1
|
Lehmann S, Schraen-Maschke S, Buée L, Vidal JS, Delaby C, Hirtz C, Blanc F, Paquet C, Allinquant B, Bombois S, Gabelle A, Hanon O. Clarifying the association of CSF Aβ, tau, BACE1, and neurogranin with AT(N) stages in Alzheimer disease. Mol Neurodegener 2024; 19:66. [PMID: 39380095 PMCID: PMC11460012 DOI: 10.1186/s13024-024-00755-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 09/13/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Current AT(N) stratification for Alzheimer's disease (AD) accounts for complex combinations of amyloid (A), tau proteinopathy (T) and neurodegeneration (N) signatures. Understanding the transition between these different stages is a major challenge, especially in view of the recent development of disease modifying therapy. METHODS This is an observational study, CSF levels of Tau, pTau181, pTau217, Aβ38/40/42, sAPPα/β, BACE1 and neurogranin were measured in the BALTAZAR cohort of cognitively impaired patients and in the Alzheimer's Disease Neuroimaging Initiative (ADNI). Biomarkers levels were related to the AT(N) framework. (A) and (T) were defined in BALTAZAR with CSF Aβ42/40 ratio and pTau217 respectively, and in ADNI with amyloid and tau PET. (N) was defined using total CSF tau in both cohorts. RESULTS As expected, CSF Aβ42 decreased progressively with the AD continuum going from the A-T-N- to the A + T + N + profile. On the other hand, Tau and pTau181 increased progressively with the disease. The final transition from A + T + N- to A + T + N + led to a sharp increase in Aβ38, Aβ42 and sAPP levels. Synaptic CSF biomarkers BACE1 and neurogranin, were lowest in the initial A + T-N- stage and increased with T + and N + . CSF pTau181 and total tau were closely related in both cohorts. CONCLUSIONS The early transition to an A + phenotype (A + T-N-) primarily impacts synaptic function. The appearance of T + and then N + is associated with a significant and progressive increase in pathological Alzheimer's disease biomarkers. Our main finding is that CSF pTau181 is an indicator of N + rather than T + , and that N + is associated with elevated levels of BACE1 protein and beta-amyloid peptides. This increase may potentially fuel the amyloid cascade in a positive feedback loop. Overall, our data provide further insights into understanding the interconnected pathological processes of amyloid, tau, and neurodegeneration underlying Alzheimer's disease.
Collapse
Affiliation(s)
- Sylvain Lehmann
- LBPC-PPC, Université de Montpellier, INM INSERM, IRMB CHU de Montpellier, 80 av Fliche, Montpellier, 34295, France.
| | - Susanna Schraen-Maschke
- Univ. Lille, Inserm, CHU Lille, UMR-S-U1172, LiCEND, Lille Neuroscience & Cognition, LabEx DISTALZ, Lille, F-59000, France
| | - Luc Buée
- Univ. Lille, Inserm, CHU Lille, UMR-S-U1172, LiCEND, Lille Neuroscience & Cognition, LabEx DISTALZ, Lille, F-59000, France
| | - Jean-Sébastien Vidal
- Université Paris Cité, EA 4468, APHP, Hospital Broca, Memory Resource and Research Centre of de Paris-Broca-Ile de France, Paris, F-75013, France
| | - Constance Delaby
- LBPC-PPC, Université de Montpellier, INM INSERM, IRMB CHU de Montpellier, 80 av Fliche, Montpellier, 34295, France
- Sant Pau Memory Unit, Hospital de la Santa Creu i Sant Pau - Biomedical Research Institute Sant Pau - Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Christophe Hirtz
- LBPC-PPC, Université de Montpellier, INM INSERM, IRMB CHU de Montpellier, 80 av Fliche, Montpellier, 34295, France
| | - Frédéric Blanc
- Université de Strasbourg, CHRU de Strasbourg, Memory Resource and Research Centre of Strasbourg/Colmar, French National Centre for Scientific Research (CNRS), ICube Laboratory and Fédération de Médecine Translationnelle de Strasbourg (FMTS), Team Imagerie Multimodale Intégrative en Santé (IMIS)/Neurocrypto, Strasbourg, F-67000, France
| | - Claire Paquet
- Université Paris Cité, GHU APHP Nord Lariboisière Fernand Widal, Centre de Neurologie Cognitive, Paris, F-75010, France
| | - Bernadette Allinquant
- UMR-S1266, Université Paris Cité, Institute of Psychiatry and Neuroscience, Inserm, Paris, France
| | - Stéphanie Bombois
- Univ. Lille, Inserm, CHU Lille, UMR-S-U1172, LiCEND, Lille Neuroscience & Cognition, LabEx DISTALZ, Lille, F-59000, France
- Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Neurologie, Centre des Maladies Cognitives et Comportementales, GH Pitié-Salpêtrière, Paris, France
| | - Audrey Gabelle
- Université de Montpellier, Memory Research and Resources Center, Department of Neurology, Inserm INM NeuroPEPs team, Montpellier, F-34000, France
| | - Olivier Hanon
- Université Paris Cité, EA 4468, APHP, Hospital Broca, Memory Resource and Research Centre of de Paris-Broca-Ile de France, Paris, F-75013, France
| |
Collapse
|
2
|
Mehdipour Ghazi M, Selnes P, Timón-Reina S, Tecelão S, Ingala S, Bjørnerud A, Kirsebom BE, Fladby T, Nielsen M. Comparative analysis of multimodal biomarkers for amyloid-beta positivity detection in Alzheimer's disease cohorts. Front Aging Neurosci 2024; 16:1345417. [PMID: 38469163 PMCID: PMC10925621 DOI: 10.3389/fnagi.2024.1345417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/12/2024] [Indexed: 03/13/2024] Open
Abstract
Introduction Efforts to develop cost-effective approaches for detecting amyloid pathology in Alzheimer's disease (AD) have gained significant momentum with a focus on biomarker classification. Recent research has explored non-invasive and readily accessible biomarkers, including magnetic resonance imaging (MRI) biomarkers and some AD risk factors. Methods In this comprehensive study, we leveraged a diverse dataset, encompassing participants with varying cognitive statuses from multiple sources, including cohorts from the Alzheimer's Disease Neuroimaging Initiative (ADNI) and our in-house Dementia Disease Initiation (DDI) cohort. As brain amyloid plaques have been proposed as sufficient for AD diagnosis, our primary aim was to assess the effectiveness of multimodal biomarkers in identifying amyloid plaques, using deep machine learning methodologies. Results Our findings underscore the robustness of the utilized methods in detecting amyloid beta positivity across multiple cohorts. Additionally, we investigated the potential of demographic data to enhance MRI-based amyloid detection. Notably, the inclusion of demographic risk factors significantly improved our models' ability to detect amyloid-beta positivity, particularly in early-stage cases, exemplified by an average area under the ROC curve of 0.836 in the unimpaired DDI cohort. Discussion These promising, non-invasive, and cost-effective predictors of MRI biomarkers and demographic variables hold the potential for further refinement through considerations like APOE genotype and plasma markers.
Collapse
Affiliation(s)
- Mostafa Mehdipour Ghazi
- Department of Computer Science, Pioneer Centre for Artificial Intelligence, University of Copenhagen, Copenhagen, Denmark
| | - Per Selnes
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, Campus Ahus, University of Oslo, Oslo, Norway
| | | | - Sandra Tecelão
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway
| | - Silvia Ingala
- Department of Radiology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Atle Bjørnerud
- Department of Physics, University of Oslo, Oslo, Norway
- Unit for Computational Radiology and Artificial Intelligence, Oslo University Hospital, Oslo, Norway
| | - Bjørn-Eivind Kirsebom
- Department of Neurology, University Hospital of North Norway, Tromsø, Norway
- Department of Psychology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Tormod Fladby
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, Campus Ahus, University of Oslo, Oslo, Norway
| | - Mads Nielsen
- Department of Computer Science, Pioneer Centre for Artificial Intelligence, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
3
|
Xu Y, Jiang H, Zhu B, Cao M, Feng T, Sun Z, Du G, Zhao Z. Advances and applications of fluids biomarkers in diagnosis and therapeutic targets of Alzheimer's disease. CNS Neurosci Ther 2023. [PMID: 37144603 DOI: 10.1111/cns.14238] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 01/25/2023] [Accepted: 04/12/2023] [Indexed: 05/06/2023] Open
Abstract
AIMS Alzheimer's disease (AD) is a neurodegenerative disease with challenging early diagnosis and effective treatments due to its complex pathogenesis. AD patients are often diagnosed after the appearance of the typical symptoms, thereby delaying the best opportunity for effective measures. Biomarkers could be the key to resolving the challenge. This review aims to provide an overview of application and potential value of AD biomarkers in fluids, including cerebrospinal fluid, blood, and saliva, in diagnosis and treatment. METHODS A comprehensive search of the relevant literature was conducted to summarize potential biomarkers for AD in fluids. The paper further explored the biomarkers' utility in disease diagnosis and drug target development. RESULTS Research on biomarkers mainly focused on amyloid-β (Aβ) plaques, Tau protein abnormal phosphorylation, axon damage, synaptic dysfunction, inflammation, and related hypotheses associated with AD mechanisms. Aβ42 , total Tau (t-Tau), and phosphorylated Tau (p-Tau), have been endorsed for their diagnostic and predictive capability. However, other biomarkers remain controversial. Drugs targeting Aβ have shown some efficacy and those that target BACE1 and Tau are still undergoing development. CONCLUSION Fluid biomarkers hold considerable potential in the diagnosis and drug development of AD. However, improvements in sensitivity and specificity, and approaches for managing sample impurities, need to be addressed for better diagnosis.
Collapse
Affiliation(s)
- Yanan Xu
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- School of Pharmacy, Capital Medical University, Beijing, China
| | - Hailun Jiang
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Bin Zhu
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Mingnan Cao
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Tao Feng
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhongshi Sun
- Department of Pharmacy, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Guanhua Du
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Zhigang Zhao
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- School of Pharmacy, Capital Medical University, Beijing, China
| |
Collapse
|
4
|
Medina-Vera D, Enache D, Tambaro S, Abuhashish E, Rosell-Valle C, Winblad B, Rodríguez de Fonseca F, Bereczki E, Nilsson P. Translational potential of synaptic alterations in Alzheimer's disease patients and amyloid precursor protein knock-in mice. Brain Commun 2023; 5:fcad001. [PMID: 36687391 PMCID: PMC9851419 DOI: 10.1093/braincomms/fcad001] [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: 02/21/2022] [Revised: 09/19/2022] [Accepted: 01/03/2023] [Indexed: 01/09/2023] Open
Abstract
Synaptic dysfunction is an early event in Alzheimer's disease. Post-mortem studies suggest that alterations in synaptic proteins are associated with cognitive decline in Alzheimer's disease. We measured the concentration of three synaptic proteins, zinc transporter protein 3, dynamin1 and AMPA glutamate receptor 3 in cerebrospinal fluid of subjects with mild cognitive impairment (n = 18) and Alzheimer's disease (n = 18) and compared the levels to cognitively and neurologically healthy controls (n = 18) by using ELISA assay. In addition, we aimed to assess the translational potential of these synaptic proteins in two established amyloid precursor protein knock-in Alzheimer's disease mouse models by assessing the cerebrospinal fluid, hippocampal and cortical synaptic protein concentrations. Using ELISA, we measured in parallel these three proteins in cerebrospinal fluid and/or brain of 12- and 24-month-old AppNL-F and AppNL-G-F knock-in mice and AppWt control mice. The regional distribution and expression of these proteins were explored upon aging of the App knock-in models by quantitative immunofluorescence microscopy. Notably, we found a significant increase in concentrations of zinc transporter protein 3 and AMPA glutamate receptor 3 in cerebrospinal fluid of both patient groups compared with cognitively healthy controls. Dynamin1 concentration was significantly higher in Alzheimer's disease patients. Remarkably, patients with mild cognitive impairment who converted to Alzheimer's disease (n = 7) within 2 years exhibited elevated baseline cerebrospinal fluid zinc transporter protein 3 concentrations compared with mild cognitive impairment patients who did not convert (n = 11). Interestingly, similar to the alterations in Alzheimer's disease subjects, cerebrospinal fluid AMPA glutamate receptor 3 concentration was significantly higher in AppNL-G-F knock-in mice when compared with wild-type controls. Furthermore, we have detected age and brain regional specific changes of the three synaptic proteins in the hippocampus and prefrontal cortex of both AppNL-F and AppNL-G-F knock-in mice. Notably, all the three cerebrospinal fluid synaptic protein concentrations correlated negatively with concentrations in hippocampal lysates. The elevated zinc transporter protein 3 concentrations in the cerebrospinal fluid of converter versus non-converter mild cognitive impairment patients suggests a prospective role of zinc transporter 3 in differentiating dementia patients of the biological continuum of Alzheimer's disease. The increased cerebrospinal fluid concentrations of synaptic proteins in both patient groups, potentially reflecting synaptic alterations in the brain, were similarly observed in the amyloid precursor protein knock-in mouse models highlighting the translational potential of these proteins as markers for synaptic alterations. These synaptic markers could potentially help reduce the current disparities between human and animal model-based studies aiding the translation of preclinical discoveries of pathophysiological changes into clinical research.
Collapse
Affiliation(s)
- Dina Medina-Vera
- Instituto de Investigación Biomédica de Málaga-IBIMA, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Málaga 29010, Spain,Facultad de Ciencias, Universidad de Málaga, Málaga 29010, Spain,Facultad de Medicina, Universidad de Málaga, Málaga 29010, Spain
| | - Daniela Enache
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, 17164 Solna, Sweden
| | - Simone Tambaro
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, 17164 Solna, Sweden
| | - Ethar Abuhashish
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, 17164 Solna, Sweden
| | - Cristina Rosell-Valle
- Instituto de Investigación Biomédica de Málaga-IBIMA, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Málaga 29010, Spain
| | - Bengt Winblad
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, 17164 Solna, Sweden,Theme Inflammation and Aging, Karolinska University Hospital, 17164 Solna, Sweden
| | - Fernando Rodríguez de Fonseca
- Instituto de Investigación Biomédica de Málaga-IBIMA, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Málaga 29010, Spain
| | - Erika Bereczki
- Correspondence to: Erika Bereczki Department of NVS, Center for Alzheimer Research Division of Neurogeriatrics, Karolinska Institutet BioClinicum J10:30, 17 164, Stockholm, Sweden E-mail:
| | | |
Collapse
|
5
|
Cano A, Ettcheto M, Bernuz M, Puerta R, Esteban de Antonio E, Sánchez-López E, Souto EB, Camins A, Martí M, Pividori MI, Boada M, Ruiz A. Extracellular vesicles, the emerging mirrors of brain physiopathology. Int J Biol Sci 2023; 19:721-743. [PMID: 36778117 PMCID: PMC9910004 DOI: 10.7150/ijbs.79063] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 11/01/2022] [Indexed: 01/11/2023] Open
Abstract
Extracellular vesicles are secreted by a wide variety of cells, and their primary functions include intercellular communication, immune responses, human reproduction, and synaptic plasticity. Their molecular cargo reflects the physiological processes that their cells of origin are undergoing. Thus, many studies have suggested that extracellular vesicles could be a promising biomarker tool for many diseases, mainly due to their biological relevance and easy accessibility to a broad range of body fluids. Moreover, since their biological composition leads them to cross the blood-brain barrier bidirectionally, growing evidence points to extracellular vesicles as emerging mirrors of brain diseases processes. In this regard, this review explores the biogenesis and biological functions of extracellular vesicles, their role in different physiological and pathological processes, their potential in clinical practice, and the recent outstanding studies about the role of exosomes in major human brain diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), or brain tumors.
Collapse
Affiliation(s)
- Amanda Cano
- Ace Alzheimer Center Barcelona - International University of Catalunya (UIC), Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Institute of Nanoscience and Nanotechnology (IN2UB), Barcelona, Spain.,Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Spain
| | - Miren Ettcheto
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Spain
| | - Mireia Bernuz
- Biosensing and Bioanalysis Group, Institut de Biotecnologia i de Biomedicina (IBB-UAB), Mòdul B Parc de Recerca UAB, Campus Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,Grup de Sensors i Biosensors, Departament de Química, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Raquel Puerta
- Ace Alzheimer Center Barcelona - International University of Catalunya (UIC), Barcelona, Spain
| | | | - Elena Sánchez-López
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Institute of Nanoscience and Nanotechnology (IN2UB), Barcelona, Spain.,Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Spain.,Unit of Synthesis and Biomedical Applications of Peptides, IQAC-CSIC, 08034 Barcelona, Spain
| | - Eliana B Souto
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto, Porto, Portugal.,REQUIMTE/UCIBIO, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Antonio Camins
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Spain
| | - Mercè Martí
- Biosensing and Bioanalysis Group, Institut de Biotecnologia i de Biomedicina (IBB-UAB), Mòdul B Parc de Recerca UAB, Campus Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - María Isabel Pividori
- Biosensing and Bioanalysis Group, Institut de Biotecnologia i de Biomedicina (IBB-UAB), Mòdul B Parc de Recerca UAB, Campus Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,Grup de Sensors i Biosensors, Departament de Química, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Mercè Boada
- Ace Alzheimer Center Barcelona - International University of Catalunya (UIC), Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Agustín Ruiz
- Ace Alzheimer Center Barcelona - International University of Catalunya (UIC), Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| |
Collapse
|
6
|
Saunders TS, Gadd DA, Spires‐Jones TL, King D, Ritchie C, Muniz‐Terrera G. Associations between cerebrospinal fluid markers and cognition in ageing and dementia: A systematic review. Eur J Neurosci 2022; 56:5650-5713. [PMID: 35338546 PMCID: PMC9790745 DOI: 10.1111/ejn.15656] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/08/2022] [Accepted: 03/13/2022] [Indexed: 12/30/2022]
Abstract
A biomarker associated with cognition in neurodegenerative dementias would aid in the early detection of disease progression, complement clinical staging and act as a surrogate endpoint in clinical trials. The current systematic review evaluates the association between cerebrospinal fluid protein markers of synapse loss and neuronal injury and cognition. We performed a systematic search which revealed 67 studies reporting an association between cerebrospinal fluid markers of interest and neuropsychological performance. Despite the substantial heterogeneity between studies, we found some evidence for an association between neurofilament-light and worse cognition in Alzheimer's diseases, frontotemporal dementia and typical cognitive ageing. Moreover, there was an association between cerebrospinal fluid neurogranin and cognition in those with an Alzheimer's-like cerebrospinal fluid biomarker profile. Some evidence was found for cerebrospinal fluid neuronal pentraxin-2 as a correlate of cognition across dementia syndromes. Due to the substantial heterogeneity of the field, no firm conclusions can be drawn from this review. Future research should focus on improving standardization and reporting as well as establishing the importance of novel markers such as neuronal pentraxin-2 and whether such markers can predict longitudinal cognitive decline.
Collapse
Affiliation(s)
- Tyler S. Saunders
- UK Dementia Research InstituteThe University of EdinburghEdinburghUK
- Center for Discovery Brain SciencesThe University of EdinburghEdinburghUK
- Center for Clinical Brain SciencesThe University of EdinburghEdinburghUK
- Center for Dementia PreventionThe University of EdinburghEdinburghUK
| | - Danni A. Gadd
- Center for Genomic and Experimental Medicine, Institute of Genetics and Molecular MedicineUniversity of EdinburghEdinburghUK
| | - Tara L. Spires‐Jones
- UK Dementia Research InstituteThe University of EdinburghEdinburghUK
- Center for Discovery Brain SciencesThe University of EdinburghEdinburghUK
| | - Declan King
- UK Dementia Research InstituteThe University of EdinburghEdinburghUK
- Center for Discovery Brain SciencesThe University of EdinburghEdinburghUK
| | - Craig Ritchie
- Center for Clinical Brain SciencesThe University of EdinburghEdinburghUK
- Center for Dementia PreventionThe University of EdinburghEdinburghUK
| | - Graciela Muniz‐Terrera
- Center for Clinical Brain SciencesThe University of EdinburghEdinburghUK
- Center for Dementia PreventionThe University of EdinburghEdinburghUK
| |
Collapse
|
7
|
Timsina J, Gomez-Fonseca D, Wang L, Do A, Western D, Alvarez I, Aguilar M, Pastor P, Henson RL, Herries E, Xiong C, Schindler SE, Fagan AM, Bateman RJ, Farlow M, Morris JC, Perrin R, Moulder K, Hassenstab J, Chhatwal J, Mori H, ADNI, DIAN Consortia, Sung YJ, Cruchaga C. Comparative Analysis of Alzheimer's Disease Cerebrospinal Fluid Biomarkers Measurement by Multiplex SOMAscan Platform and Immunoassay-Based Approach. J Alzheimers Dis 2022; 89:193-207. [PMID: 35871346 PMCID: PMC9562128 DOI: 10.3233/jad-220399] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND The SOMAscan assay has an advantage over immunoassay-based methods because it measures a large number of proteins in a cost-effective manner. However, the performance of this technology compared to the routinely used immunoassay techniques needs to be evaluated. OBJECTIVE We performed comparative analyses of SOMAscan and immunoassay-based protein measurements for five cerebrospinal fluid (CSF) proteins associated with Alzheimer's disease (AD) and neurodegeneration: NfL, Neurogranin, sTREM2, VILIP-1, and SNAP-25. METHODS We compared biomarkers measured in ADNI (N = 689), Knight-ADRC (N = 870), DIAN (N = 115), and Barcelona-1 (N = 92) cohorts. Raw protein values were transformed using z-score in order to combine measures from the different studies. sTREM2 and VILIP-1 had more than one analyte in SOMAscan; all available analytes were evaluated. Pearson's correlation coefficients between SOMAscan and immunoassays were calculated. Receiver operating characteristic curve and area under the curve were used to compare prediction accuracy of these biomarkers between the two platforms. RESULTS Neurogranin, VILIP-1, and NfL showed high correlation between SOMAscan and immunoassay measures (r > 0.9). sTREM2 had a fair correlation (r > 0.6), whereas SNAP-25 showed weak correlation (r = 0.06). Measures in both platforms provided similar predicted performance for all biomarkers except SNAP-25 and one of the sTREM2 analytes. sTREM2 showed higher AUC for SOMAscan based measures. CONCLUSION Our data indicate that SOMAscan performs as well as immunoassay approaches for NfL, Neurogranin, VILIP-1, and sTREM2. Our study shows promise for using SOMAscan as an alternative to traditional immunoassay-based measures. Follow-up investigation will be required for SNAP-25 and additional established biomarkers.
Collapse
Affiliation(s)
- Jigyasha Timsina
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Duber Gomez-Fonseca
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Lihua Wang
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Anh Do
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Dan Western
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Ignacio Alvarez
- Memory Disorders Unit, Department of Neurology, University Hospital Mutua Terrassa, Terrassa, Spain
| | - Miquel Aguilar
- Memory Disorders Unit, Department of Neurology, University Hospital Mutua Terrassa, Terrassa, Spain
| | - Pau Pastor
- Memory Disorders Unit, Department of Neurology, University Hospital Mutua Terrassa, Terrassa, Spain
| | - Rachel L. Henson
- Hope Center for Neurologic Diseases, Washington University in St. Louis, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Elizabeth Herries
- Hope Center for Neurologic Diseases, Washington University in St. Louis, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Chengjie Xiong
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Suzanne E. Schindler
- Hope Center for Neurologic Diseases, Washington University in St. Louis, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Anne M. Fagan
- Hope Center for Neurologic Diseases, Washington University in St. Louis, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Randall J. Bateman
- Hope Center for Neurologic Diseases, Washington University in St. Louis, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Martin Farlow
- Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana University Health, Indianapolis, IN, USA
| | - John C. Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Richard Perrin
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Krista Moulder
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jason Hassenstab
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jasmeer Chhatwal
- Massachusetts General Hospital, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Hiroshi Mori
- Dept. of Clinical Neuroscience, Osaka City University Medical School, Nagaoka Sutoku University, Japan
| | | | | | - Yun Ju Sung
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurologic Diseases, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
8
|
Yoong SQ, Lu J, Xing H, Gyanwali B, Tan YQ, Wu XV. The prognostic utility of CSF neurogranin in predicting future cognitive decline in the Alzheimer's disease continuum: A systematic review and meta-analysis with narrative synthesis. Ageing Res Rev 2021; 72:101491. [PMID: 34688925 DOI: 10.1016/j.arr.2021.101491] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 09/13/2021] [Accepted: 10/15/2021] [Indexed: 01/08/2023]
Abstract
Core cerebrospinal fluid (CSF) biomarkers (Aβ42, T-tau, P-tau) were included as supporting diagnostic criteria for Alzheimer's Disease (AD), but they lack the power to predict AD progression. On the other hand, a new biomarker CSF Neurogranin (Ng) has been shown to predict cognitive decline. This systematic review aims to synthesise the prognostic utility of CSF Ng in predicting cognitive decline in the AD continuum. Seven databases were searched systematically from inception to 30 September 2020. Participants were 55 years or older, who had baseline and at least one follow-up cognitive assessments. Risk of bias was assessed using the Quality in Prognosis Studies tool. Meta-analysis was conducted by pooling standardised beta coefficients and adjusted hazard ratios. Thirteen studies were included and high-quality evidence suggests that CSF Ng predicts Mini-Mental State Examination (MMSE) decline in Aβ+ mild cognitive impairment (MCI). Moderate quality evidence showed that CSF Ng could predict the decline of memory and executive function in MCI. Narrative synthesis found that CSF Ng/Aβ42 was also likely to predict cognitive decline. More studies are required to validate the use of CSF Ng as an AD prognostic marker and its application in future development of drug treatment and diagnosis.
Collapse
|
9
|
Siafarikas N, Kirsebom BE, Srivastava DP, Eriksson CM, Auning E, Hessen E, Selbaek G, Blennow K, Aarsland D, Fladby T. Cerebrospinal fluid markers for synaptic function and Alzheimer type changes in late life depression. Sci Rep 2021; 11:20375. [PMID: 34645914 PMCID: PMC8514484 DOI: 10.1038/s41598-021-99794-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 09/28/2021] [Indexed: 01/11/2023] Open
Abstract
To explore markers for synaptic function and Alzheimer disease (AD) pathology in late life depression (LLD), predementia AD and normal controls (NC). A cross-sectional study to compare cerebrospinal fluid (CSF) levels of neurogranin (Ng), Beta-site amyloid-precursor-protein cleaving enzyme1 (BACE1), Ng/BACE1 ratio and Amyloid-β 42/40 ratio, phosphorylated-tau and total-tau in LLD with (LLD AD) or without (LLD NoAD) AD pathology, predementia AD and normal controls (NC). We included 145 participants (NC = 41; predementia AD = 66 and LLD = 38). LLD comprised LLD AD (n = 16), LLD NoAD (n = 19), LLD with non-AD typical changes (n = 3, excluded). LLD AD (pADJ < 0.05) and predementia AD (pADJ < 0.0001) showed significantly higher Ng than NC. BACE1 and Ng/BACE1 ratio were altered similarly. Compared to LLD NoAD, LLD AD showed significantly higher Ng (pADJ < 0.001), BACE1 (pADJ < 0.05) and Ng/BACE1 ratio (pADJ < 0.01). All groups had significantly lower Aβ 42/40 ratio than NC (predementia AD and LLD AD, p < 0.0001; LLD NoAD, p < 0.05). Both LLD groups performed similarly on tests of memory and executive function, but significantly poorer than NC. Synaptic function in LLD depended on AD pathology. LLD showed an association to Amyloid dysmetabolism. The LLD groups performed poorer cognitively than NC. LLD AD may be conceptualized as "predementia AD with depression".
Collapse
Affiliation(s)
- Nikias Siafarikas
- Department of Geriatric Psychiatry, Akershus University Hospital, Sykehusveien 25, 1478, Lørenskog, Norway.
- Faculty of Medicine, University of Oslo, Oslo, Norway.
| | - Bjørn-Eivind Kirsebom
- Department of Neurology, University Hospital of North Norway, Tromsø, Norway
- Department of Psychology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Deepak P Srivastava
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 9NU, UK
| | - Cecilia M Eriksson
- Department of Geriatric Psychiatry, Akershus University Hospital, Sykehusveien 25, 1478, Lørenskog, Norway
| | - Eirik Auning
- Department of Geriatric Psychiatry, Akershus University Hospital, Sykehusveien 25, 1478, Lørenskog, Norway
| | - Erik Hessen
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Geir Selbaek
- Faculty of Medicine, University of Oslo, Oslo, Norway
- Norwegian National Advisory Unit On Aging and Health, Vestfold Hospital Trust, Tønsberg, Norway
- Department of Geriatric Medicine, Oslo University Hospital, Oslo, Norway
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Dag Aarsland
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
| | - Tormod Fladby
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, University of Oslo, Campus Ahus, Oslo, Norway
| |
Collapse
|
10
|
Bergland AK, Proitsi P, Kirsebom BE, Soennesyn H, Hye A, Larsen AI, Xu J, Legido-Quigley C, Rajendran L, Fladby T, Aarsland D. Exploration of Plasma Lipids in Mild Cognitive Impairment due to Alzheimer's Disease. J Alzheimers Dis 2021; 77:1117-1127. [PMID: 32804144 DOI: 10.3233/jad-200441] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Lipids have important structural roles in cell membranes and changes to these membrane lipids may influence β- and γ-secretase activities and thus contribute to Alzheimer's disease (AD) pathology. OBJECTIVE To explore baseline plasma lipid profiling in participants with mild cognitive impairment (MCI) with and without AD pathology. METHODS We identified 261 plasma lipids using reversed-phase liquid chromatography/mass spectrometry in cerebrospinal fluid amyloid positive (Aβ+) or negative (Aβ-) participants with MCI as compared to controls. Additionally, we analyzed the potential associations of plasma lipid profiles with performance on neuropsychological tests at baseline and after two years. RESULTS Sphingomyelin (SM) concentrations, particularly, SM(d43:2), were lower in MCI Aβ+ individuals compared to controls. Further, SM(d43:2) was also nominally reduced in MCI Aβ+ individuals compared to MCI Aβ-. No plasma lipids were associated with performance on primary neuropsychological tests at baseline or between the two time points after correction for multiple testing. CONCLUSION Reduced plasma concentrations of SM were associated with AD.
Collapse
Affiliation(s)
- Anne Katrine Bergland
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Petroula Proitsi
- Maurice Wohl Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Bjørn-Eivind Kirsebom
- Department of Neurology, University Hospital of North Norway, Tromsø, Norway.,Department of Psychology, Faculty of Health Sciences, UiT, The Arctic University of Norway, Tromsø, Norway
| | - Hogne Soennesyn
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Abdul Hye
- Maurice Wohl Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Alf Inge Larsen
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Cardiology, Stavanger University Hospital, Stavanger, Norway
| | - Jin Xu
- Maurice Wohl Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.,Institute of Pharmaceutical Science, King's College London, London, UK
| | - Cristina Legido-Quigley
- Institute of Pharmaceutical Science, King's College London, London, UK.,Systems Medicine, Steno Diabetes Centre, Copenhagen, Denmark
| | - Lawrence Rajendran
- UK Dementia Research Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Tormod Fladby
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway.,Institute of Clinical Medicine, Campus Ahus, University of Oslo, Oslo, Norway
| | - Dag Aarsland
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway.,UK Dementia Research Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| |
Collapse
|
11
|
Enache D, Pereira JB, Jelic V, Winblad B, Nilsson P, Aarsland D, Bereczki E. Increased Cerebrospinal Fluid Concentration of ZnT3 Is Associated with Cognitive Impairment in Alzheimer's Disease. J Alzheimers Dis 2021; 77:1143-1155. [PMID: 32925049 DOI: 10.3233/jad-200498] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Cognitive deficits arising in the course of Alzheimer's disease (AD), dementia with Lewy bodies (DLB), and Parkinson's disease with dementia (PDD) are directly linked to synaptic loss. Postmortem studies suggest that zinc transporter protein 3 (ZnT3), AMPA glutamate receptor 3 (GluA3), and Dynamin1 are associated with cognitive decline in AD and Lewy body dementia patients. OBJECTIVE We aimed to evaluate the diagnostic value of ZnT3, GluA3, and Dynamin 1 in the cerebrospinal fluid (CSF) of patients with dementia due to AD, DLB, and PDD compared to cognitively normal subjective cognitive decline (SCD) patients in a retrospective study. In addition, we assessed the relationship between synaptic markers and age, sex, cognitive impairment, and depressive symptoms as well as CSF amyloid, phosphorylated tau (p-tau), and total tau (T-tau). METHODS Commercially available ELISA immunoassay was used to measure the levels of proteins in a total of 97 CSF samples from AD (N = 24), PDD (N = 18), DLB (N = 27), and SCD (N = 28) patients. Cognitive impairment was assessed using the Mini-Mental State Examination (MMSE). RESULTS We found a significant increase in the concentrations of ZnT3, GluA3, and Dynamin1 in AD (p = 0.002) and of ZnT3 and Dynamin 1 in DLB (p = 0.001, p = 0.002) when compared to SCD patients. Changes in ZnT3 concentrations correlated with MMSE scores in AD (p = 0.011), and with depressive symptoms in SCD (p = 0.041). CONCLUSION We found alteration of CSF levels of synaptic proteins in AD, PDD, and DLB. Our results reveal distinct changes in CSF concentrations of ZnT3 that could reflect cognitive impairment in AD with implications for future prognostic and diagnostic marker development.
Collapse
Affiliation(s)
- Daniela Enache
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Joana B Pereira
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Clinical Geriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Vesna Jelic
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Clinical Geriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Bengt Winblad
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Per Nilsson
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Dag Aarsland
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden.,Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.,Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Erika Bereczki
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
12
|
Lazarev VF, Tsolaki M, Mikhaylova ER, Benken KA, Shevtsov MA, Nikotina AD, Lechpammer M, Mitkevich VA, Makarov AA, Moskalev AA, Kozin SA, Margulis BA, Guzhova IV, Nudler E. Extracellular GAPDH Promotes Alzheimer Disease Progression by Enhancing Amyloid-β Aggregation and Cytotoxicity. Aging Dis 2021; 12:1223-1237. [PMID: 34341704 PMCID: PMC8279520 DOI: 10.14336/ad.2020.1230] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 12/30/2020] [Indexed: 01/10/2023] Open
Abstract
Neuronal cell death at late stages of Alzheimer's disease (AD) causes the release of cytosolic proteins. One of the most abundant such proteins, glyceraldehyde-3-phosphate dehydrogenase (GAPDH), forms stable aggregates with extracellular amyloid-β (Aβ). We detect these aggregates in cerebrospinal fluid (CSF) from AD patients at levels directly proportional to the progressive stages of AD. We found that GAPDH forms a covalent bond with Q15 of Aβ that is mediated by transglutaminase (tTG). The Q15A substitution weakens the interaction between Aβ and GAPDH and reduces Aβ-GAPDH cytotoxicity. Lentivirus-driven GAPDH overexpression in two AD animal models increased the level of apoptosis of hippocampal cells, neural degeneration, and cognitive dysfunction. In contrast, in vivo knockdown of GAPDH reversed these pathogenic abnormalities suggesting a pivotal role of GAPDH in Aβ-stimulated neurodegeneration. CSF from animals with enhanced GAPDH expression demonstrates increased cytotoxicity in vitro. Furthermore, RX-624, a specific GAPDH small molecular ligand reduced accumulation of Aβ aggregates and reversed memory deficit in AD transgenic mice. These findings argue that extracellular GAPDH compromises Aβ clearance and accelerates neurodegeneration, and, thus, is a promising pharmacological target for AD.
Collapse
Affiliation(s)
- Vladimir F Lazarev
- Institute of Cytology of the Russian Academy of Sciences (RAS), Petersburg, Russia.
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
| | - Magda Tsolaki
- 1 University Department of Neurology, AHEPA hospital Aristotle University of Thessaloniki and Greek Alzheimer Association, Thessaloniki, Greece.
| | - Elena R Mikhaylova
- Institute of Cytology of the Russian Academy of Sciences (RAS), Petersburg, Russia.
| | | | - Maxim A Shevtsov
- Institute of Cytology of the Russian Academy of Sciences (RAS), Petersburg, Russia.
- Klinikum rechts der Isar, Technische Universität München, Munich, Germany.
| | - Alina D Nikotina
- Institute of Cytology of the Russian Academy of Sciences (RAS), Petersburg, Russia.
| | - Mirna Lechpammer
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA.
| | - Vladimir A Mitkevich
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
| | - Alexander A Makarov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
| | - Alexey A Moskalev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
- Institute of Biology of Komi Scientific Centre of The Ural Branch of The Russian Academy of Sciences, Kommunisticheskaya, Russia.
| | - Sergey A Kozin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
| | - Boris A Margulis
- Institute of Cytology of the Russian Academy of Sciences (RAS), Petersburg, Russia.
| | - Irina V Guzhova
- Institute of Cytology of the Russian Academy of Sciences (RAS), Petersburg, Russia.
| | - Evgeny Nudler
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA.
- Howard Hughes Medical Institute, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
13
|
Peng Q, Zhang Z. The fluid biomarkers of Alzheimer’s disease. BRAIN SCIENCE ADVANCES 2021. [DOI: 10.26599/bsa.2021.9050001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Alzheimer’s disease (AD) is the most common neurodegenerative disorder. However, it still has no available disease‐modifying therapies. Its pathology cascade begins decades before symptomatic presentation. For these reasons, highly sensitive and highly specific fluid biomarkers should be developed for the early diagnosis of AD. In this study, the well‐established and emerging fluid biomarkers of AD are summarized, and recent advances on their role in early diagnosis and progression monitoring as well as their correlations with AD pathology are highlighted. Future prospects and related research directions are also discussed.
Collapse
Affiliation(s)
- Qinyu Peng
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
14
|
Piersson AD, Mohamad M, Suppiah S, Rajab NF. Topographical patterns of whole-brain structural alterations in association with genetic risk, cerebrospinal fluid, positron emission tomography biomarkers of Alzheimer’s disease, and neuropsychological measures. Clin Transl Imaging 2021. [DOI: 10.1007/s40336-021-00440-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
15
|
Cano A, Turowski P, Ettcheto M, Duskey JT, Tosi G, Sánchez-López E, García ML, Camins A, Souto EB, Ruiz A, Marquié M, Boada M. Nanomedicine-based technologies and novel biomarkers for the diagnosis and treatment of Alzheimer's disease: from current to future challenges. J Nanobiotechnology 2021; 19:122. [PMID: 33926475 PMCID: PMC8086346 DOI: 10.1186/s12951-021-00864-x] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/17/2021] [Indexed: 02/07/2023] Open
Abstract
Increasing life expectancy has led to an aging population, which has consequently increased the prevalence of dementia. Alzheimer's disease (AD), the most common form of dementia worldwide, is estimated to make up 50-80% of all cases. AD cases are expected to reach 131 million by 2050, and this increasing prevalence will critically burden economies and health systems in the next decades. There is currently no treatment that can stop or reverse disease progression. In addition, the late diagnosis of AD constitutes a major obstacle to effective disease management. Therefore, improved diagnostic tools and new treatments for AD are urgently needed. In this review, we investigate and describe both well-established and recently discovered AD biomarkers that could potentially be used to detect AD at early stages and allow the monitoring of disease progression. Proteins such as NfL, MMPs, p-tau217, YKL-40, SNAP-25, VCAM-1, and Ng / BACE are some of the most promising biomarkers because of their successful use as diagnostic tools. In addition, we explore the most recent molecular strategies for an AD therapeutic approach and nanomedicine-based technologies, used to both target drugs to the brain and serve as devices for tracking disease progression diagnostic biomarkers. State-of-the-art nanoparticles, such as polymeric, lipid, and metal-based, are being widely investigated for their potential to improve the effectiveness of both conventional drugs and novel compounds for treating AD. The most recent studies on these nanodevices are deeply explained and discussed in this review.
Collapse
Affiliation(s)
- Amanda Cano
- Research Center and Memory Clinic, Fundació ACE. Institut Català de Neurociències Aplicades, International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain.
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
- Institute of Nanoscience and Nanotechnology (IN2UB), Barcelona, Spain.
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain.
| | - Patric Turowski
- UCL Institute of Ophthalmology, University College of London, London, UK
| | - Miren Ettcheto
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
| | - Jason Thomas Duskey
- Nanotech Lab, Te.Far.T.I, Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Umberto Veronesi Foundation, 20121, Milano, Italy
| | - Giovanni Tosi
- Nanotech Lab, Te.Far.T.I, Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Elena Sánchez-López
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), Barcelona, Spain
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
| | - Maria Luisa García
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), Barcelona, Spain
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
| | - Antonio Camins
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
| | - Eliana B Souto
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
- CEB - Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal
| | - Agustín Ruiz
- Research Center and Memory Clinic, Fundació ACE. Institut Català de Neurociències Aplicades, International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Marta Marquié
- Research Center and Memory Clinic, Fundació ACE. Institut Català de Neurociències Aplicades, International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Mercè Boada
- Research Center and Memory Clinic, Fundació ACE. Institut Català de Neurociències Aplicades, International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| |
Collapse
|
16
|
Álvarez I, Diez-Fairen M, Aguilar M, González JM, Ysamat M, Tartari JP, Carcel M, Alonso A, Brix B, Arendt P, Pastor P. Added value of cerebrospinal fluid multimarker analysis in diagnosis and progression of dementia. Eur J Neurol 2021; 28:1142-1152. [PMID: 33236496 DOI: 10.1111/ene.14658] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 11/16/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND PURPOSE Recently, some emerging cerebrospinal fluid (CSF) markers have been proposed as diagnostic tools for Alzheimer disease (AD) that can have an effect on disease progression. We analyze the accuracy of these CSF markers for diagnosis of AD in reference to brain amyloid positron emission tomography (PET). We also investigated whether they help in differentiating AD from other dementias and examined their influence in tracing the progression to dementia. METHODS Amyloid-β (Aβ) 1-42, total tau (t-tau), phosphorylated tau, Aβ40 , Aβ38 , beta-site amyloid precursor protein cleaving enzyme 1 (BACE-1), neurogranin (ng), phosphorylated neurofilament heavy-chain, and α-synuclein (α-syn) CSF levels were analyzed in 319 subjects, among whom 57 also underwent an amyloid PET scan. We also analyzed longitudinal clinical data from 239 subjects. RESULTS Emerging CSF markers, especially ng/BACE-1 ratio (area under the curve = 0.77) and their combinations with core AD CSF markers (all AUCs >0.85), showed high accuracy to discriminate amyloid PET positivity. Subjects with AD had higher CSF BACE-1, ng, and α-syn levels than those with non-AD dementia. CSF t-tau/α-syn ratio was higher in subjects with dementia with Lewy bodies than in those with frontotemporal dementia. Most emerging/core AD ratios predicted a faster conversion from mild cognitive impairment (MCI) stage to AD and appeared to be helpful when core AD CSF markers were discordant. In addition, the rate of cognitive decline was associated with all CSF core AD markers, several emerging/core AD two-marker ratios, and CSF ng levels. CONCLUSIONS These results suggest that emerging biomarkers in conjunction with core AD markers improve diagnosis of AD, are associated with the conversion from MCI into AD, and predict a faster progression of dementia.
Collapse
Affiliation(s)
- Ignacio Álvarez
- Memory Disorders Unit, Department of Neurology, Hospital Universitari Mutua de Terrassa, Terrassa, Spain
- Fundació per a la Recerca Biomèdica i Social Mútua de Terrassa, Terrassa, Spain
| | - Monica Diez-Fairen
- Memory Disorders Unit, Department of Neurology, Hospital Universitari Mutua de Terrassa, Terrassa, Spain
- Fundació per a la Recerca Biomèdica i Social Mútua de Terrassa, Terrassa, Spain
| | - Miquel Aguilar
- Memory Disorders Unit, Department of Neurology, Hospital Universitari Mutua de Terrassa, Terrassa, Spain
- Fundació per a la Recerca Biomèdica i Social Mútua de Terrassa, Terrassa, Spain
| | - Jose Manuel González
- Centre de Tecnologia Diagnòstica, Hospital Universitari Mutua de Terrassa, Terrassa, Spain
| | - Montse Ysamat
- Centre de Tecnologia Diagnòstica, Hospital Universitari Mutua de Terrassa, Terrassa, Spain
| | - Juan Pablo Tartari
- Memory Disorders Unit, Department of Neurology, Hospital Universitari Mutua de Terrassa, Terrassa, Spain
- Fundació per a la Recerca Biomèdica i Social Mútua de Terrassa, Terrassa, Spain
| | - Maria Carcel
- Memory Disorders Unit, Department of Neurology, Hospital Universitari Mutua de Terrassa, Terrassa, Spain
- Fundació per a la Recerca Biomèdica i Social Mútua de Terrassa, Terrassa, Spain
| | - Alvaro Alonso
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Britta Brix
- Institute of Experimental Immunology, affiliated to EUROIMMUN Medizinische Labordiagnostika AG, Luebeck, Germany
| | - Philipp Arendt
- Institute of Experimental Immunology, affiliated to EUROIMMUN Medizinische Labordiagnostika AG, Luebeck, Germany
| | - Pau Pastor
- Memory Disorders Unit, Department of Neurology, Hospital Universitari Mutua de Terrassa, Terrassa, Spain
- Fundació per a la Recerca Biomèdica i Social Mútua de Terrassa, Terrassa, Spain
| |
Collapse
|
17
|
Compta Y, Revesz T. Neuropathological and Biomarker Findings in Parkinson's Disease and Alzheimer's Disease: From Protein Aggregates to Synaptic Dysfunction. JOURNAL OF PARKINSONS DISEASE 2021; 11:107-121. [PMID: 33325398 PMCID: PMC7990431 DOI: 10.3233/jpd-202323] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
There is mounting evidence that Parkinson’s disease (PD) and Alzheimer’s disease (AD) share neuropathological hallmarks, while similar types of biomarkers are being applied to both. In this review we aimed to explore similarities and differences between PD and AD at both the neuropathology and the biomarker levels, specifically focusing on protein aggregates and synapse dysfunction. Thus, amyloid-β peptide (Aβ) and tau lesions of the Alzheimer-type are common in PD and α-synuclein Lewy-type aggregates are frequent findings in AD. Modern neuropathological techniques adding to routine immunohistochemistry might take further our knowledge of these diseases beyond protein aggregates and down to their presynaptic and postsynaptic terminals, with potential mechanistic and even future therapeutic implications. Translation of neuropathological discoveries to the clinic remains challenging. Cerebrospinal fluid (CSF) and positron emission tomography (PET) markers of Aβ and tau have been shown to be reliable for AD diagnosis. Conversely, CSF markers of α-synuclein have not been that consistent. In terms of PET markers, there is no PET probe available for α-synuclein yet, while the AD PET markers range from consistent evidence of their specificity (amyloid imaging) to greater uncertainty of their reliability due to off-target binding (tau imaging). CSF synaptic markers are attractive, still needing more evidence, which currently suggests those might be non-specific markers of disease progression. It can be summarized that there is neuropathological evidence that protein aggregates of AD and PD are present both at the soma and the synapse. Thus, a number of CSF and PET biomarkers beyond α-synuclein, tau and Aβ might capture these different faces of protein-related neurodegeneration. It remains to be seen what the longitudinal outcomes and the potential value as surrogate markers of these biomarkers are.
Collapse
Affiliation(s)
- Yaroslau Compta
- Parkinson's Disease & Movement Disorders Unit, Neurology Service, Hospital Clínic / IDIBAPS / CIBERNED, Barcelona, Catalonia, Spain.,Institut de Neurociències, Maextu's excellence center, University of Barcelona, Barcelona, Catalonia, Spain
| | - Tamas Revesz
- Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, UK.,Reta Lila Weston Institute of Neurological Studies, UCL Queen Square Institute of Neurology, London, UK.,Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, UK
| |
Collapse
|
18
|
Clinical Utility of the Pathogenesis-Related Proteins in Alzheimer's Disease. Int J Mol Sci 2020; 21:ijms21228661. [PMID: 33212853 PMCID: PMC7698353 DOI: 10.3390/ijms21228661] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/05/2020] [Accepted: 11/13/2020] [Indexed: 12/16/2022] Open
Abstract
Research on the Aβ cascade and alternations of biomarkers in neuro-inflammation, synaptic dysfunction, and neuronal injury followed by Aβ have progressed. But the question is how to use the biomarkers. Here, we examine the evidence and pathogenic implications of protein interactions and the time order of alternation. After the deposition of Aβ, the change of tau, neurofilament light chain (NFL), and neurogranin (Ng) is the main alternation and connection to others. Neuro-inflammation, synaptic dysfunction, and neuronal injury function is exhibited prior to the structural and metabolic changes in the brain following Aβ deposition. The time order of such biomarkers compared to the tau protein is not clear. Despite the close relationship between biomarkers and plaque Aβ deposition, several factors favor one or the other. There is an interaction between some proteins that can predict the brain amyloid burden. The Aβ cascade hypothesis could be the pathway, but not all subjects suffer from Alzheimer's disease (AD) within a long follow-up, even with very elevated Aβ. The interaction of biomarkers and the time order of change require further research to identify the right subjects and right molecular target for precision medicine therapies.
Collapse
|
19
|
Mazzucchi S, Palermo G, Campese N, Galgani A, Della Vecchia A, Vergallo A, Siciliano G, Ceravolo R, Hampel H, Baldacci F. The role of synaptic biomarkers in the spectrum of neurodegenerative diseases. Expert Rev Proteomics 2020; 17:543-559. [PMID: 33028119 DOI: 10.1080/14789450.2020.1831388] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION The quest for reliable fluid biomarkers tracking synaptic disruption is supported by the evidence of a tight association between synaptic density and cognitive performance in neurodegenerative diseases (NDD), especially Alzheimer's disease (AD). AREAS COVERED Neurogranin (Ng) is a post-synaptic protein largely expressed in neurons involved in the memory networks. Currently, Ng measured in CSF is the most promising synaptic biomarker. Several studies show Ng elevated in AD dementia with a hippocampal phenotype as well as in MCI individuals who progress to AD. Ng concentrations are also increased in Creutzfeldt Jacob Disease where widespread and massive synaptic disintegration takes place. Ng does not discriminate Parkinson's disease from atypical parkinsonisms, nor is it altered in Huntington disease. CSF synaptosomal-associated protein 25 (SNAP-25) and synaptotagmin-1 (SYT-1) are emerging candidates. EXPERT OPINION CSF Ng revealed a role as a diagnostic and prognostic biomarker in NDD. Ng increase seems to be very specific for typical AD phenotype, probably for a prevalent hippocampal involvement. Synaptic biomarkers may serve different context-of-use in AD and other NDD including prognosis, diagnosis, and tracking synaptic damage - a critical pathophysiological mechanism in NDD - thus representing reliable tools for a precision medicine-oriented approach to NDD.
Collapse
Affiliation(s)
- Sonia Mazzucchi
- Department of Clinical and Experimental Medicine, University of Pisa , Pisa, Italy
| | - Giovanni Palermo
- Department of Clinical and Experimental Medicine, University of Pisa , Pisa, Italy
| | - Nicole Campese
- Department of Clinical and Experimental Medicine, University of Pisa , Pisa, Italy
| | - Alessandro Galgani
- Department of Clinical and Experimental Medicine, University of Pisa , Pisa, Italy
| | | | - Andrea Vergallo
- Sorbonne University, GRC N° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de L'hôpital , Paris, France.,Brain & Spine Institute (ICM), INSERM U1127 , Paris, France.,Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Pitié-Salpêtrière Hospital, AP-HP , Paris, France
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa , Pisa, Italy
| | - Roberto Ceravolo
- Department of Clinical and Experimental Medicine, University of Pisa , Pisa, Italy
| | - Harald Hampel
- Sorbonne University, GRC N° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de L'hôpital , Paris, France
| | - Filippo Baldacci
- Department of Clinical and Experimental Medicine, University of Pisa , Pisa, Italy.,Sorbonne University, GRC N° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de L'hôpital , Paris, France
| |
Collapse
|
20
|
Hampel H, Lista S, Vanmechelen E, Zetterberg H, Giorgi FS, Galgani A, Blennow K, Caraci F, Das B, Yan R, Vergallo A. β-Secretase1 biological markers for Alzheimer's disease: state-of-art of validation and qualification. Alzheimers Res Ther 2020; 12:130. [PMID: 33066807 PMCID: PMC7566058 DOI: 10.1186/s13195-020-00686-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 09/15/2020] [Indexed: 01/09/2023]
Abstract
β-Secretase1 (BACE1) protein concentrations and rates of enzyme activity, analyzed in human bodily fluids, are promising candidate biological markers for guidance in clinical trials investigating BACE1 inhibitors to halt or delay the dysregulation of the amyloid-β pathway in Alzheimer's disease (AD). A robust body of evidence demonstrates an association between cerebrospinal fluid/blood BACE1 biomarkers and core pathophysiological mechanisms of AD, such as brain protein misfolding and aggregration, neurodegeneration, and synaptic dysfunction.In pharmacological trials, BACE1 candidate biomarkers may be applied to a wide set of contexts of use (CoU), including proof of mechanism, dose-finding, response and toxicity dose estimation. For clinical CoU, BACE1 biomarkers show good performance for prognosis and disease prediction.The roadmap toward validation and qualification of BACE1 biomarkers requires standardized pre-analytical and analytical protocols to reduce inter-site variance that may have contributed to inconsistent results.BACE1 biomarker-drug co-development programs, including biomarker-guided outcomes and endpoints, may support the identification of sub-populations with a higher probability to benefit from BACE1 inhibitors with a reduced risk of adverse effects, in line with the evolving precision medicine paradigm.
Collapse
Affiliation(s)
- Harald Hampel
- Sorbonne University, GRC no 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Simone Lista
- Sorbonne University, GRC no 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
- Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l'hôpital, F-75013, Paris, France
- Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Boulevard de l'hôpital, F-75013, Paris, France
| | | | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & 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
| | - Filippo Sean Giorgi
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Alessandro Galgani
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | - Filippo Caraci
- Department of Drug Sciences, University of Catania, Catania, Italy
- Oasi Research Institute-IRCCS, Troina, Italy
| | - Brati Das
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Riqiang Yan
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Andrea Vergallo
- Sorbonne University, GRC no 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France.
| |
Collapse
|
21
|
Mavroudis IA, Petridis F, Chatzikonstantinou S, Kazis D. A meta-analysis on CSF neurogranin levels for the diagnosis of Alzheimer's disease and mild cognitive impairment. Aging Clin Exp Res 2020; 32:1639-1646. [PMID: 31463927 DOI: 10.1007/s40520-019-01326-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 08/14/2019] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Neurogranin is a postsynaptic protein involved in long-term potentiation and synaptic plasticity. Recent studies have shown increased neurogranin levels in the cerebrospinal fluid of Alzheimer's disease patients, and in patients with mild cognitive impairment. METHOD We searched the online databases for studies on neurogranin cerebrospinal fluid levels in Alzheimer's disease, mild cognitive impairment and other neurodegenerative disorders, and we did a meta-analysis to clarify whether this can be a reliable biomarker for the diagnosis of Alzheimer's disease, and the discrimination from other disorders. RESULTS The present meta-analysis showed that neurogranin CSF levels are significantly higher in AD patients compared to NC [SMD: 268.26, 95% CI (143.47, 393.04), Z = 4.21, P = 0.0001], MCI [SMD: 23.45 (15.97, 30.92), Z = 6.15, P < 0.00001], FTD [SMD: 1.91 (0.92, 2.89), Z = 3.80, P < 0.0001], but no significant difference was found between AD and LBD [SMD: 138.51 (- 14.92, 291.95), Z = 1.77, P = 0.08]. Comparison of stable MCI and MCI that progressed to AD showed significantly higher levels in the CSF of MCI patients who progressed to AD, compared to stable MCI patients [SMD: 230.84 (12.54, 449.14), Z = 2.07, P = 0.04]. Neurogranin can also be a useful biomarker for the differentiation MCI and NC, but not between MCI and FTD or LBD. CONCLUSION Neurogranin could be added to the panel of existing biomarkers for a more accurate diagnosis and progress of AD and assessment of underlying pathological changes in the brain.
Collapse
|
22
|
Tible M, Sandelius Å, Höglund K, Brinkmalm A, Cognat E, Dumurgier J, Zetterberg H, Hugon J, Paquet C, Blennow K. Dissection of synaptic pathways through the CSF biomarkers for predicting Alzheimer disease. Neurology 2020; 95:e953-e961. [PMID: 32586895 DOI: 10.1212/wnl.0000000000010131] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 02/15/2020] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To assess the ability of a combination of synaptic CSF biomarkers to separate Alzheimer disease (AD) and non-AD disorders and to help in the differential diagnosis between neurocognitive diseases. METHODS This was a retrospective cross-sectional monocentric study. All participants explored with CSF assessments for neurocognitive decline were invited to participate. After complete clinical and imaging evaluations, 243 patients were included. CSF synaptic (GAP-43, neurogranin, SNAP-25 total, SNAP-25aa40, synaptotagmin-1) and AD biomarkers were blindly quantified with ELISA or mass spectrometry. Statistical analysis compared CSF levels between the various groups of AD dementias (n = 81), mild cognitive impairment (MCI)-AD (n = 30), other MCI (n = 49), other dementias (OD) (n = 49), and neurologic controls (n = 35) and their discriminatory powers. RESULTS All synaptic biomarkers were significantly increased in patients with MCI-AD and AD-dementia compared to the other groups. All synaptic biomarkers could efficiently discriminate AD dementias from OD (AUC ≥0.80). All but synaptotagmin were also able to discriminate patients with MCI-AD from controls (area under the curve [AUC] ≥0.85) and those with AD dementias from controls (AUC ≥0.80). Overall, CSF SNAP-25aa40 had the highest discriminative power (AUC 0.93 between patients with AD dementias and controls or OD, AUC 0.90 between those with MCI-AD and controls). Higher levels were associated with 2 alleles of APOE ε4. CONCLUSION All synaptic biomarkers tested had a good discriminatory power to distinguish patients with AD abnormal CSF from those with non-AD disorders. SNAP25aa40 demonstrated the highest power to discriminate AD CSF-positive patients from patients without AD and neurologic controls in this cohort. CLASSIFICATION OF EVIDENCE This retrospective study provides Class II evidence that CSF synaptic biomarkers discriminate patients with AD from those without AD.
Collapse
Affiliation(s)
- Marion Tible
- From the Université de Paris INSERM U1144 (M.T., E.C., J.D., J.H., C.P.), France; Clinical Neurochemistry Laboratory (A.S., K.H., A.B., H.Z., K.B.), Sahlgrenska University Hospital; Institute of Neuroscience and Physiology (A.S., K.H., A.B., H.Z., K.B.), Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; and Center of Cognitive Neurology (E.C., J.D., J.H., C.P.), Lariboisière Fernand-Widal Hospital, APHP, Paris, France
| | - Åsa Sandelius
- From the Université de Paris INSERM U1144 (M.T., E.C., J.D., J.H., C.P.), France; Clinical Neurochemistry Laboratory (A.S., K.H., A.B., H.Z., K.B.), Sahlgrenska University Hospital; Institute of Neuroscience and Physiology (A.S., K.H., A.B., H.Z., K.B.), Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; and Center of Cognitive Neurology (E.C., J.D., J.H., C.P.), Lariboisière Fernand-Widal Hospital, APHP, Paris, France
| | - Kina Höglund
- From the Université de Paris INSERM U1144 (M.T., E.C., J.D., J.H., C.P.), France; Clinical Neurochemistry Laboratory (A.S., K.H., A.B., H.Z., K.B.), Sahlgrenska University Hospital; Institute of Neuroscience and Physiology (A.S., K.H., A.B., H.Z., K.B.), Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; and Center of Cognitive Neurology (E.C., J.D., J.H., C.P.), Lariboisière Fernand-Widal Hospital, APHP, Paris, France
| | - Ann Brinkmalm
- From the Université de Paris INSERM U1144 (M.T., E.C., J.D., J.H., C.P.), France; Clinical Neurochemistry Laboratory (A.S., K.H., A.B., H.Z., K.B.), Sahlgrenska University Hospital; Institute of Neuroscience and Physiology (A.S., K.H., A.B., H.Z., K.B.), Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; and Center of Cognitive Neurology (E.C., J.D., J.H., C.P.), Lariboisière Fernand-Widal Hospital, APHP, Paris, France
| | - Emmanuel Cognat
- From the Université de Paris INSERM U1144 (M.T., E.C., J.D., J.H., C.P.), France; Clinical Neurochemistry Laboratory (A.S., K.H., A.B., H.Z., K.B.), Sahlgrenska University Hospital; Institute of Neuroscience and Physiology (A.S., K.H., A.B., H.Z., K.B.), Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; and Center of Cognitive Neurology (E.C., J.D., J.H., C.P.), Lariboisière Fernand-Widal Hospital, APHP, Paris, France
| | - Julien Dumurgier
- From the Université de Paris INSERM U1144 (M.T., E.C., J.D., J.H., C.P.), France; Clinical Neurochemistry Laboratory (A.S., K.H., A.B., H.Z., K.B.), Sahlgrenska University Hospital; Institute of Neuroscience and Physiology (A.S., K.H., A.B., H.Z., K.B.), Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; and Center of Cognitive Neurology (E.C., J.D., J.H., C.P.), Lariboisière Fernand-Widal Hospital, APHP, Paris, France
| | - Henrik Zetterberg
- From the Université de Paris INSERM U1144 (M.T., E.C., J.D., J.H., C.P.), France; Clinical Neurochemistry Laboratory (A.S., K.H., A.B., H.Z., K.B.), Sahlgrenska University Hospital; Institute of Neuroscience and Physiology (A.S., K.H., A.B., H.Z., K.B.), Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; and Center of Cognitive Neurology (E.C., J.D., J.H., C.P.), Lariboisière Fernand-Widal Hospital, APHP, Paris, France
| | - Jacques Hugon
- From the Université de Paris INSERM U1144 (M.T., E.C., J.D., J.H., C.P.), France; Clinical Neurochemistry Laboratory (A.S., K.H., A.B., H.Z., K.B.), Sahlgrenska University Hospital; Institute of Neuroscience and Physiology (A.S., K.H., A.B., H.Z., K.B.), Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; and Center of Cognitive Neurology (E.C., J.D., J.H., C.P.), Lariboisière Fernand-Widal Hospital, APHP, Paris, France
| | - Claire Paquet
- From the Université de Paris INSERM U1144 (M.T., E.C., J.D., J.H., C.P.), France; Clinical Neurochemistry Laboratory (A.S., K.H., A.B., H.Z., K.B.), Sahlgrenska University Hospital; Institute of Neuroscience and Physiology (A.S., K.H., A.B., H.Z., K.B.), Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; and Center of Cognitive Neurology (E.C., J.D., J.H., C.P.), Lariboisière Fernand-Widal Hospital, APHP, Paris, France.
| | - Kaj Blennow
- From the Université de Paris INSERM U1144 (M.T., E.C., J.D., J.H., C.P.), France; Clinical Neurochemistry Laboratory (A.S., K.H., A.B., H.Z., K.B.), Sahlgrenska University Hospital; Institute of Neuroscience and Physiology (A.S., K.H., A.B., H.Z., K.B.), Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; and Center of Cognitive Neurology (E.C., J.D., J.H., C.P.), Lariboisière Fernand-Widal Hospital, APHP, Paris, France
| |
Collapse
|
23
|
Liu W, Lin H, He X, Chen L, Dai Y, Jia W, Xue X, Tao J, Chen L. Neurogranin as a cognitive biomarker in cerebrospinal fluid and blood exosomes for Alzheimer's disease and mild cognitive impairment. Transl Psychiatry 2020; 10:125. [PMID: 32350238 PMCID: PMC7190828 DOI: 10.1038/s41398-020-0801-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 03/12/2020] [Accepted: 03/25/2020] [Indexed: 12/31/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder with clinical, biological, and pathological features occurring along a continuum from normal to end-stage disease. Currently, the diagnosis of AD depends on clinical assessments and post-mortem neuropathology, which is unbenefited early diagnosis and progressive monitoring. In recent years, clinical studies have reported that the level of cerebrospinal fluid (CSF) and blood neurogranin (Ng) are closely related to the occurrence and subsequent progression of AD. Therefore, the study used meta-analysis to identify the CSF and blood Ng levels for the development of diagnosis biomarker of patients with AD and mild cognitive impairment (MCI). We searched the Pubmed, Embase, Cochrane Library, and Web of Science databases. A total of 24 articles eligible for inclusion and exclusion criteria were assessed, including 4661 individuals, consisting of 1518 AD patients, 1501 MCI patients, and 1642 healthy control subjects. The level of CSF Ng significantly increased in patients with AD and MCI compared with healthy control subjects (SMD: 0.84 [95% CI: 0.70-0.98], P < 0.001; SMD: 0.53 [95% CI: 0.40-0.66], P = 0.008), and higher in AD patients than in MCI patients (SMD: 0.18 [95% CI: 0.07-0.30], P = 0.002), and CSF Ng level of patients with MCI-AD who progressed from MCI to AD was significantly higher than that of patients with stable MCI (sMCI) (SMD: 0.71 [95% CI: 0.25-1.16], P = 0.002). Moreover, the concentration of Ng in blood plasma exosomes of patients with AD and MCI was lower than that of healthy control subjects (SMD: -6.657 [95% CI: -10.558 to -2.755], P = 0.001; and SMD: -3.64 [95% CI: -6.50 to -0.78], P = 0.013), and which in patients with AD and MCI-AD were also lower than those in patients with sMCI (P < 0.001). Furthermore, regression analysis showed a negative relationship between MMSE scores and CSF Ng levels in MCI patients (slope = -0.249 [95% CI: -0.003 to -0.495], P = 0.047). Therefore, the Ng levels increased in CSF, but decreased in blood plasma exosomes of patients with AD and MCI-AD, and highly associated with cognitive declines. These findings provide the clinical evidence that CSF and blood exosomes Ng can be used as a cognitive biomarker for AD and MCI-AD, and further studies are needed to define the specific range of Ng values for diagnosis at the different stages of AD.
Collapse
Affiliation(s)
- Weilin Liu
- grid.411504.50000 0004 1790 1622College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian China ,grid.266902.90000 0001 2179 3618Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - Huawei Lin
- grid.411504.50000 0004 1790 1622College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian China
| | - Xiaojun He
- grid.411504.50000 0004 1790 1622The Academy of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian China
| | - Lewen Chen
- grid.411504.50000 0004 1790 1622The Academy of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian China
| | - Yaling Dai
- grid.411504.50000 0004 1790 1622The Academy of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian China
| | - Weiwei Jia
- grid.411504.50000 0004 1790 1622The Academy of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian China
| | - Xiehua Xue
- grid.411504.50000 0004 1790 1622Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian China
| | - Jing Tao
- grid.411504.50000 0004 1790 1622College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian China
| | - Lidian Chen
- The Academy of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China.
| |
Collapse
|
24
|
Galasko D, Xiao M, Xu D, Smirnov D, Salmon DP, Dewit N, Vanbrabant J, Jacobs D, Vanderstichele H, Vanmechelen E, Alzheimer's Disease Neuroimaging Initiative (ADNI), Worley P. Synaptic biomarkers in CSF aid in diagnosis, correlate with cognition and predict progression in MCI and Alzheimer's disease. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2019; 5:871-882. [PMID: 31853477 PMCID: PMC6911971 DOI: 10.1016/j.trci.2019.11.002] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
INTRODUCTION Amyloid, Tau, and neurodegeneration biomarkers can stage Alzheimer's Disease (AD). Synaptic biomarkers may help track cognition. METHODS In cognitively normal controls, Mild Cognitive Impairment (MCI) and AD, we investigated CSF biomarkers in relation to cognitive measures and as predictors of cognitive and global decline. RESULTS There were 90 normal controls (mean age 73.0, 58% women), 57 MCI (mean age 74.3, 35% women), and 46 AD (mean age 70.7, 41% women). CSF Aβ1-42 and Neuronal Pentraxin 2 (NPTX2) were decreased, and CSF Tau, neurogranin, and SNAP25 increased in AD versus controls. Aβ1-42/Tau or NPTX2/Tau discriminated AD and controls best. NPTX2/Tau correlated strongly with cognition in AD and MCI and predicted a 2-3-year decline. We replicated findings in the ADNI cohort. DISCUSSION CSF synaptic biomarkers, particularly NPTX2, which regulates synaptic homeostasis, relate to cognition and predict progression in AD beyond Aβ1-42 and Tau. This is relevant for prognosis and clinical trials.
Collapse
Affiliation(s)
- Douglas Galasko
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Meifang Xiao
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Desheng Xu
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Denis Smirnov
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - David P. Salmon
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | | | | | | | | | | | | | - Paul Worley
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|