1
|
Milos T, Vuic B, Balic N, Farkas V, Nedic Erjavec G, Svob Strac D, Nikolac Perkovic M, Pivac N. Cerebrospinal fluid in the differential diagnosis of Alzheimer's disease: an update of the literature. Expert Rev Neurother 2024; 24:1063-1079. [PMID: 39233323 DOI: 10.1080/14737175.2024.2400683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 09/01/2024] [Indexed: 09/06/2024]
Abstract
INTRODUCTION The importance of cerebrospinal fluid (CSF) biomarkers in Alzheimer's disease (AD) diagnosis is rapidly increasing, and there is a growing interest in the use of CSF biomarkers in monitoring the response to therapy, especially in the light of newly available approaches to the therapy of neurodegenerative diseases. AREAS COVERED In this review we discuss the most relevant measures of neurodegeneration that are being used to distinguish patients with AD from healthy controls and individuals with mild cognitive impairment, in order to provide an overview of the latest information available in the scientific literature. We focus on markers related to amyloid processing, markers associated with neurofibrillary tangles, neuroinflammation, neuroaxonal injury and degeneration, synaptic loss and dysfunction, and markers of α-synuclein pathology. EXPERT OPINION In addition to neuropsychological evaluation, core CSF biomarkers (Aβ42, t-tau, and p-tau181) have been recommended for improvement of timely, accurate and differential diagnosis of AD, as well as to assess the risk and rate of disease progression. In addition to the core CSF biomarkers, various other markers related to synaptic dysfunction, neuroinflammation, and glial activation (neurogranin, SNAP-25, Nfl, YKL-40, TREM2) are now investigated and have yet to be validated for future potential clinical use in AD diagnosis.
Collapse
Affiliation(s)
- Tina Milos
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia
| | - Barbara Vuic
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia
| | - Nikola Balic
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia
| | - Vladimir Farkas
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia
| | | | | | | | - Nela Pivac
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia
- University of Applied Sciences Hrvatsko Zagorje Krapina, Krapina, Croatia
| |
Collapse
|
2
|
Agnello L, Gambino CM, Ciaccio AM, Piccoli T, Blandino V, Scazzone C, Lo Sasso B, Del Ben F, Ciaccio M. Exploring the effect of APOE ε4 on biomarkers of neurodegeneration in Alzheimer's disease. Clin Chim Acta 2024; 562:119876. [PMID: 39025198 DOI: 10.1016/j.cca.2024.119876] [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: 06/26/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND AND AIMS This study aims to assess the association between APOE genotype and biomarkers of neurodegeneration in Alzheimer's disease (AD). METHODS We performed a retrospective observational study at the University Hospital "P. Giaccone" in Palermo, Italy. We enrolled patients with cognitive decline, including AD. For each patient, we measured amyloid beta (Aβ)42, Aβ40, tau protein phosphorylated at threonine 181 (pTau), total tau (tTau), neurogranin, alpha-synuclein, and neurofilament light chain (NfL) in cerebrospinal fluid (CSF). RESULTS The study population consisted of 194 patients (123 AD and 71 non-AD). AD patients have significantly lower Aβ42 levels and Aβ42/40 ratio and higher pTau, tTau, and NfLs levels than non-AD patients. In AD patients, the APOEε4 allele is associated with a significantly lower Aβ42/40 ratio and higher levels of pTau, tTau, neurogranin, and alpha-synuclein. This association is not observed in non-AD patients. CONCLUSIONS This study provides evidence of the significant impact of the APOE ε4 allele on neurodegenerative biomarkers in AD patients, highlighting its role in exacerbating amyloid and tau pathology as well as synaptic degeneration.
Collapse
Affiliation(s)
- Luisa Agnello
- Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University of Palermo, Palermo, Italy
| | - Caterina Maria Gambino
- Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University of Palermo, Palermo, Italy; Department of Laboratory Medicine, University Hospital "P. Giaccone", Palermo, Italy
| | - Anna Maria Ciaccio
- Internal Medicine and Medical Specialties "G. D'Alessandro", Department of Health Promotion, Maternal and Infant Care, University of Palermo, Palermo, Italy
| | - Tommaso Piccoli
- Unit of Neurology, Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University of Palermo, Palermo, Italy
| | - Valeria Blandino
- Unit of Neurology, Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University of Palermo, Palermo, Italy
| | - Concetta Scazzone
- Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University of Palermo, Palermo, Italy
| | - Bruna Lo Sasso
- Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University of Palermo, Palermo, Italy; Department of Laboratory Medicine, University Hospital "P. Giaccone", Palermo, Italy
| | - Fabio Del Ben
- Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO)-IRCCS, Aviano, Italy
| | - Marcello Ciaccio
- Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University of Palermo, Palermo, Italy; Department of Laboratory Medicine, University Hospital "P. Giaccone", Palermo, Italy.
| |
Collapse
|
3
|
Li M, Shen T, Yao R, Sun H, Liu X, Li Z, Zhang J. Mitochondrial dysfunction is associated with cognitive impairment in adults with OSA without dementia. Sleep Med 2024; 119:234-243. [PMID: 38704871 DOI: 10.1016/j.sleep.2024.04.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 04/24/2024] [Indexed: 05/07/2024]
Abstract
STUDY OBJECTIVES Increased reactive oxygen species associated with loss of mitochondrial function affect synaptic activity, which is an important mechanism underlying cognitive decline. This study assesses the role of mitochondrial proteins in neuron-derived exosomes (NDEs) on cognitive impairment in patients with obstructive sleep apnea (OSA) without dementia. METHODS Analyses were conducted in 268 study participants with complete polysomnography data, cognitive tests, and important clinical data available. NDEs were isolated immunochemically for enzyme-linked immunosorbent assay quantification of mitochondrial proteins, i.e., humanin and mitochondrial open reading frame of the 12S rRNA-c (MOTS-c), and synaptic protein, i.e., neurogranin (NRGN). A mediation analysis of the relationship between sleep parameters and cognition was performed using humanin, MOTS-c, and NRGN values as a mediating factor. Twenty-two patients with moderate to severe OSA who received CPAP therapy were followed up, and humanin, MOTS-c and NRGN levels were reassessed after 1 year of treatment. RESULTS All participants were divided into the OSA + MCI group (n = 91), OSA-MCI group (n = 89), MCI group (MCI without OSA) (n = 38) and control group (normal cognitive state without OSA) (n = 50). The mean CD63-normalized NDE levels of humanin, MOTS-c, and NRGN in the OSA + MCI group were higher than those in the OSA-MCI and control groups. The NDE levels of humanin, MOTS-c, and NRGN in the MCI group were lower than those in controls. The odds of cognitive impairment in patients with OSA were higher with higher NDE levels of humanin, MOTS-c, and NRGN (odds ratio (OR): 2.100, 95 % confidence interval (CI): 1.646-2.679, P < 0.001; OR: 5.453, 95 % CI: 3.112-9.556, P < 0.001; OR: 3.115, 95 % CI: 2.163-4.484, P < 0.001). The impaired cognitive performance was associated with higher NDE levels of humanin (β: 0.505, SE: 0.048, P < 0.001), MOTS-c (β: 0.580, SE: 0.001, P < 0.001), and NRGN (β: 0.585, SE: 0.553, P < 0.001). The relationship between sleep parameters (mean SaO2 and T90) and MoCA scores was mediated by the NDE levels of humanin, MOTS-c, and NRGN with the proportion of mediation varying from 35.33 % to 149.07 %. Receiver operating characteristic curve revealed an area under the curve of 0.905 for humanin, 0.873 for MOTS-c, and 0.934 for NRGN to predict MCI in OSA patients without dementia. Increased humanin, MOTS-c, and NRGN levels significantly decreased after CPAP treatment. CONCLUSIONS Mitochondrial dysfunction is implicated in cognitive impairment in OSA patients without dementia, and mainly mediates the association between intermittent hypoxia and cognitive impairment in adults with OSA without dementia. Mitochondrial dysfunction can be partially reversible by CPAP treatment. Mitochondrial proteins can be used as markers of cognitive impairment in patients with OSA.
Collapse
Affiliation(s)
- Mengfan Li
- Department of Neurology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, China
| | - Tengqun Shen
- Department of Resident Standardized Training Management, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, China
| | - Ran Yao
- Department of Neurology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, China
| | - Hairong Sun
- Department of Neurology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, China
| | - Xiaoxiao Liu
- Department of Neurology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, China
| | - Zhenguang Li
- Department of Neurology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, China
| | - Jinbiao Zhang
- Department of Neurology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, China.
| |
Collapse
|
4
|
Kızılay T, Akbayir E, Erol R, Demir AS, Özkan Yaşargün D, Yilmaz V, Tuzun E, Turkoglu R. Altered Cerebrospinal Fluid Neurofilament Light Chain but Not Neurogranin Levels Are Associated with Response to Ocrelizumab Treatment in Relapsing-Remitting Multiple Sclerosis: A Preliminary Study. Eur Neurol 2024; 87:203-210. [PMID: 38754397 DOI: 10.1159/000539376] [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: 12/19/2023] [Accepted: 05/08/2024] [Indexed: 05/18/2024]
Abstract
INTRODUCTION Ocrelizumab is a CD20-targeting monoclonal antibody used for treatment of multiple sclerosis (MS). Serum and cerebrospinal fluid (CSF) neurofilament light (NFL) chain levels are reduced in MS patients under ocrelizumab treatment indicating a preventive action against neuro-axonal degeneration. Our aim, in this preliminary study, was to explore the impact of ocrelizumab treatment on synaptic integrity through assessment of neurogranin levels. METHODS Thirteen relapsing-remitting multiple sclerosis (RRMS) patients resistant to first-line immunomodulating agents were enrolled and followed up for 24 months under ocrelizumab treatment. Disease activity was monitored by periodic EDSS, MSSS, and cranial-spinal MRI assessments. No evidence of disease activity (NEDA)-3 was determined, and CSF levels of NFL (marker of neuro-axonal integrity) and neurogranin (marker of synaptic integrity) were measured by ELISA at baseline and 12-month ocrelizumab treatment. RESULTS Seven RRMS patients, who preserved NEDA-3 status during 24-month follow-up, showed ≥30% NFL level decrease, whereas 6 patients with stable/increased NFL levels displayed relapse, MRI lesion, or disability progression. Although most RRMS patients exhibited increased CSF levels of neurogranin under ocrelizumab treatment, patients with and without neurogranin level increase did not differ in terms of clinical features and NEDA-3 status. Baseline neurogranin levels negatively correlated with baseline EDSS scores. CONCLUSION Our results confirm that NFL effectively monitors treatment response of RRMS patients under ocrelizumab treatment. Neurogranin does not appear to exhibit a similar benefit in screening of RRMS disease activity. Nevertheless, lower neurogranin levels are associated with increased disability in RRMS indicating a potential disease activity biomarker function.
Collapse
Affiliation(s)
- Tuğçe Kızılay
- Neurology Clinic, Haydarpasa Numune Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Ece Akbayir
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Ruziye Erol
- Neurology Clinic, Haydarpasa Numune Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Ayça Simay Demir
- Neurology Clinic, Haydarpasa Numune Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Duygu Özkan Yaşargün
- Neurology Clinic, Haydarpasa Numune Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Vuslat Yilmaz
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Erdem Tuzun
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Recai Turkoglu
- Neurology Clinic, Haydarpasa Numune Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| |
Collapse
|
5
|
Wagemann O, Liu H, Wang G, Shi X, Bittner T, Scelsi MA, Farlow MR, Clifford DB, Supnet-Bell C, Santacruz AM, Aschenbrenner AJ, Hassenstab JJ, Benzinger TLS, Gordon BA, Coalier KA, Cruchaga C, Ibanez L, Perrin RJ, Xiong C, Li Y, Morris JC, Lah JJ, Berman SB, Roberson ED, van Dyck CH, Galasko D, Gauthier S, Hsiung GYR, Brooks WS, Pariente J, Mummery CJ, Day GS, Ringman JM, Mendez PC, St. George-Hyslop P, Fox NC, Suzuki K, Okhravi HR, Chhatwal J, Levin J, Jucker M, Sims JR, Holdridge KC, Proctor NK, Yaari R, Andersen SW, Mancini M, Llibre-Guerra J, Bateman RJ, McDade E. Downstream Biomarker Effects of Gantenerumab or Solanezumab in Dominantly Inherited Alzheimer Disease: The DIAN-TU-001 Randomized Clinical Trial. JAMA Neurol 2024; 81:582-593. [PMID: 38683602 PMCID: PMC11059071 DOI: 10.1001/jamaneurol.2024.0991] [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: 10/18/2023] [Accepted: 02/01/2024] [Indexed: 05/01/2024]
Abstract
Importance Effects of antiamyloid agents, targeting either fibrillar or soluble monomeric amyloid peptides, on downstream biomarkers in cerebrospinal fluid (CSF) and plasma are largely unknown in dominantly inherited Alzheimer disease (DIAD). Objective To investigate longitudinal biomarker changes of synaptic dysfunction, neuroinflammation, and neurodegeneration in individuals with DIAD who are receiving antiamyloid treatment. Design, Setting, and Participants From 2012 to 2019, the Dominantly Inherited Alzheimer Network Trial Unit (DIAN-TU-001) study, a double-blind, placebo-controlled, randomized clinical trial, investigated gantenerumab and solanezumab in DIAD. Carriers of gene variants were assigned 3:1 to either drug or placebo. The present analysis was conducted from April to June 2023. DIAN-TU-001 spans 25 study sites in 7 countries. Biofluids and neuroimaging from carriers of DIAD gene variants in the gantenerumab, solanezumab, and placebo groups were analyzed. Interventions In 2016, initial dosing of gantenerumab, 225 mg (subcutaneously every 4 weeks) was increased every 8 weeks up to 1200 mg. In 2017, initial dosing of solanezumab, 400 mg (intravenously every 4 weeks) was increased up to 1600 mg every 4 weeks. Main Outcomes and Measures Longitudinal changes in CSF levels of neurogranin, soluble triggering receptor expressed on myeloid cells 2 (sTREM2), chitinase 3-like 1 protein (YKL-40), glial fibrillary acidic protein (GFAP), neurofilament light protein (NfL), and plasma levels of GFAP and NfL. Results Of 236 eligible participants screened, 43 were excluded. A total of 142 participants (mean [SD] age, 44 [10] years; 72 female [51%]) were included in the study (gantenerumab, 52 [37%]; solanezumab, 50 [35%]; placebo, 40 [28%]). Relative to placebo, gantenerumab significantly reduced CSF neurogranin level at year 4 (mean [SD] β = -242.43 [48.04] pg/mL; P < .001); reduced plasma GFAP level at year 1 (mean [SD] β = -0.02 [0.01] ng/mL; P = .02), year 2 (mean [SD] β = -0.03 [0.01] ng/mL; P = .002), and year 4 (mean [SD] β = -0.06 [0.02] ng/mL; P < .001); and increased CSF sTREM2 level at year 2 (mean [SD] β = 1.12 [0.43] ng/mL; P = .01) and year 4 (mean [SD] β = 1.06 [0.52] ng/mL; P = .04). Solanezumab significantly increased CSF NfL (log) at year 4 (mean [SD] β = 0.14 [0.06]; P = .02). Correlation analysis for rates of change found stronger correlations between CSF markers and fluid markers with Pittsburgh compound B positron emission tomography for solanezumab and placebo. Conclusions and Relevance This randomized clinical trial supports the importance of fibrillar amyloid reduction in multiple AD-related processes of neuroinflammation and neurodegeneration in CSF and plasma in DIAD. Additional studies of antiaggregated amyloid therapies in sporadic AD and DIAD are needed to determine the utility of nonamyloid biomarkers in determining disease modification. Trial Registration ClinicalTrials.gov Identifier: NCT04623242.
Collapse
Affiliation(s)
- Olivia Wagemann
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Haiyan Liu
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| | - Guoqiao Wang
- Department of Biostatistics, Washington University in St Louis, St Louis, Missouri
| | - Xinyu Shi
- Department of Biostatistics, Washington University in St Louis, St Louis, Missouri
| | | | - Marzia A. Scelsi
- F. Hoffmann-La Roche Products Ltd, Welwyn Garden City, United Kingdom
| | - Martin R. Farlow
- Department of Neurology, Indiana University School of Medicine, Indianapolis
| | - David B. Clifford
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| | - Charlene Supnet-Bell
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| | - Anna M. Santacruz
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| | | | - Jason J. Hassenstab
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| | | | - Brian A. Gordon
- Department of Radiology, Washington University in St Louis, St Louis, Missouri
| | | | - Carlos Cruchaga
- Department of Psychiatry, Washington University in St Louis, St Louis, Missouri
| | - Laura Ibanez
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri
- Department of Psychiatry, Washington University in St Louis, St Louis, Missouri
| | - Richard J. Perrin
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri
- Department of Pathology and Immunology, Washington University in St Louis, St Louis, Missouri
| | - Chengjie Xiong
- Department of Biostatistics, Washington University in St Louis, St Louis, Missouri
| | - Yan Li
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| | - John C. Morris
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| | - James J. Lah
- Department of Neurology, School of Medicine Emory University, Atlanta, Georgia
| | - Sarah B. Berman
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Erik D. Roberson
- Department of Neurology, University of Alabama at Birmingham, Birmingham
| | | | - Douglas Galasko
- Department of Neurology, University of California, San Diego
| | - Serge Gauthier
- Department of Neurology & Psychiatry, McGill University, Montréal, Québec, Canada
| | - Ging-Yuek R. Hsiung
- Department of Neurology, University of British Columbia, Vancouver, British Columbia, Canada
| | - William S. Brooks
- Neuroscience Research Australia, Sydney, New South Wales, Australia
- School of Clinical Medicine, University of New South Wales, Randwick, New South Wales, Australia
| | - Jérémie Pariente
- Department of Neurology, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Catherine J. Mummery
- Dementia Research Centre, Institute of Neurology, University College London, London, United Kingdom
| | - Gregory S. Day
- Department of Neurology, Mayo Clinic Florida, Jacksonville
| | - John M. Ringman
- Department of Neurology, University of Southern California, Los Angeles
| | - Patricio Chrem Mendez
- Fundación Para la Lucha Contra las Enfermedades Neurológicas de la Infancia (FLENI), Buenos Aires, Argentina
| | | | - Nick C. Fox
- Dementia Research Centre, Institute of Neurology, University College London, London, United Kingdom
| | | | - Hamid R. Okhravi
- Department of Geriatrics, Eastern Virginia Medical School, Norfolk
| | - Jasmeer Chhatwal
- Department of Neurology, Massachusetts General and Brigham & Women’s Hospitals, Harvard Medical School, Boston
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Mathias Jucker
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | | | | | | | - Roy Yaari
- Eli Lilly and Company, Indianapolis, Indiana
| | | | | | - Jorge Llibre-Guerra
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| | - Randall J. Bateman
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| | - Eric McDade
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| |
Collapse
|
6
|
Gobom J, Brinkmalm A, Brinkmalm G, Blennow K, Zetterberg H. Alzheimer's Disease Biomarker Analysis Using Targeted Mass Spectrometry. Mol Cell Proteomics 2024; 23:100721. [PMID: 38246483 PMCID: PMC10926085 DOI: 10.1016/j.mcpro.2024.100721] [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: 11/14/2023] [Revised: 12/30/2023] [Accepted: 01/04/2024] [Indexed: 01/23/2024] Open
Abstract
Alzheimer's disease (AD) is characterized by several neuropathological changes, mainly extracellular amyloid aggregates (plaques), intraneuronal inclusions of phosphorylated tau (tangles), as well as neuronal and synaptic degeneration, accompanied by tissue reactions to these processes (astrocytosis and microglial activation) that precede neuronal network disturbances in the symptomatic phase of the disease. A number of biomarkers for these brain tissue changes have been developed, mainly using immunoassays. In this review, we discuss how targeted mass spectrometry (TMS) can be used to validate and further characterize classes of biomarkers reflecting different AD pathologies, such as tau- and amyloid-beta pathologies, synaptic dysfunction, lysosomal dysregulation, and axonal damage, and the prospect of using TMS to measure these proteins in clinical research and diagnosis. TMS advantages and disadvantages in relation to immunoassays are discussed, and complementary aspects of the technologies are discussed.
Collapse
Affiliation(s)
- Johan Gobom
- 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.
| | - Ann Brinkmalm
- 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
| | - Gunnar Brinkmalm
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK; UK Dementia Research Institute at UCL, 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, Wisconsin, USA.
| |
Collapse
|
7
|
Bivona G, Iemmolo M, Ghersi G. Cerebrospinal and Blood Biomarkers in Alzheimer's Disease: Did Mild Cognitive Impairment Definition Affect Their Clinical Usefulness? Int J Mol Sci 2023; 24:16908. [PMID: 38069230 PMCID: PMC10706963 DOI: 10.3390/ijms242316908] [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: 11/06/2023] [Revised: 11/26/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Despite Alzheimer's Disease (AD) being known from the times of Alois Alzheimer, who lived more than one century ago, many aspects of the disease are still obscure, including the pathogenesis, the clinical spectrum definition, and the therapeutic approach. Well-established biomarkers for AD come from the histopathological hallmarks of the disease, which are Aβ and phosphorylated Tau protein aggregates. Consistently, cerebrospinal fluid (CSF) Amyloid β (Aβ) and phosphorylated Tau level measurements are currently used to detect AD presence. However, two central biases affect these biomarkers. Firstly, incomplete knowledge of the pathogenesis of diseases legitimates the search for novel molecules that, reasonably, could be expressed by neurons and microglia and could be detected in blood simpler and earlier than the classical markers and in a higher amount. Further, studies have been performed to evaluate whether CSF biomarkers can predict AD onset in Mild Cognitive Impairment (MCI) patients. However, the MCI definition has changed over time. Hence, the studies on MCI patients seem to be biased at the beginning due to the imprecise enrollment and heterogeneous composition of the miscellaneous MCI subgroup. Plasma biomarkers and novel candidate molecules, such as microglia biomarkers, have been tentatively investigated and could represent valuable targets for diagnosing and monitoring AD. Also, novel AD markers are urgently needed to identify molecular targets for treatment strategies. This review article summarizes the main CSF and blood AD biomarkers, underpins their advantages and flaws, and mentions novel molecules that can be used as potential biomarkers for AD.
Collapse
Affiliation(s)
- Giulia Bivona
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy
| | - Matilda Iemmolo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy
| | - Giulio Ghersi
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy
| |
Collapse
|
8
|
Morrow CB, Leoutsakos J, Yan H, Onyike C, Kamath V. Weight Change and Neuropsychiatric Symptoms in Alzheimer's Disease and Frontotemporal Dementia: Associations with Cognitive Decline. J Alzheimers Dis Rep 2023; 7:767-774. [PMID: 37662607 PMCID: PMC10473120 DOI: 10.3233/adr-230034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 06/29/2023] [Indexed: 09/05/2023] Open
Abstract
Weight changes, neuropsychiatric symptoms (NPS), and cognitive decline often coincide in Alzheimer's disease (AD) and frontotemporal dementia (FTD); however, the direction of their relationship remains unclear. This study aims to clarify the connection between weight changes, NPS, and cognition in AD and FTD. We found that cognitive decline was associated with decreased body mass index (BMI) in AD, while BMI gain was associated with increased conversion to FTD. Elevated NPS were associated with decreased BMI in AD and increased BMI in FTD. Identifying early changes in NPS and BMI may facilitate the detection of cognitive decline, providing an opportunity for early intervention.
Collapse
Affiliation(s)
- Christopher B. Morrow
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jeannie Leoutsakos
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Haijuan Yan
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Chiadi Onyike
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Vidyulata Kamath
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
9
|
Advanced Overview of Biomarkers and Techniques for Early Diagnosis of Alzheimer's Disease. Cell Mol Neurobiol 2023:10.1007/s10571-023-01330-y. [PMID: 36847930 DOI: 10.1007/s10571-023-01330-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 02/15/2023] [Indexed: 03/01/2023]
Abstract
The development of early non-invasive diagnosis methods and identification of novel biomarkers are necessary for managing Alzheimer's disease (AD) and facilitating effective prognosis and treatment. AD has multi-factorial nature and involves complex molecular mechanism, which causes neuronal degeneration. The primary challenges in early AD detection include patient heterogeneity and lack of precise diagnosis at the preclinical stage. Several cerebrospinal fluid (CSF) and blood biomarkers have been proposed to show excellent diagnosis ability by identifying tau pathology and cerebral amyloid beta (Aβ) for AD. Intense research endeavors are being made to develop ultrasensitive detection techniques and find potent biomarkers for early AD diagnosis. To mitigate AD worldwide, understanding various CSF biomarkers, blood biomarkers, and techniques that can be used for early diagnosis is imperative. This review attempts to provide information regarding AD pathophysiology, genetic and non-genetic factors associated with AD, several potential blood and CSF biomarkers, like neurofilament light, neurogranin, Aβ, and tau, along with biomarkers under development for AD detection. Besides, numerous techniques, such as neuroimaging, spectroscopic techniques, biosensors, and neuroproteomics, which are being explored to aid early AD detection, have been discussed. The insights thus gained would help in finding potential biomarkers and suitable techniques for the accurate diagnosis of early AD before cognitive dysfunction.
Collapse
|
10
|
Feng RC, Dong YH, Hong XL, Su Y, Wu XV. Effects of anthocyanin-rich supplementation on cognition of the cognitively healthy middle-aged and older adults: a systematic review and meta-analysis of randomized controlled trials. Nutr Rev 2023; 81:287-303. [PMID: 35960187 DOI: 10.1093/nutrit/nuac055] [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: 11/13/2022] Open
Abstract
CONTEXT The prevalence of age-related cognitive decline has been on the rise as the global population age, putting the independence and quality of life of elderly at risk. Anthocyanin, as a subclass of dietary flavonoids, may have a beneficial impact on cognitive outcomes. OBJECTIVES To examine the effects of dietary anthocyanin supplementation on cognition of the cognitively healthy middle-aged and older adults. DATA SOURCES PubMed, ScienceDirect, CINAHL, EMBASE, ProQuest and Cochrane databases were searched. DATA EXTRACTION AND ANALYSIS Thirteen studies were included in this meta-analysis. Anthocyanin-rich supplementation was found to significantly improve the processing speed of the older adults (95%CI 0.08, 0.44; P = 0.004). No significant differences were observed between intervention and control groups on memory, attention, executive function and psychomotor performance. Current neuroimaging studies have found promising effects of anthocyanin supplementation on brain activation and cerebral perfusion. CONCLUSION Anthocyanin-rich supplementation may preserve cognitive processing speed and neuro-activities in older adults, which improves their daily functioning and quality of life. This review provides useful insights to guide direction and methodological designs for future studies to explore the underlying mechanisms of anthocyanins. SYSTEMATIC REVIEW AND META-ANALYSIS REGISTRATION PROSPERO registration No. CRD42021228007.
Collapse
Affiliation(s)
- Ruo Chen Feng
- is with the High-Dependency Unit, Tan Tock Seng Hospital, National Health Group, Singapore
| | - Yan Hong Dong
- are with the Alice Lee Centre for Nursing Studies, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,is with the Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Xian Li Hong
- is with the Medical Intensive Care Unit, Tan Tock Seng Hospital, National Health Group, Singapore
| | - Ya Su
- is with the Shanghai Jiao Tong University, School of Nursing, Shanghai, China.,is with the Faculty of Health Sciences, Hokkaido University, Japan
| | - Xi Vivien Wu
- are with the Alice Lee Centre for Nursing Studies, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,is with the NUSMED Healthy Longevity Translational Research Programme, National University of Singapore, Singapore
| |
Collapse
|
11
|
Stecker M. A Perspective: Challenges in Dementia Research. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:1368. [PMID: 36295529 PMCID: PMC9609997 DOI: 10.3390/medicina58101368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/21/2022] [Accepted: 09/27/2022] [Indexed: 11/17/2022]
Abstract
Although dementia is a common and devastating disease that has been studied intensely for more than 100 years, no effective disease modifying treatment has been found. At this impasse, new approaches are important. The purpose of this paper is to provide, in the context of current research, one clinician's perspective regarding important challenges in the field in the form of specific challenges. These challenges not only illustrate the scope of the problems inherent in finding treatments for dementia, but can also be specific targets to foster discussion, criticism and new research. One common theme is the need to transform research activities from small projects in individual laboratories/clinics to larger multinational projects, in which each clinician and researcher works as an integral part. This transformation will require collaboration between researchers, large corporations, regulatory/governmental authorities and the general population, as well as significant financial investments. However, the costs of transforming the approach are small in comparison with the cost of dementia.
Collapse
Affiliation(s)
- Mark Stecker
- Fresno Institute of Neuroscience, Fresno, CA 93720, USA
| |
Collapse
|
12
|
Fong TG, Inouye SK. The inter-relationship between delirium and dementia: the importance of delirium prevention. Nat Rev Neurol 2022; 18:579-596. [PMID: 36028563 PMCID: PMC9415264 DOI: 10.1038/s41582-022-00698-7] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2022] [Indexed: 12/30/2022]
Abstract
Delirium and dementia are two frequent causes of cognitive impairment among older adults and have a distinct, complex and interconnected relationship. Delirium is an acute confusional state characterized by inattention, cognitive dysfunction and an altered level of consciousness, whereas dementia is an insidious, chronic and progressive loss of a previously acquired cognitive ability. People with dementia have a higher risk of developing delirium than the general population, and the occurrence of delirium is an independent risk factor for subsequent development of dementia. Furthermore, delirium in individuals with dementia can accelerate the trajectory of the underlying cognitive decline. Delirium prevention strategies can reduce the incidence of delirium and associated adverse outcomes, including falls and functional decline. Therefore, delirium might represent a modifiable risk factor for dementia, and interventions that prevent or minimize delirium might also reduce or prevent long-term cognitive impairment. Additionally, understanding the pathophysiology of delirium and the connection between delirium and dementia might ultimately lead to additional treatments for both conditions. In this Review, we explore mechanisms that might be common to both delirium and dementia by reviewing evidence on shared biomarkers, and we discuss the importance of delirium recognition and prevention in people with dementia. In this Review, Fong and Inouye explore mechanisms that might be common to both delirium and dementia. They present delirium as a possible modifiable risk factor for dementia and discuss the importance of delirium prevention strategies in reducing this risk. Delirium and dementia are frequent causes of cognitive impairment among older adults and have a distinct, complex and interconnected relationship. Delirium prevention strategies have been shown to reduce not only the incidence of delirium but also the incidence of adverse outcomes associated with delirium such as falls and functional decline. Adverse outcomes associated with delirium, such as the onset of dementia symptoms in individuals with preclinical dementia, and/or the acceleration of cognitive decline in individuals with dementia might also be delayed by the implementation of delirium prevention strategies. Evidence regarding the association of systemic inflammatory and neuroinflammatory biomarkers with delirium is variable, possibly as a result of co-occurring dementia pathology or disruption of the blood–brain barrier. Alzheimer disease pathology, even prior to the onset of symptoms, might have an effect on delirium risk, with potential mechanisms including neuroinflammation and gene–protein interactions with the APOE ε4 allele. Novel strategies, including proteomics, multi-omics, neuroimaging, transcranial magnetic stimulation and EEG, are beginning to reveal how changes in cerebral blood flow, spectral power and connectivity can be associated with delirium; further work is needed to expand these findings to patients with delirium superimposed upon dementia.
Collapse
Affiliation(s)
- Tamara G Fong
- Aging Brain Center, Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA, USA. .,Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
| | - Sharon K Inouye
- Aging Brain Center, Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA, USA.,Division of Gerontology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
13
|
Metabolites and Biomarker Compounds of Neurodegenerative Diseases in Cerebrospinal Fluid. Metabolites 2022; 12:metabo12040343. [PMID: 35448530 PMCID: PMC9031591 DOI: 10.3390/metabo12040343] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/29/2022] [Accepted: 04/11/2022] [Indexed: 12/25/2022] Open
Abstract
Despite recent advances in diagnostic procedures for neurological disorders, it is still difficult to definitively diagnose some neurodegenerative diseases without neuropathological examination of autopsied brain tissue. As pathological processes in the brain are frequently reflected in the components of cerebrospinal fluid (CSF), CSF samples are sometimes useful for diagnosis. After CSF is secreted from the choroid plexus epithelial cells in the ventricles, some flows in the brain, some is mixed with intracerebral interstitial fluid, and some is excreted through two major drainage pathways, i.e., the intravascular periarterial drainage pathway and the glymphatic system. Accordingly, substances produced by metabolic and pathological processes in the brain may be detectable in CSF. Many papers have reported changes in the concentration of substances in the CSF of patients with metabolic and neurological disorders, some of which can be useful biomarkers of the disorders. In this paper, we show the significance of glucose- and neurotransmitter-related CSF metabolites, considering their transporters in the choroid plexus; summarize the reported candidates of CSF biomarkers for neurodegenerative diseases, including amyloid-β, tau, α-synuclein, microRNAs, and mitochondrial DNA; and evaluate their potential as efficient diagnostic tools.
Collapse
|
14
|
Zhang Y, Wu KM, Yang L, Dong Q, Yu JT. Tauopathies: new perspectives and challenges. Mol Neurodegener 2022; 17:28. [PMID: 35392986 PMCID: PMC8991707 DOI: 10.1186/s13024-022-00533-z] [Citation(s) in RCA: 124] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 03/23/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Tauopathies are a class of neurodegenerative disorders characterized by neuronal and/or glial tau-positive inclusions. MAIN BODY Clinically, tauopathies can present with a range of phenotypes that include cognitive/behavioral-disorders, movement disorders, language disorders and non-specific amnestic symptoms in advanced age. Pathologically, tauopathies can be classified based on the predominant tau isoforms that are present in the inclusion bodies (i.e., 3R, 4R or equal 3R:4R ratio). Imaging, cerebrospinal fluid (CSF) and blood-based tau biomarkers have the potential to be used as a routine diagnostic strategy and in the evaluation of patients with tauopathies. As tauopathies are strongly linked neuropathologically and genetically to tau protein abnormalities, there is a growing interest in pursuing of tau-directed therapeutics for the disorders. Here we synthesize emerging lessons on tauopathies from clinical, pathological, genetic, and experimental studies toward a unified concept of these disorders that may accelerate the therapeutics. CONCLUSIONS Since tauopathies are still untreatable diseases, efforts have been made to depict clinical and pathological characteristics, identify biomarkers, elucidate underlying pathogenesis to achieve early diagnosis and develop disease-modifying therapies.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, 12th Wulumuqi Zhong Road, Shanghai, 200040 China
| | - Kai-Min Wu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, 12th Wulumuqi Zhong Road, Shanghai, 200040 China
| | - Liu Yang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, 12th Wulumuqi Zhong Road, Shanghai, 200040 China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, 12th Wulumuqi Zhong Road, Shanghai, 200040 China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, 12th Wulumuqi Zhong Road, Shanghai, 200040 China
| |
Collapse
|
15
|
Roveta F, Cermelli A, Boschi S, Ferrandes F, Grassini A, Marcinnò A, Spina M, Rubino E, Borsello T, Vercelli A, Rainero I. Synaptic Proteins as Fluid Biomarkers in Alzheimer's Disease: A Systematic Review and Meta-Analysis. J Alzheimers Dis 2022; 90:1381-1393. [PMID: 36278349 DOI: 10.3233/jad-220515] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Synaptic disruption precedes neuronal death and correlates with clinical features of Alzheimer's disease (AD). The identification of fluid biomarkers of synaptic damage is emerging as a goal for early and accurate diagnosis of the disease. OBJECTIVE To perform a systematic review and meta-analysis to determine whether fluid biomarkers of synaptic damage are impaired in AD. METHODS PubMed, Scopus, EMBASE, and Web of Science were searched for articles reporting synaptic proteins as fluid biomarkers in AD and cognitively unimpaired (CU) individuals. Pooled effect sizes were determined using the Hedge G method with random effects. Questions adapted from the Quality Assessment of Diagnostic Accuracy Studies were applied for quality assessment. A protocol for this study has been previously registered in PROSPERO (registration number: CRD42021277487). RESULTS The search strategy identified 204 articles that were assessed for eligibility. A total of 23 studies were included in the systematic review and 15 were included in the meta-analysis. For Neurogranin, 827 AD and 1,237 CU subjects were included in the meta-analysis, showing a significant increase in cerebrospinal fluid of patients with AD compared to CU individuals, with an effect size of 1.01 (p < 0.001). A significant increase in SNAP-25 and GAP-43 levels in CSF of patients with AD was observed. CONCLUSION Neurogranin, SNAP-25, and GAP-43 are possible biomarkers of synaptic damage in AD, and other potential synaptic biomarkers are emerging. This meta-analysis also revealed that there are still relatively few studies investigating these biomarkers in patients with AD or other dementias and showed wide heterogeneity in literature.
Collapse
Affiliation(s)
- Fausto Roveta
- Department of Neuroscience Rita Levi Montalcini, University of Torino, Torino, Italy
| | - Aurora Cermelli
- Department of Neuroscience Rita Levi Montalcini, University of Torino, Torino, Italy
| | - Silvia Boschi
- Department of Neuroscience Rita Levi Montalcini, University of Torino, Torino, Italy
| | - Fabio Ferrandes
- Department of Neuroscience Rita Levi Montalcini, University of Torino, Torino, Italy
| | - Alberto Grassini
- Department of Neuroscience Rita Levi Montalcini, University of Torino, Torino, Italy
| | - Andrea Marcinnò
- Department of Neuroscience Rita Levi Montalcini, University of Torino, Torino, Italy
| | - Margherita Spina
- Department of Neuroscience Rita Levi Montalcini, University of Torino, Torino, Italy
| | - Elisa Rubino
- Department of Neuroscience Rita Levi Montalcini, University of Torino, Torino, Italy
| | - Tiziana Borsello
- Department of Pharmacological and Biomolecular Sciences University of Milano, Milan, Italy
- Mario Negri Institute for Pharmacological Research, University of Milano, Milan, Italy
| | - Alessandro Vercelli
- Department of Neuroscience Rita Levi Montalcini, University of Torino, Torino, Italy
- Neuroscience Institute Cavalieri Ottolenghi, University of Torino, Orbassano, Italy
| | - Innocenzo Rainero
- Department of Neuroscience Rita Levi Montalcini, University of Torino, Torino, Italy
| |
Collapse
|