjubljanaLjubljanaSlovenia
- Faculty of MedicineUniversity of LjubljanaLjubljanaSlovenia
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, The Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseUCL Institute of Neurology, Queen SquareLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Hong Kong Center for Neurodegenerative DiseasesHong KongChina
- School of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Thomas K. Karikari
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, The Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of PsychiatrySchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Saša Čučnik
- Department of NeurologyUniversity Medical Centre LjubljanaLjubljanaSlovenia
- Faculty of PharmacyUniversity of LjubljanaLjubljanaSlovenia
- Department of RheumatologyUniversity Medical Centre LjubljanaLjubljanaSlovenia
| | - Kaj Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, The Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Paris Brain Institute, ICM, Pitié‐Salpêtrière HospitalSorbonne UniversityParisFrance
- Neurodegenerative Disorder Research CenterDivision of Life Sciences and Medicineand Department of NeurologyInstitute on Aging and Brain DisordersUniversity of Science and Technology of China and First Affiliated Hospital of USTCHefeiP.R. China
| |
Collapse
|
49
|
Syk M, Tornvind E, Gallwitz M, Fällmar D, Amandusson Å, Rothkegel H, Danfors T, Thulin M, Rasmusson AJ, Cervenka S, Pollak TA, Endres D, van Elst LT, Bodén R, Nilsson BM, Nordmark G, Burman J, Cunningham JL. An exploratory study of the damage markers NfL, GFAP, and t-Tau, in cerebrospinal fluid and other findings from a patient cohort enriched for suspected autoimmune psychiatric disease. Transl Psychiatry 2024; 14:304. [PMID: 39048548 PMCID: PMC11269634 DOI: 10.1038/s41398-024-03021-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 06/27/2024] [Accepted: 07/10/2024] [Indexed: 07/27/2024] Open
Abstract
There is growing evidence suggesting that immunological mechanisms play a significant role in the development of psychiatric symptoms in certain patient subgroups. However, the relationship between clinical red flags for suspected autoimmune psychiatric disease and signs of central nervous system (CNS) pathology (e.g., routine cerebrospinal fluid (CSF) alterations, CNS damage markers, neurophysiological or neuroimaging findings) has received limited attention. Here, we aimed to describe the prevalence and distribution of potential CNS pathologies in psychiatric patients in relation to clinical red flags for autoimmune psychiatric disease and psychiatric symptoms. CSF routine findings and CNS damage markers; neurofilament light chain protein (NfL), glial fibrillary acidic protein (GFAP) and total Tau (t-Tau), in CSF from 127 patients with psychiatric disease preselected for suspected immunological involvement were related to recently proposed clinical red flags, psychiatric features, and MRI and EEG findings. Twenty-one percent had abnormal routine CSF findings and 27% had elevated levels of CNS damage markers. Six percent had anti-neuronal antibodies in serum and 2% had these antibodies in the CSF. Sixty-six percent of patients examined with MRI (n = 88) had alterations, mostly atrophy or nonspecific white matter lesions. Twenty-seven percent of patients with EEG recordings (n = 70) had abnormal findings. Elevated NfL levels were associated with comorbid autoimmunity and affective dysregulation symptoms. Elevated t-Tau was associated with catatonia and higher ratings of agitation/hyperactivity. Elevated GFAP was associated with acute onset, atypical presentation, infectious prodrome, tics, depressive/anxiety symptom ratings and overall greater psychiatric symptom burden. In conclusion, preselection based on suspected autoimmune psychiatric disease identifies a population with a high prevalence of CSF alterations suggesting CNS pathology. Future studies should examine the value of these markers in predicting treatment responses.
Collapse
Affiliation(s)
- Mikaela Syk
- Department of Medical Sciences, Psychiatry, Uppsala University, Uppsala, Sweden
| | - Emma Tornvind
- Department of Medical Sciences, Psychiatry, Uppsala University, Uppsala, Sweden
| | - Maike Gallwitz
- Department of Medical Sciences, Psychiatry, Uppsala University, Uppsala, Sweden
| | - David Fällmar
- Department of Surgical Sciences, Neuroradiology, Uppsala University, Uppsala, Sweden
| | - Åsa Amandusson
- Department of Medical Sciences, Clinical Neurophysiology, Uppsala University, Uppsala, Sweden
| | - Holger Rothkegel
- Department of Medical Sciences, Clinical Neurophysiology, Uppsala University, Uppsala, Sweden
| | - Torsten Danfors
- Department of Surgical Sciences, Radiology, Uppsala University, Uppsala, Sweden
| | - Måns Thulin
- Department of Mathematics, Uppsala University, Uppsala, Sweden
| | - Annica J Rasmusson
- Department of Medical Sciences, Psychiatry, Uppsala University, Uppsala, Sweden
| | - Simon Cervenka
- Department of Medical Sciences, Psychiatry, Uppsala University, Uppsala, Sweden
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institute & Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - Thomas A Pollak
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Dominique Endres
- Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ludger Tebartz van Elst
- Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Robert Bodén
- Department of Medical Sciences, Psychiatry, Uppsala University, Uppsala, Sweden
| | - Björn M Nilsson
- Department of Medical Sciences, Psychiatry, Uppsala University, Uppsala, Sweden
| | - Gunnel Nordmark
- Department of Medical Sciences, Rheumatology, Uppsala University, Uppsala, Sweden
| | - Joachim Burman
- Department of Medical Sciences, Neurology, Uppsala University, Uppsala, Sweden
| | - Janet L Cunningham
- Department of Medical Sciences, Psychiatry, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
50
|
Chae S, Yang E, Moon WJ, Kim JH. Deep Cascade of Convolutional Neural Networks for Quantification of Enlarged Perivascular Spaces in the Basal Ganglia in Magnetic Resonance Imaging. Diagnostics (Basel) 2024; 14:1504. [PMID: 39061641 PMCID: PMC11276133 DOI: 10.3390/diagnostics14141504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/07/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
In this paper, we present a cascaded deep convolution neural network (CNN) for assessing enlarged perivascular space (ePVS) within the basal ganglia region using T2-weighted MRI. Enlarged perivascular spaces (ePVSs) are potential biomarkers for various neurodegenerative disorders, including dementia and Parkinson's disease. Accurate assessment of ePVS is crucial for early diagnosis and monitoring disease progression. Our approach first utilizes an ePVS enhancement CNN to improve ePVS visibility and then employs a quantification CNN to predict the number of ePVSs. The ePVS enhancement CNN selectively enhances the ePVS areas without the need for additional heuristic parameters, achieving a higher contrast-to-noise ratio (CNR) of 113.77 compared to Tophat, Clahe, and Laplacian-based enhancement algorithms. The subsequent ePVS quantification CNN was trained and validated using fourfold cross-validation on a dataset of 76 participants. The quantification CNN attained 88% accuracy at the image level and 94% accuracy at the subject level. These results demonstrate significant improvements over traditional algorithm-based methods, highlighting the robustness and reliability of our deep learning approach. The proposed cascaded deep CNN model not only enhances the visibility of ePVS but also provides accurate quantification, making it a promising tool for evaluating neurodegenerative disorders. This method offers a novel and significant advancement in the non-invasive assessment of ePVS, potentially aiding in early diagnosis and targeted treatment strategies.
Collapse
Affiliation(s)
- Seunghye Chae
- Medical Research Institute, Samsung Medical Center, Seoul 06351, Republic of Korea;
| | - Ehwa Yang
- School of Medicine, Sungkyunkwan University, Seoul 06351, Republic of Korea
| | - Won-Jin Moon
- Department of Radiology, Konkuk University Medical Center, School of Medicine, Konkuk University, Seoul 05030, Republic of Korea
| | - Jae-Hun Kim
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| |
Collapse
|