1
|
Cumplido-Mayoral I, Sánchez-Benavides G, Vilor-Tejedor N, López-Martos D, Brugulat-Serrat A, Milà-Alomà M, Falcon C, Cacciaglia R, Minguillón C, Fauria K, Kollmorgen G, Quijano-Rubio C, Molinuevo JL, Grau-Rivera O, Suárez-Calvet M, Vilaplana V, Gispert JD. Neuroimaging-derived biological brain age and its associations with glial reactivity and synaptic dysfunction cerebrospinal fluid biomarkers. Mol Psychiatry 2025:10.1038/s41380-025-02961-x. [PMID: 40221600 DOI: 10.1038/s41380-025-02961-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 01/07/2025] [Accepted: 03/18/2025] [Indexed: 04/14/2025]
Abstract
Magnetic resonance Imaging (MRI)-derived brain-age prediction is a promising biomarker of biological brain aging. Accelerated brain aging has been found in Alzheimer's disease (AD) and other neurodegenerative diseases. However, no previous studies have investigated the relationship between specific pathophysiological pathways in AD and biological brain aging. Here, we studied whether glial reactivity and synaptic dysfunction are associated with biological brain aging in the earliest stages of the Alzheimer's continuum, and if these mechanisms are differently associated with AD-related cortical atrophy. We further evaluated their effects on cognitive decline. We included 380 cognitively unimpaired individuals from the ALFA+ study, for which we computed their brain-age deltas by subtracting chronological age from their brain age predicted by machine learning algorithms. We studied the cross-sectional linear associations between brain-age delta and cerebrospinal fluid (CSF) biomarkers of synaptic dysfunction (neurogranin, GAP43, synaptotagmin-1, SNAP25, and α-synuclein), glial reactivity (sTREM2, YKL-40, GFAP, and S100b) and inflammation (interleukin-6). We also studied the cross-sectional linear associations between AD signature and these CSF biomarkers, We further evaluated the mechanisms linking baseline brain-age delta and longitudinal cognitive decline by performing mediation analyses. To reproduce our findings on an independent cohort, we included 152 cognitively unimpaired and 310 mild cognitive impaired (MCI) individuals from the ADNI study. We found that higher CSF sTREM2 was associated with a younger brain-age after adjusting for AD pathology, both in ALFA+ cognitively unimpaired and in ADNI MCI individuals. Furthermore, we found that CSF sTREM2 fully mediated the link between older brain-age and cognitive decline in ALFA+. In summary, we showed that the protective microglial state reflected by higher CSF sTREM2 has a beneficial impact on biological brain aging that may partly explains the variability in cognitive decline in early AD stages, independently of AD pathology.
Collapse
Affiliation(s)
- Irene Cumplido-Mayoral
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
- Hospital del Mar Medical Research Institute, Barcelona, Spain
| | - Gonzalo Sánchez-Benavides
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Hospital del Mar Medical Research Institute, Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Natalia Vilor-Tejedor
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Department of Genetics, Radboud University, Nijmegen, Netherlands
| | - David López-Martos
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Hospital del Mar Medical Research Institute, Barcelona, Spain
| | - Anna Brugulat-Serrat
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Hospital del Mar Medical Research Institute, Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
- Global Brain Health Institute., San Francisco, CA, USA
| | - Marta Milà-Alomà
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Hospital del Mar Medical Research Institute, Barcelona, Spain
- Department of Veterans Affairs Medical Center, Northern California Institute for Research and Education (NCIRE), San Francisco, CA, USA
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Carles Falcon
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Hospital del Mar Medical Research Institute, Barcelona, Spain
| | - Raffaele Cacciaglia
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Hospital del Mar Medical Research Institute, Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Carolina Minguillón
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Hospital del Mar Medical Research Institute, Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Karine Fauria
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | | | | | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
| | - Oriol Grau-Rivera
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Hospital del Mar Medical Research Institute, Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
- Servei de Neurologia, Hospital del Mar, Barcelona, Spain
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.
- Hospital del Mar Medical Research Institute, Barcelona, Spain.
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain.
- Servei de Neurologia, Hospital del Mar, Barcelona, Spain.
| | - Verónica Vilaplana
- Department of Signal Theory and Communications, Universitat Politècnica de Catalunya, Barcelona, Spain
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.
- Hospital del Mar Medical Research Institute, Barcelona, Spain.
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain.
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| |
Collapse
|
2
|
Zhang B, Zhang D, Chen K, Wu T. Silibinin's role in counteracting neuronal apoptosis and synaptic dysfunction in Alzheimer's disease models. Apoptosis 2025; 30:861-879. [PMID: 39833635 DOI: 10.1007/s10495-024-02073-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/27/2024] [Indexed: 01/22/2025]
Abstract
This study investigates silibinin's capacity to mitigate Alzheimer's disease (AD) pathologies with a particular emphasis on its effects on apoptosis and synaptic dysfunction in AD models. Employing APP/PS1 transgenic mice and SH-SY5Y neuroblastoma cell lines, our research assessed the efficacy of silibinin in reducing amyloid-beta (Aβ) deposition, neuroinflammation, and neuronal apoptosis. Our results demonstrate that silibinin significantly decreases Aβ accumulation and neuroinflammation and robustly inhibits apoptosis in neuronal cells. Additionally, silibinin enhances the expression of synaptic proteins, thereby supporting synaptic integrity. Through network pharmacology analysis, we identified potential targets of silibinin in Aβ metabolism and synaptic functions. Mechanistically, our findings suggest that silibinin promotes neuronal survival predominantly via the modulation of the Fyn/GluN2B/CaMKIIα signaling pathway, which protects against Aβ1-42-induced apoptosis. These insights highlight silibinin's potential as a therapeutic agent for AD, particularly its role in reducing neuronal apoptosis and maintaining synaptic function.
Collapse
Affiliation(s)
- Baohui Zhang
- Department of Neurobiology, China Medical University, Shenyang, 110122, China
- Journal Center, China Medical University, Shenyang, 110122, China
| | - Di Zhang
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China
| | - Keyan Chen
- Department of Laboratory Animal Science, China Medical University, No. 77, Puhe Road, Shenbei New District, Shenyang, Liaoning Province, 110122, China.
| | - Tengfei Wu
- Department of Laboratory Animal Science, China Medical University, No. 77, Puhe Road, Shenbei New District, Shenyang, Liaoning Province, 110122, China.
| |
Collapse
|
3
|
Wolner SH, Gleerup HS, Musaeus CS, Høgh P, Ashton NJ, Brinkmalm A, Nilsson J, Grötschel L, Zetterberg H, Blennow K, Hasselbalch SG, Walls AB, Simonsen AH. Synaptosomal-Associated Protein 25 kDA (SNAP-25) Levels in Cerebrospinal Fluid: Implications for Alzheimer's Disease Diagnosis and Monitoring. Synapse 2025; 79:e70010. [PMID: 39912369 PMCID: PMC11800177 DOI: 10.1002/syn.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 01/05/2025] [Accepted: 01/19/2025] [Indexed: 02/07/2025]
Abstract
Synaptic degeneration has been linked to cognitive decline. The presynaptic protein, synaptosomal-associated protein 25 kDA (SNAP-25), is crucial for synaptic transmission and has been suggested as a biomarker in Alzheimer's disease (AD). In the current study, we investigated the ability of SNAP-25 to differentiate between heterogenous dementia etiologies and whether SNAP-25 could be a staging marker in AD. SNAP-25 in the cerebrospinal fluid (CSF) from a retrospective (n = 187) and a prospective (n = 134) cohort was investigated with immunoprecipitation mass spectrometry (IP-MS) and single-molecule array (Simoa), respectively. Both cohorts consisted of healthy controls (HC) and patients with cognitive decline of different etiologies. CSF SNAP-25 concentration was higher in AD and non-neurodegenerative diseases (i.e., vascular dementia) compared with controls but did not differ between AD and non-AD neurodegenerative diseases. We found a trend toward an association between SNAP-25 and disease burden when comparing HC, mild cognitive impairment due to AD, and AD. CSF SNAP-25 concentrations were strongly associated with CSF phosphorylated tau (p-tau) concentrations, thus strengthening the link between synaptic dysfunction and tau pathophysiology in AD. Our initial findings suggest that SNAP-25 may be a potential biomarker for differentiating AD from dementia due to other etiologies. However, due to the significant association between SNAP-25 and p-tau proteins, the clinical utility of SNAP-25 as a diagnostic biomarker for AD may be limited, while SNAP-25 may be useful for monitoring disease progression or treatment response.
Collapse
Affiliation(s)
- Sofia Hjorth Wolner
- Danish Dementia Research Centre, Department of NeurologyCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
| | - Helena Sophia Gleerup
- Danish Dementia Research Centre, Department of NeurologyCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
| | - Christian Sandøe Musaeus
- Danish Dementia Research Centre, Department of NeurologyCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
| | - Peter Høgh
- Regional Dementia Research Centre, Department of NeurologyZealand University HospitalRoskildeDenmark
- Department of Clinical MedicineUniversity of CopenhagenCopenhagenDenmark
| | - Nicholas J. Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and PhysiologySahlgrenska Academy at the University of GothenburgMölndalSweden
- Institute of Psychiatry, Psychology and Neuroscience Maurice Wohl Institute Clinical Neuroscience InstituteKing's College LondonLondonUK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS FoundationLondonUK
- Centre for Age‐Related MedicineStavanger University HospitalStavangerNorway
| | - Ann Brinkmalm
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and PhysiologySahlgrenska Academy at the University of GothenburgMölndalSweden
| | - Johanna Nilsson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and PhysiologySahlgrenska Academy at the University of GothenburgMölndalSweden
| | - Lana Grötschel
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and PhysiologySahlgrenska Academy at the University of GothenburgMölndalSweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and PhysiologySahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseUCL Institute of Neurology, Queen SquareLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Hong Kong Center for Neurodegenerative DiseasesClear Water BayHong KongChina
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public HealthUniversity of Wisconsin–MadisonMadisonWisconsinUSA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and PhysiologySahlgrenska Academy at the University of GothenburgMölndalSweden
- Institute of Psychiatry, Psychology and Neuroscience Maurice Wohl Institute Clinical Neuroscience InstituteKing's College LondonLondonUK
- Paris Brain Institute, ICM, Pitié‐Salpêtrière HospitalSorbonne UniversityParisFrance
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain DisordersUniversity of Science and Technology of China and First Affiliated Hospital of USTCHefeiChina
| | - Steen Gregers Hasselbalch
- Danish Dementia Research Centre, Department of NeurologyCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
- Department of Clinical MedicineUniversity of CopenhagenCopenhagenDenmark
| | - Anne Byriel Walls
- Department of Drug Design and Pharmacology, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- Capital Region Hospital PharmacyRigshospitaletCopenhagenDenmark
| | - Anja Hviid Simonsen
- Danish Dementia Research Centre, Department of NeurologyCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
| |
Collapse
|
4
|
Lee AJ, Howard E, Saltiel N, Hayes JP, Hayes SM. Neuronal and glial dysfunction, white matter hyperintensities and cognition in ageing and Alzheimer's disease. Brain Commun 2025; 7:fcaf068. [PMID: 39995657 PMCID: PMC11848269 DOI: 10.1093/braincomms/fcaf068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 01/08/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
This cross-sectional study examined associations between multiple fluid biomarkers of neuronal and glial dysfunction (plasma neurofilament light chain, CSF growth-associated protein 43 and CSF soluble triggering receptor expressed on myeloid cells 2), total white matter hyperintensity volume and episodic memory and executive function performance in the context of Alzheimer's disease biomarker status. A total of 563 participants (mean age = 71.9 years, standard deviation = 7.2) from the Alzheimer's Disease Neuroimaging Initiative were classified by the amyloid-β/tau/neurodegeneration framework into no Alzheimer's disease pathology (n = 176), suspected non-Alzheimer's disease pathophysiology (n = 87) or Alzheimer's disease continuum (n = 300) groups. Participants completed baseline neuropsychological assessment, plasma/CSF biomarker collection and MRI. Analyses explored the relative contributions of biomarkers to episodic memory and executive function performance and whether relationships varied by amyloid-β/tau/neurodegeneration group status. Across all participants, neurofilament light chain (β ^ = -0.14, P < 0.001) and growth-associated protein 43 (β ^ = -0.13, P < 0.001) were the strongest biomarkers associated with episodic memory performance, such that greater levels were associated with worse episodic memory. There was a group by growth-associated protein 43 interaction with episodic memory: greater growth-associated protein 43 was associated with lower episodic memory performance in participants classified as Alzheimer's disease continuum relative to the no Alzheimer's disease pathology group (β ^ = -0.26, P < 0.001). No robust associations between biomarkers and executive function performance or between soluble triggering receptor expressed on myeloid cells 2, white matter hyperintensity volume and cognition were observed. Biomarkers of neuro-axonal injury and synaptic dysfunction may independently contribute to episodic memory performance across participants with differing amyloid-β/tau/neurodegeneration profiles. Growth-associated protein 43 may predict worse episodic memory performance in participants with greater Alzheimer's disease pathology. These biomarkers of neuronal dysfunction may serve as domain-specific cognitive correlates in the context of Alzheimer's disease biomarker status.
Collapse
Affiliation(s)
- Ann J Lee
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA
| | - Erica Howard
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA
| | - Nicole Saltiel
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA
| | - Jasmeet P Hayes
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA
- Chronic Brain Injury Initiative, The Ohio State University, Columbus, OH 43210, USA
| | - Scott M Hayes
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA
- Chronic Brain Injury Initiative, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
5
|
Xu ZX, Zhang JL, Li FZ, Xu B, Xia J, Wang P, Xie GJ. AnMei decoction ameliorates cognitive impairment in rats with chronic sleep deprivation by mitigating hippocampal neuroinflammation and restoring synaptic architecture. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119101. [PMID: 39537118 DOI: 10.1016/j.jep.2024.119101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 11/16/2024]
Abstract
SIGNIFICANCE OF ETHNOPHARMACOLOGY AnMei Decoction (AMD) is a renowned herbal prescription that has been widely demonstrated to have positive therapeutic effects on sleep disorders, depression, and cognitive impairments. However, the molecular mechanisms underlying AMD's resistance to sleep deprivation-induced cognitive impairment remain to be further investigated. RESEARCH OBJECTIVE To clarify whether AMD may alleviate neuroinflammation by inhibiting NLRP3/Caspase1 signaling pathway and repair neuronal damage by regulating BDNF/TrkB pathway, thereby improving cognitive dysfunction in rats with chronic sleep deprivation. MATERIALS AND METHODS LC-MS/MS was used to detect the active components in AMD. After behavioral tests, HE staining, Nissl staining, immunofluorescence, immunohistochemistry, transmission electron microscopy, and Golgi staining were performed to assess the effects of AMD on chronic sleep deprivation. Western blot was used to detect the expression of hippocampal proteins NLRP3, Caspase-1, BDNF, p-TrkB, TrkB, Bax, Bcl-2, GAP43, PSD95, SNAP25, SYN, STX1A, and VAMP2. Hippocampal transcriptome sequencing was employed to observe differentially expressed genes after AMD intervention. RESULTS A total of 15 active components were identified from the AMD extract. AMD effectively improved the exploration and learning and memory abilities of sleep-deprived rats. AMD reduced neuroinflammation by inhibiting the NLRP3/Caspase-1 pathway and repaired neuronal damage by regulating the BDNF/TrkB pathway. Simultaneously, AMD upregulated the expression of BDNF, p-TrkB, Bcl-2, GAP43, PSD95, SNAP25, SYN, STX1A, and VAMP2 proteins and inhibited the expression of NLRP3, Caspase-1, and Bax proteins. Analysis of GO and KEGG pathway enrichment for the differentially expressed inflammation-related pathways may be involved in the therapeutic mechanism of AMD on sleep deprivation. CONCLUSION AMD can effectively inhibit the NLRP3/Caspase1 signaling pathway to alleviate neuroinflammation, regulate the BDNF/TrkB pathway to maintain hippocampal neuronal viability, repair synaptic structural damage, and improve cognitive impairment in the sleep deprivation model.
Collapse
Affiliation(s)
- Zi-Xuan Xu
- Basic Medicine College, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Jun-Lu Zhang
- Basic Medicine College, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Fei-Zhou Li
- Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430006, China.
| | - Bo Xu
- Basic Medicine College, Hubei University of Chinese Medicine, Wuhan, 430065, China; Engineering Research Center, Hubei University of Chinese Medicine, Wuhan, 430065, China; Hubei Shizhen Laboratory, Wuhan, 430006, China.
| | - Jing Xia
- Engineering Research Center, Hubei University of Chinese Medicine, Wuhan, 430065, China; Hubei Shizhen Laboratory, Wuhan, 430006, China.
| | - Ping Wang
- Engineering Research Center, Hubei University of Chinese Medicine, Wuhan, 430065, China; Hubei Shizhen Laboratory, Wuhan, 430006, China.
| | - Guang-Jing Xie
- Engineering Research Center, Hubei University of Chinese Medicine, Wuhan, 430065, China; Hubei Shizhen Laboratory, Wuhan, 430006, China; College of Physical Education and Health, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| |
Collapse
|
6
|
Xu B, Xiao K, Jia X, Cao R, Liang D, A R, Zhang W, Li C, Gao L, Chen C, Shi Q, Dong X. β-synuclein in cerebrospinal fluid as a potential biomarker for distinguishing human prion diseases from Alzheimer's and Parkinson's disease. Alzheimers Res Ther 2025; 17:39. [PMID: 39920821 PMCID: PMC11806663 DOI: 10.1186/s13195-025-01688-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 01/30/2025] [Indexed: 02/09/2025]
Abstract
BACKGROUND β-synuclein (β-syn), mainly expressed in central nerve system, is one of the biomarkers in cerebrospinal fluid (CSF) and blood for synaptic damage, which has been reported to be elevated in CSF and blood of the patients of prion diseases (PrDs). METHODS We analyzed 314 CSF samples from patients in China National Surveillance for CJD. The diagnostic groups of the 223 patients with PrDs included sporadic Creutzfeldt-Jacob disease (sCJD), genetic CJD (gCJD), fatal familial insomnia (FFI) and Gerstmann-Straussler-Scheinker (GSS). 91 patients with non-PrDs comprised Alzheimer's disease (AD), Parkinson's disease (PD), viral encephalitis (VE) or autoimmune encephalitis (AE) were enrolled in the control groups. The CSF β-syn levels were measured by a commercial microfluidic ELISA. The Mann-Whitney U test and Kruskal-Wallis H test were employed to analyze two or more sets of continuous variables. Multiple linear regression was also performed to evaluate the factors for CSF β-syn levels. Receiver operating characteristics (ROC) curves and area under the curve (AUC) values were used to assess the diagnostic performance of β-syn. RESULTS The median of β-syn levels (2074 pg/ml; IQR: 691 to 4332) of all PrDs was significantly higher than that of non-PrDs group (504 pg/ml; IQR: 126 to 3374). The CSF β-syn values in the cohorts of sCJD, T188K-gCJD, E200K-gCJD and P102L-GSS were remarkably higher than that of the group of AD + PD, but similar as that of the group of VE + AE. The elevated CSF β-syn in sCJD and gCJD cases was statistically associated with CSF 14-3-3 positive and appearance of mutism. ROC curve analysis identified satisfied performance for distinguishing from AD + PD, with high AUC values in sCJD (0.7640), T188K-gCJD (0.8489), E200K-gCJD (0.8548), P102L-GSS (0.7689) and D178N-FFI (0.7210), respectively. CONCLUSION Our data here indicate that CSF β-syn is a potential biomarker for distinguishing PrDs (gCJD, sCJD and GSS) from AD and PD, but is much less efficient from VE and AE. These findings have critical implications for early diagnosis and monitoring of synaptic integrity in prion diseases.
Collapse
Grants
- 2021SKLID504, 2019SKLID401, 2019SKLID603, and 2021SKLID503 SKLID Development Grants from the National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, China CDC
- 2021SKLID504, 2019SKLID401, 2019SKLID603, and 2021SKLID503 SKLID Development Grants from the National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, China CDC
- 2021SKLID504, 2019SKLID401, 2019SKLID603, and 2021SKLID503 SKLID Development Grants from the National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, China CDC
- 2021SKLID504, 2019SKLID401, 2019SKLID603, and 2021SKLID503 SKLID Development Grants from the National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, China CDC
- 2021SKLID504, 2019SKLID401, 2019SKLID603, and 2021SKLID503 SKLID Development Grants from the National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, China CDC
- 2021SKLID504, 2019SKLID401, 2019SKLID603, and 2021SKLID503 SKLID Development Grants from the National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, China CDC
- 2021SKLID504, 2019SKLID401, 2019SKLID603, and 2021SKLID503 SKLID Development Grants from the National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, China CDC
- 2021SKLID504, 2019SKLID401, 2019SKLID603, and 2021SKLID503 SKLID Development Grants from the National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, China CDC
- 2021SKLID504, 2019SKLID401, 2019SKLID603, and 2021SKLID503 SKLID Development Grants from the National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, China CDC
- 2021SKLID504, 2019SKLID401, 2019SKLID603, and 2021SKLID503 SKLID Development Grants from the National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, China CDC
- 2021SKLID504, 2019SKLID401, 2019SKLID603, and 2021SKLID503 SKLID Development Grants from the National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, China CDC
- 2021SKLID504, 2019SKLID401, 2019SKLID603, and 2021SKLID503 SKLID Development Grants from the National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, China CDC
Collapse
Affiliation(s)
- Bing Xu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Kang Xiao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiaoxi Jia
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Rundong Cao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Donglin Liang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ruhan A
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Weiwei Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Chunjie Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Liping Gao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Cao Chen
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Qi Shi
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.
- China Academy of Chinese Medical Sciences, Beijing, China.
| | - Xiaoping Dong
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China.
- China Academy of Chinese Medical Sciences, Beijing, China.
- Shanghai Institute of Infectious Disease and Biosafety, Shanghai, China.
| |
Collapse
|
7
|
Taddei RN, E Duff K. Synapse vulnerability and resilience underlying Alzheimer's disease. EBioMedicine 2025; 112:105557. [PMID: 39891995 PMCID: PMC11833146 DOI: 10.1016/j.ebiom.2025.105557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/24/2024] [Accepted: 01/03/2025] [Indexed: 02/03/2025] Open
Abstract
Synapse preservation is key for healthy cognitive ageing, and synapse loss represents a critical anatomical basis of cognitive dysfunction in Alzheimer's disease (AD), predicting dementia onset, severity, and progression. Synapse loss is viewed as a primary pathologic event, preceding neuronal loss and brain atrophy in AD. Synapses may, therefore, represent one of the earliest and clinically most meaningful targets of the neuropathologic processes driving AD dementia. The synapse loss in AD is highly selective and targets particularly vulnerable synapses while leaving others, termed resilient, largely unaffected. Yet, the anatomic and molecular hallmarks of the vulnerable and resilient synapse populations and their association with AD neuropathologic changes (e.g. amyloid-β plaques and tau tangles) and memory dysfunction remain poorly understood. Characterising the selectively vulnerable and resilient synapses in AD may be key to understanding the mechanisms of cognitive preservation versus loss and enable the development of robust biomarkers and disease-modifying therapies for dementia.
Collapse
Affiliation(s)
- Raquel N Taddei
- Neurology Department, Massachusetts General Hospital, Harvard Medical School, Boston, USA; UK Dementia Research Institute at UCL, Institute of Neurology, University College London, UK.
| | - Karen E Duff
- UK Dementia Research Institute at UCL, Institute of Neurology, University College London, UK
| |
Collapse
|
8
|
Cao M, Chen J, Chen G, Ouyang W, Tong J. Preoperative Blood-Brain Barrier Integrity Influence on the Impact of Anesthesia and Surgery on Mice Brain. Anesth Analg 2024:00000539-990000000-01077. [PMID: 39689007 DOI: 10.1213/ane.0000000000007330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
BACKGROUND Brain homeostasis imbalance, characterized by cognitive dysfunction and delirium, frequently occurs in the elderly after surgery. Investigating why this complication only affects part of patients undergoing the same surgery, and anesthesia remains intriguing. This study tested the role of preoperative blood-brain barrier (BBB) integrity in the occurrence of postoperative brain homeostasis imbalance using mice with conditional BBB damage. METHODS Preoperative BBB breakdown was induced in End-SCL-Cre-ctnnb1fl//fl (iCKO) mice by administering tamoxifen (intraperitoneal [i.p.]). This breakdown was assessed using Evans Blue (EB) leakage and immunoglobulin G (IgG) staining. Postoperative brain homeostasis imbalance was evaluated through the Novel Object Recognition test, the Barnes Maze, and neuroinflammation tests. Synapse loss was detected by colabeling synaptophysin and PSD-95, followed by Western blotting. The role of astrocytes in this pathogenesis was evaluated by comparing cognitive behaviors, hippocampal gene expression, and astrocytic phagocytosis of synaptophysin in iCKO mice with and without genetic inhibition of perioperative astrocyte activity. RESULTS Tamoxifen treatment (30 mg/kg/d) induced BBB breakdown of iCKO mice in a time-dependent manner (analysis of variance [ANOVA] for time, P = .0006), but not in their littermate control mice (nCKO, P > .999). A 3-day tamoxifen treatment induced slight BBB breakdown (EB leakage: 95% confidence interval [CI], 13.9-204.8, P = .013; IgG level: 95% CI, 12.6-51.4: P = .001), but did not cause significant cognitive impairment in the Novel Object Recognition test in iCKO mice (95% CI, -7.99 to 6.12; P > .999). Anesthesia and surgery-induced significant cognitive impairment (all P < .0001 for the Novel Object Recognition test, Barnes Maze test), neuroinflammation, and synaptic loss in iCKO mice with 3-day tamoxifen treatment, but not in nCKO mice with the same treatment. Inhibiting astrocyte activity alleviated the impact of anesthesia and surgery on cognitive function (all P < .0001 for the Novel Object Recognition test, Barnes Maze test), gene expression, and synapse pruning in iCKO mice with 3-day tamoxifen treatment. CONCLUSIONS Preoperative BBB integrity influences the impact of anesthesia and surgery on the brain, with astrocytes modulating this effect. These findings partly explain the heterogeneity in the occurrence of postoperative brain homeostasis imbalance.
Collapse
Affiliation(s)
- Mengya Cao
- Department of Anesthesiology, Hunan Province Key Laboratory of Brain Homeostasis, Third Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Jie Chen
- Department of Anesthesiology, Hunan Province Key Laboratory of Brain Homeostasis, Third Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Gong Chen
- Department of Anesthesiology, Hunan Province Key Laboratory of Brain Homeostasis, Third Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
- Department of Anesthesiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Wen Ouyang
- Department of Anesthesiology, Hunan Province Key Laboratory of Brain Homeostasis, Third Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
- Department of Anesthesiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Jianbin Tong
- Department of Anesthesiology, Hunan Province Key Laboratory of Brain Homeostasis, Third Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
- Department of Anesthesiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
- Brain Research Center, Central South University, Changsha, Hunan, P.R. China
| |
Collapse
|
9
|
Zuglianello C, França AP, de Souza BS, Agnes JP, Prediger RD, Lemos-Senna E. Intranasal administration of dextran-pramlintide polyelectrolyte complex-coated nanoemulsions improves cognitive impairments in a mouse model of Alzheimer's disease. Int J Biol Macromol 2024; 281:136158. [PMID: 39362444 DOI: 10.1016/j.ijbiomac.2024.136158] [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: 05/01/2024] [Revised: 09/15/2024] [Accepted: 09/28/2024] [Indexed: 10/05/2024]
Abstract
Nasal delivery has emerged as a non-invasive route to administer drugs for brain delivery. In particular, polyelectrolyte complexes-based nanocarriers have been demonstrated to be advantageous for nasal delivery of peptide drugs and vaccines. Pramlintide (Pram) is a peptide that emerges as a novel neuroprotective strategy to modify the pathogenesis of Alzheimer's disease (AD). In this study, we examined the effects of the intranasal administration of dextran-pramlintide polyelectrolyte complex-coated nanoemulsions (PEC-NEDexS/Pram) in an experimental model of AD induced by intracerebroventricular (i.c.v.) infusion of amyloid-beta (Aβ1-42) peptide in mice. PEC-NEDexS/Pram displayed droplet size lower than 200 nm and a negatively charged surface. The locomotor activity of the animals was not affected by the i.c.v. Aβ1-42 injection or Pram treatment. On the other hand, the intranasal administration of PEC-NEDexS/Pram at a dose of 100 μg/day for 14 consecutive days restored the impairment induced by Aβ1-42 injection in the discriminative learning and the short-term spatial reference memory of mice. However, Pram treatment did not alter the Aβ1-42-induced anhedonic behavior, oxidative stress parameters, or the pre-synaptic SNAP-25 and post-synaptic PSD-95 levels in the hippocampus and prefrontal cortex. These findings indicate cognitive-enhancing properties of intranasal Pram administration in an animal model of AD.
Collapse
Affiliation(s)
- Carine Zuglianello
- Laboratório de Farmacotécnica, Departamento de Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal de Santa Catarina, Florianópolis 88040-370, Santa Catarina, Brazil
| | - Angela P França
- Laboratório Experimental de Doenças Neurodegenerativas, Centro de Ciências Biológicas, Departamento de Farmacologia, Universidade Federal de Santa Catarina, Florianópolis 88049-900, Santa Catarina, Brazil
| | - Bruna S de Souza
- Laboratório Experimental de Doenças Neurodegenerativas, Centro de Ciências Biológicas, Departamento de Farmacologia, Universidade Federal de Santa Catarina, Florianópolis 88049-900, Santa Catarina, Brazil
| | - Jonathan P Agnes
- Laboratório de Farmacologia e Bioquímica do Câncer, Centro de Ciências Biológicas, Departamento de Farmacologia, Universidade Federal de Santa Catarina, Florianópolis 88049-900, SC, Brazil
| | - Rui D Prediger
- Laboratório Experimental de Doenças Neurodegenerativas, Centro de Ciências Biológicas, Departamento de Farmacologia, Universidade Federal de Santa Catarina, Florianópolis 88049-900, Santa Catarina, Brazil
| | - Elenara Lemos-Senna
- Laboratório de Farmacotécnica, Departamento de Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal de Santa Catarina, Florianópolis 88040-370, Santa Catarina, Brazil.
| |
Collapse
|
10
|
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
|
11
|
Van Eldik LJ, Siemers ER, Collins EC, Gold M, Henley D, Johannsen P, Möbius HJ, Shulman M, Zhou J, Carrillo M, Weber C. Understanding recent advances in non-amyloid/non-tau (NANT) biomarkers and therapeutic targets in Alzheimer's disease. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2024; 10:e70014. [PMID: 39748839 PMCID: PMC11694522 DOI: 10.1002/trc2.70014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 08/09/2024] [Indexed: 01/04/2025]
Abstract
The Alzheimer's disease (AD) research community continues to make great strides in expanding approaches for early detection and treatment of the disease, including recent advances in our understanding of fundamental AD pathophysiology beyond the classical targets: beta-amyloid and tau. Recent clinical trial readouts implicate a variety of non-amyloid/non-tau (NANT) approaches that show promise in slowing cognitive decline for people with AD. The Alzheimer's Association Research Roundtable (AARR) meeting held on December 13-14, 2022, reviewed the current state of NANT targets on underlying AD pathophysiology and their contribution to cognitive decline, the current data on a diverse range of NANT biomarkers and therapeutic targets, and the integration of NANT concepts in clinical trial designs. Participants also discussed the current definition of therapies that target underlying AD pathophysiology, what endpoints best define what is considered a meaningful change beyond the current approved definition for clinical efficacy, and how the recent NANT findings should inform the development of future guidelines for AD classification and personalized treatment strategies. Highlights The Alzheimer's Association Research Roundtable (AARR) convened leaders from industry, academia, and government to review the current state of non-beta amyloid and non-tau (NANT) targets on underlying Alzheimer's disease (AD) pathophysiology.The totality of scientific and clinical evidence supports the hypothesis that emerging NANT targets play a role in cognitive decline and neurodegeneration in AD. New biomarkers based on NANT targets must be globally developed and implemented with specific consideration of fluid biomarkers as a cost-effective clinical option, to ensure better, more equitable treatment options for AD.
Collapse
Affiliation(s)
- Linda J. Van Eldik
- Sanders‐Brown Center on Aging and Department of NeuroscienceUniversity of KentuckyLexingtonKentuckyUSA
| | | | - Emily C. Collins
- Imaging Research and DevelopmentEli Lilly and CompanyIndianapolisIndianaUSA
| | | | - David Henley
- Research and DevelopmentJanssen Pharmaceuticals, Inc.TitusvilleNew JerseyUSA
| | | | | | - Melanie Shulman
- Alzheimer's and Dementia Research UnitBiogenCambridgeMassachusettsUSA
| | - Jin Zhou
- Eisai, Inc., Clinical ResearchNeurology Business GroupNutleyNew JerseyUSA
| | | | | |
Collapse
|
12
|
Jurasova V, Andel R, Katonova A, Veverova K, Zuntychova T, Horakova H, Vyhnalek M, Kolarova T, Matoska V, Blennow K, Hort J. CSF neurogranin levels as a biomarker in Alzheimer's disease and frontotemporal lobar degeneration: a cross-sectional analysis. Alzheimers Res Ther 2024; 16:199. [PMID: 39242539 PMCID: PMC11378641 DOI: 10.1186/s13195-024-01566-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 08/24/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND There is initial evidence suggesting that biomarker neurogranin (Ng) may distinguish Alzheimer's disease (AD) from other neurodegenerative diseases. Therefore, we assessed (a) the discriminant ability of cerebrospinal fluid (CSF) Ng levels to distinguish between AD and frontotemporal lobar degeneration (FTLD) pathology and between different stages within the same disease, (b) the relationship between Ng levels and cognitive performance in both AD and FTLD pathology, and (c) whether CSF Ng levels vary by apolipoprotein E (APOE) polymorphism in the AD continuum. METHODS Participants with subjective cognitive decline (SCD) (n = 33), amnestic mild cognitive impairment (aMCI) due to AD (n = 109), AD dementia (n = 67), MCI due to FTLD (n = 25), and FTLD dementia (n = 29) were recruited from the Czech Brain Aging Study. One-way analysis of covariance (ANCOVA) assessed Ng levels in diagnostic subgroups. Linear regressions evaluated the relationship between CSF Ng levels, memory scores, and APOE polymorphism. RESULTS Ng levels were higher in aMCI-AD patients compared to MCI-FTLD (F[1, 134] = 15.16, p < .001), and in AD-dementia compared to FTLD-dementia (F[1, 96] = 4.60, p = .029). Additionally, Ng levels were higher in FTLD-dementia patients compared to MCI-FTLD (F[1, 54]= 4.35, p = .034), lower in SCD participants compared to aMCI-AD (F[1, 142] = 10.72, p = .001) and AD-dementia (F[1, 100] = 20.90, p < .001), and did not differ between SCD participants and MCI-FTLD (F[1, 58]= 1.02, p = .491) or FTLD-dementia (F[1, 62]= 2.27, p = .051). The main effect of diagnosis across the diagnostic subgroups on Aβ1-42/Ng ratio was significant too (F[4, 263]=, p < .001). We found a non-significant association between Ng levels and memory scores overall (β=-0.25, p = .154) or in AD diagnostic subgroups, and non-significant differences in this association between overall AD APOE ε4 carriers and non-carriers (β=-0.32, p = .358). CONCLUSIONS In this first study to-date to assess MCI and dementia due to AD or FTLD within one study, elevated CSF Ng appears to be an early biomarker of AD-related impairment, but its role as a biomarker appears to diminish after dementia diagnosis, whereby dementia-related underlying processes in AD and FTLD may begin to merge. The Aβ1-42/Ng ratio discriminated AD from FTLD patients better than Ng alone. CSF Ng levels were not related to memory in AD or FTLD, suggesting that Ng may be a marker of the biological signs of disease state rather than cognitive deficits.
Collapse
Affiliation(s)
- Vanesa Jurasova
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University, Motol University Hospital, Prague, Czech Republic.
| | - Ross Andel
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University, Motol University Hospital, Prague, Czech Republic
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ, USA
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Alzbeta Katonova
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University, Motol University Hospital, Prague, Czech Republic
| | - Katerina Veverova
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University, Motol University Hospital, Prague, Czech Republic
| | - Terezie Zuntychova
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University, Motol University Hospital, Prague, Czech Republic
| | - Hana Horakova
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University, Motol University Hospital, Prague, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Martin Vyhnalek
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University, Motol University Hospital, Prague, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Tereza Kolarova
- Department of Clinical Biochemistry, Hematology and Immunology, Homolka Hospital, Prague, Czech Republic
| | - Vaclav Matoska
- Department of Clinical Biochemistry, Hematology and Immunology, Homolka Hospital, Prague, Czech Republic
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Jakub Hort
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University, Motol University Hospital, Prague, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| |
Collapse
|
13
|
Knorr U, Simonsen AH, Nilsson J, Brinkmalm A, Zetterberg H, Blennow K, Knudsen MB, Forman J, Hasselbalch SG, Kessing LV. Cerebrospinal fluid synaptic biomarker changes in bipolar disorder - A longitudinal case-control study. J Affect Disord 2024; 358:250-259. [PMID: 38723679 DOI: 10.1016/j.jad.2024.05.034] [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: 01/18/2024] [Revised: 05/03/2024] [Accepted: 05/04/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND This exploratory study investigated cerebrospinal fluid (CSF) synaptic protein biomarkers in bipolar disorder (BD), aiming to highlight the neurobiological basis of the disorder. With shared cognitive impairment features between BD and Alzheimer's disease, and considering increased dementia risk in BD patients, the study explores potential connections. METHODS Fifty-nine well-characterized patients with BD and thirty-seven healthy control individuals were examined and followed for one year. Synaptic proteins encompassing neuronal pentraxins (NPTX)1, NPTX2, and NPTX-receptor, 14-3-3 protein family epsilon, and zeta/delta, activating protein-2 complex subunit beta, synucleins beta-synuclein and gamma-synuclein, complexin-2, phosphatidylethanolamine-binding protein 1, rab GDP dissociation inhibitor alpha, and syntaxins 1B and 7 were measured in CSF using a microflow liquid chromatography-mass spectrometric multiple reaction monitoring set-up. Biomarker levels were compared between BD and HC and in BD before, during, and after mood episodes. RESULTS The synaptic proteins revealed no statistically significant differences between BD and HC, neither at baseline, one-year follow-up, or in terms of changes from baseline to follow-up. Moreover, the CSF synaptic protein levels in patients with BD were unaltered compared to baseline when they stabilized in euthymia following an affective episode and at one-year follow-up. LIMITATION It is uncertain what the CSF biomarker concentrations reflect since we yet do not know the mechanisms of release of these proteins, and we are uncertain of what increased or decreased levels reflect. CONCLUSION This first-ever investigation of a panel of CSF protein biomarkers of synaptic dysfunction in patients with BD and HC individuals found no statistically significant differences cross-sectionally or longitudinally.
Collapse
Affiliation(s)
- Ulla Knorr
- Copenhagen Affective Disorder Research Center (CADIC), Psychiatric Center Copenhagen, Department Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Denmark.
| | - Anja Hviid Simonsen
- Danish Dementia Research Center, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Johanna Nilsson
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Ann Brinkmalm
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, University College London, Queen Square, London, United Kingdom; UK Dementia Research Institute University College London, London, United Kingdom; Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China; Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France; Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, PR China
| | - Mark Bech Knudsen
- Section of Biostatistics, Department of Public Health, University of Copenhagen, Denmark
| | - Julie Forman
- Section of Biostatistics, Department of Public Health, University of Copenhagen, Denmark
| | - Steen Gregers Hasselbalch
- Danish Dementia Research Center, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Denmark
| | - Lars Vedel Kessing
- Copenhagen Affective Disorder Research Center (CADIC), Psychiatric Center Copenhagen, Department Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Denmark
| |
Collapse
|
14
|
Azzolini F, Dolcetti E, Bruno A, Rovella V, Centonze D, Buttari F. Physical exercise and synaptic protection in human and pre-clinical models of multiple sclerosis. Neural Regen Res 2024; 19:1768-1771. [PMID: 38103243 PMCID: PMC10960279 DOI: 10.4103/1673-5374.389359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/11/2023] [Accepted: 09/14/2023] [Indexed: 12/18/2023] Open
Abstract
In multiple sclerosis, only immunomodulatory and immunosuppressive drugs are recognized as disease-modifying therapies. However, in recent years, several data from pre-clinical and clinical studies suggested a possible role of physical exercise as disease-modifying therapy in multiple sclerosis. Current evidence is sparse and often conflicting, and the mechanisms underlying the neuroprotective and antinflammatory role of exercise in multiple sclerosis have not been fully elucidated. Data, mainly derived from pre-clinical studies, suggest that exercise could enhance long-term potentiation and thus neuroplasticity, could reduce neuroinflammation and synaptopathy, and dampen astrogliosis and microgliosis. In humans, most trials focused on direct clinical and MRI outcomes, as investigating synaptic, neuroinflammatory, and pathological changes is not straightforward compared to animal models. The present review analyzed current evidence and limitations in research concerning the potential disease-modifying therapy effects of exercise in multiple sclerosis in animal models and human studies.
Collapse
Affiliation(s)
| | | | | | - Valentina Rovella
- Department of System Medicine, University of Tor Vergata, Rome, Italy
| | - Diego Centonze
- Unit of Neurology, IRCCS Neuromed, Pozzilli, Italy
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Fabio Buttari
- Unit of Neurology, IRCCS Neuromed, Pozzilli, Italy
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| |
Collapse
|
15
|
Nilsson J, Pichet Binette A, Palmqvist S, Brum WS, Janelidze S, Ashton NJ, Spotorno N, Stomrud E, Gobom J, Zetterberg H, Brinkmalm A, Blennow K, Hansson O. Cerebrospinal fluid biomarker panel for synaptic dysfunction in a broad spectrum of neurodegenerative diseases. Brain 2024; 147:2414-2427. [PMID: 38325331 PMCID: PMC11224614 DOI: 10.1093/brain/awae032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 12/31/2023] [Accepted: 01/21/2024] [Indexed: 02/09/2024] Open
Abstract
Synaptic dysfunction and degeneration is likely the key pathophysiology for the progression of cognitive decline in various dementia disorders. Synaptic status can be monitored by measuring synaptic proteins in CSF. In this study, both known and new synaptic proteins were investigated and compared as potential biomarkers of synaptic dysfunction, particularly in the context of Alzheimer's disease (AD). Seventeen synaptic proteins were quantified in CSF using two different targeted mass spectrometry assays in the prospective Swedish BioFINDER-2 study. The study included 958 individuals, characterized as having mild cognitive impairment (MCI, n = 205), AD dementia (n = 149) and a spectrum of other neurodegenerative diseases (n = 171), in addition to cognitively unimpaired individuals (CU, n = 443). Synaptic protein levels were compared between diagnostic groups and their associations with cognitive decline and key neuroimaging measures (amyloid-β-PET, tau-PET and cortical thickness) were assessed. Among the 17 synaptic proteins examined, 14 were specifically elevated in the AD continuum. SNAP-25, 14-3-3 zeta/delta, β-synuclein, and neurogranin exhibited the highest discriminatory accuracy in differentiating AD dementia from controls (areas under the curve = 0.81-0.93). SNAP-25 and 14-3-3 zeta/delta also had the strongest associations with tau-PET, amyloid-β-PET and cortical thickness at baseline and were associated with longitudinal changes in these imaging biomarkers [β(standard error, SE) = -0.056(0.0006) to 0.058(0.005), P < 0.0001]. SNAP-25 was the strongest predictor of progression to AD dementia in non-demented individuals (hazard ratio = 2.11). In contrast, neuronal pentraxins were decreased in all neurodegenerative diseases (except for Parkinson's disease), and NPTX2 showed the strongest associations with subsequent cognitive decline [longitudinal Mini-Mental State Examination: β(SE) = 0.57(0.1), P ≤ 0.0001; and mPACC: β(SE) = 0.095(0.024), P ≤ 0.001] across the AD continuum. Interestingly, utilizing a ratio of the proteins that displayed higher levels in AD, such as SNAP-25 or 14-3-3 zeta/delta, over NPTX2 improved the biomarkers' associations with cognitive decline and brain atrophy. We found 14-3-3 zeta/delta and SNAP-25 to be especially promising as synaptic biomarkers of pathophysiological changes in AD. Neuronal pentraxins were identified as general indicators of neurodegeneration and associated with cognitive decline across various neurodegenerative dementias. Cognitive decline and brain atrophy were best predicted by ratios of SNAP-25/NPTX2 and 14-3-3 zeta/delta/NPTX2.
Collapse
Affiliation(s)
- Johanna Nilsson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, S-431 80 Mölndal, Sweden
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, 211 46 Malmö, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, 211 46 Malmö, Sweden
- Memory Clinic, Skåne University Hospital, 205 02 Malmö, Sweden
| | - Wagner S Brum
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, S-431 80 Mölndal, Sweden
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035-003, Brazil
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, 211 46 Malmö, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, S-431 80 Mölndal, Sweden
- Centre for Age-Related Medicine, Stavanger University Hospital, 4011 Stavanger, Norway
- Department of Old Age Psychiatry, Maurice Wohl Clinical Neuroscience Institute, King’s College London, London SE5 9RX, UK
- NIHR Maudsley Biomedical Research Centre, South London and Maudsley NHS Foundation Trust, London SE5 8AF, UK
| | - Nicola Spotorno
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, 211 46 Malmö, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, 211 46 Malmö, Sweden
- Memory Clinic, Skåne University Hospital, 205 02 Malmö, Sweden
| | - Johan Gobom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, S-431 80 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 431 30 Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, S-431 80 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 431 30 Mölndal, Sweden
- Fluid Biomarker Laboratory, UK Dementia Research Institute at UCL, London WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London WC1N 3BG, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Ann Brinkmalm
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, S-431 80 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 431 30 Mölndal, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, S-431 80 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 431 30 Mölndal, Sweden
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, 75646 Paris, France
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei 230036, P.R. China
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, 211 46 Malmö, Sweden
- Memory Clinic, Skåne University Hospital, 205 02 Malmö, Sweden
| |
Collapse
|
16
|
Soares C, Da Ros LU, Machado LS, Rocha A, Lazzarotto G, Carello-Collar G, De Bastiani MA, Ferrari-Souza JP, Lussier FZ, Souza DO, Rosa-Neto P, Pascoal TA, Bellaver B, Zimmer ER. The glutamatergic system in Alzheimer's disease: a systematic review with meta-analysis. Mol Psychiatry 2024; 29:2261-2273. [PMID: 38366114 DOI: 10.1038/s41380-024-02473-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 01/23/2024] [Accepted: 01/31/2024] [Indexed: 02/18/2024]
Abstract
Glutamatergic neurotransmission system dysregulation may play an important role in the pathophysiology of Alzheimer's disease (AD). However, reported results on glutamatergic components across brain regions are contradictory. Here, we conducted a systematic review with meta-analysis to examine whether there are consistent glutamatergic abnormalities in the human AD brain. We searched PubMed and Web of Science (database origin-October 2023) reports evaluating glutamate, glutamine, glutaminase, glutamine synthetase, glutamate reuptake, aspartate, excitatory amino acid transporters, vesicular glutamate transporters, glycine, D-serine, metabotropic and ionotropic glutamate receptors in the AD human brain (PROSPERO #CDRD42022299518). The studies were synthesized by outcome and brain region. We included cortical regions, the whole brain (cortical and subcortical regions combined), the entorhinal cortex and the hippocampus. Pooled effect sizes were determined with standardized mean differences (SMD), random effects adjusted by false discovery rate, and heterogeneity was examined by I2 statistics. The search retrieved 6 936 articles, 63 meeting the inclusion criteria (N = 709CN/786AD; mean age 75/79). We showed that the brain of AD individuals presents decreased glutamate (SMD = -0.82; I2 = 74.54%; P < 0.001) and aspartate levels (SMD = -0.64; I2 = 89.71%; P = 0.006), and reuptake (SMD = -0.75; I2 = 83.04%; P < 0.001. We also found reduced α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPAR)-GluA2/3 levels (SMD = -0.63; I2 = 95.55%; P = 0.046), hypofunctional N-methyl-D-aspartate receptor (NMDAR) (SMD = -0.60; I2 = 91.47%; P < 0.001) and selective reduction of NMDAR-GluN2B subunit levels (SMD = -1.07; I2 = 41.81%; P < 0.001). Regional differences include lower glutamate levels in cortical areas and aspartate levels in cortical areas and in the hippocampus, reduced glutamate reuptake, reduced AMPAR-GluA2/3 in the entorhinal cortex, hypofunction of NMDAR in cortical areas, and a decrease in NMDAR-GluN2B subunit levels in the entorhinal cortex and hippocampus. Other parameters studied were not altered. Our findings show depletion of the glutamatergic system and emphasize the importance of understanding glutamate-mediated neurotoxicity in AD. This study has implications for the development of therapies and biomarkers in AD.
Collapse
Affiliation(s)
- Carolina Soares
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lucas Uglione Da Ros
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Luiza Santos Machado
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Andreia Rocha
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Gabriela Lazzarotto
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Giovanna Carello-Collar
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Marco A De Bastiani
- Graduate Program in Biological Sciences: Pharmacology and Therapeutics, UFRGS, Porto Alegre, Brazil
| | - João Pedro Ferrari-Souza
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Firoza Z Lussier
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Diogo O Souza
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Department of Biochemistry, UFRGS, Porto Alegre, Brazil
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC, Canada
- Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Tharick A Pascoal
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bruna Bellaver
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Eduardo R Zimmer
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.
- Graduate Program in Biological Sciences: Pharmacology and Therapeutics, UFRGS, Porto Alegre, Brazil.
- Department of Biochemistry, UFRGS, Porto Alegre, Brazil.
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.
- Brain Institute of Rio Grande do Sul - Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil.
- Department of Pharmacology, UFRGS, Porto Alegre, Brazil.
| |
Collapse
|
17
|
Wang YT, Ashton NJ, Servaes S, Nilsson J, Woo MS, Pascoal TA, Tissot C, Rahmouni N, Therriault J, Lussier F, Chamoun M, Gauthier S, Brinkmalm A, Zetterberg H, Blennow K, Rosa-Neto P, Benedet AL. The relation of synaptic biomarkers with Aβ, tau, glial activation, and neurodegeneration in Alzheimer's disease. Transl Neurodegener 2024; 13:27. [PMID: 38802928 PMCID: PMC11131272 DOI: 10.1186/s40035-024-00420-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 05/06/2024] [Indexed: 05/29/2024] Open
Affiliation(s)
- Yi-Ting Wang
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Johanna Nilsson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Marcel S Woo
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Neuroimmunology and Multiple Sclerosis (INIMS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tharick A Pascoal
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | | | - Mira Chamoun
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Serge Gauthier
- Alzheimer's Disease Research Unit, The McGill University Research Centre for Studies in Aging, Montreal, McGill University, Montreal, QC, Canada
| | - Ann Brinkmalm
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - 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
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53792, USA
| | - Kaj Blennow
- 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
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Andréa L Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.
| |
Collapse
|
18
|
Bartl M, Nilsson J, Dakna M, Weber S, Schade S, Xylaki M, Fernandes Gomes B, Ernst M, Muntean ML, Sixel-Döring F, Trenkwalder C, Zetterberg H, Brinkmalm A, Mollenhauer B. Lysosomal and synaptic dysfunction markers in longitudinal cerebrospinal fluid of de novo Parkinson's disease. NPJ Parkinsons Dis 2024; 10:102. [PMID: 38760408 PMCID: PMC11101466 DOI: 10.1038/s41531-024-00714-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 04/19/2024] [Indexed: 05/19/2024] Open
Abstract
Lysosomal and synaptic dysfunctions are hallmarks in neurodegeneration and potentially relevant as biomarkers, but data on early Parkinson's disease (PD) is lacking. We performed targeted mass spectrometry with an established protein panel, assessing autophagy and synaptic function in cerebrospinal fluid (CSF) of drug-naïve de novo PD, and sex-/age-matched healthy controls (HC) cross-sectionally (88 PD, 46 HC) and longitudinally (104 PD, 58 HC) over 10 years. Multiple markers of autophagy, synaptic plasticity, and secretory pathways were reduced in PD. We added samples from prodromal subjects (9 cross-sectional, 12 longitudinal) with isolated REM sleep behavior disorder, revealing secretogranin-2 already decreased compared to controls. Machine learning identified neuronal pentraxin receptor and neurosecretory protein VGF as most relevant for discriminating between groups. CSF levels of LAMP2, neuronal pentraxins, and syntaxins in PD correlated with clinical progression, showing predictive potential for motor- and non-motor symptoms as a valid basis for future drug trials.
Collapse
Affiliation(s)
- Michael Bartl
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany.
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Goettingen, Goettingen, Germany.
| | - Johanna Nilsson
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Mohammed Dakna
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
| | - Sandrina Weber
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
| | | | - Mary Xylaki
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
| | - Bárbara Fernandes Gomes
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Marielle Ernst
- Institute of Diagnostic and Interventional Neuroradiology, University Medical Center Goettingen, Goettingen, Germany
| | | | - Friederike Sixel-Döring
- Paracelsus-Elena-Klinik, Kassel, Germany
- Department of Neurology, Philipps-University, Marburg, Germany
| | - Claudia Trenkwalder
- Paracelsus-Elena-Klinik, Kassel, Germany
- Department of Neurosurgery, University Medical Center Goettingen, Goettingen, Germany
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Ann Brinkmalm
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Brit Mollenhauer
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
- Paracelsus-Elena-Klinik, Kassel, Germany
| |
Collapse
|
19
|
Alba-González A, Dragomir EI, Haghdousti G, Yáñez J, Dadswell C, González-Méndez R, Wilson SW, Tuschl K, Folgueira M. Manganese Overexposure Alters Neurogranin Expression and Causes Behavioral Deficits in Larval Zebrafish. Int J Mol Sci 2024; 25:4933. [PMID: 38732149 PMCID: PMC11084468 DOI: 10.3390/ijms25094933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/22/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
Manganese (Mn), a cofactor for various enzyme classes, is an essential trace metal for all organisms. However, overexposure to Mn causes neurotoxicity. Here, we evaluated the effects of exposure to Mn chloride (MnCl2) on viability, morphology, synapse function (based on neurogranin expression) and behavior of zebrafish larvae. MnCl2 exposure from 2.5 h post fertilization led to reduced survival (60%) at 5 days post fertilization. Phenotypical changes affected body length, eye and olfactory organ size, and visual background adaptation. This was accompanied by a decrease in both the fluorescence intensity of neurogranin immunostaining and expression levels of the neurogranin-encoding genes nrgna and nrgnb, suggesting the presence of synaptic alterations. Furthermore, overexposure to MnCl2 resulted in larvae exhibiting postural defects, reduction in motor activity and impaired preference for light environments. Following the removal of MnCl2 from the fish water, zebrafish larvae recovered their pigmentation pattern and normalized their locomotor behavior, indicating that some aspects of Mn neurotoxicity are reversible. In summary, our results demonstrate that Mn overexposure leads to pronounced morphological alterations, changes in neurogranin expression and behavioral impairments in zebrafish larvae.
Collapse
Affiliation(s)
- Anabel Alba-González
- Department of Biology, Faculty of Sciences, University of A Coruña, 15008 A Coruña, Spain; (A.A.-G.); (J.Y.)
- Centro Interdisciplinar de Química y Biología, (CICA), University of A Coruña, 15071 A Coruña, Spain
| | - Elena I. Dragomir
- Department of Cell and Developmental, University College London, London, WC1E 6BT, UK; (E.I.D.); (G.H.); (S.W.W.)
| | - Golsana Haghdousti
- Department of Cell and Developmental, University College London, London, WC1E 6BT, UK; (E.I.D.); (G.H.); (S.W.W.)
| | - Julián Yáñez
- Department of Biology, Faculty of Sciences, University of A Coruña, 15008 A Coruña, Spain; (A.A.-G.); (J.Y.)
- Centro Interdisciplinar de Química y Biología, (CICA), University of A Coruña, 15071 A Coruña, Spain
| | - Chris Dadswell
- School of Life Sciences, University of Sussex, Brighton, BN1 9QJ, UK; (C.D.); (R.G.-M.)
| | - Ramón González-Méndez
- School of Life Sciences, University of Sussex, Brighton, BN1 9QJ, UK; (C.D.); (R.G.-M.)
| | - Stephen W. Wilson
- Department of Cell and Developmental, University College London, London, WC1E 6BT, UK; (E.I.D.); (G.H.); (S.W.W.)
| | - Karin Tuschl
- UCL GOSH Institute of Child Health, University College London, London, WC1N 1EH, UK
| | - Mónica Folgueira
- Department of Biology, Faculty of Sciences, University of A Coruña, 15008 A Coruña, Spain; (A.A.-G.); (J.Y.)
- Centro Interdisciplinar de Química y Biología, (CICA), University of A Coruña, 15071 A Coruña, Spain
| |
Collapse
|
20
|
Wang X, Zhang X, Liu J, Zhang J, Liu C, Cui Y, Song Q, Hou Y, Wang Y, Zhang Q, Zhang Y, Fan Y, Jia J, Wang P. Synaptic vesicle glycoprotein 2 A in serum is an ideal biomarker for early diagnosis of Alzheimer's disease. Alzheimers Res Ther 2024; 16:82. [PMID: 38615037 PMCID: PMC11015666 DOI: 10.1186/s13195-024-01440-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/26/2024] [Indexed: 04/15/2024]
Abstract
BACKGROUND Previous studies have demonstrated that early intervention was the best plan to inhibit the progression of Alzheimer's disease (AD), which relied on the discovery of early diagnostic biomarkers. In this study, synaptic vesicle glycoprotein 2 A (SV2A) was examined to improve the early diagnostic efficiency in AD. METHODS In this study, biomarker testing was performed through the single-molecule array (Simoa). A total of 121 subjects including cognitively unimpaired controls, amnestic mild cognitive impairment (aMCI), AD and other types of dementia underwent cerebrospinal fluid (CSF) SV2A testing; 430 subjects including health controls, aMCI, AD and other types of dementia underwent serum SV2A, glial fibrillary acidic protein (GFAP), neurofilament light chain (NfL) and p-tau217 testing; 92 subjects including aMCI and AD underwent both CSF SV2A and serum SV2A testing; 115 cognitively unimpaired subjects including APOE ε4 carriers and APOE ε4 non-carriers were tested for serum SV2A, GFAP, NfL and p-tau217. Then, the efficacy of SV2A for the early diagnosis of AD and its ability to identify those at high risk of AD from a cognitively unimpaired population were further analyzed. RESULTS Both CSF and serum SV2A significantly and positively correlated with cognitive performance in patients with AD, and their levels gradually decreased with the progression of AD. Serum SV2A demonstrated excellent diagnostic efficacy for aMCI, with a sensitivity of 97.8%, which was significantly higher than those of NfL, GFAP, and p-tau217. The SV2A-positive rates ranged from 92.86 to 100% in aMCI cases that were negative for the above three biomarkers. Importantly, of all the biomarkers tested, serum SV2A had the highest positivity rate (81.82%) in individuals at risk for AD. CONCLUSIONS Serum SV2A was demonstrated to be a novel and ideal biomarker for the early diagnosis of AD, which can effectively distinguish those at high risk of AD in cognitively unimpaired populations.
Collapse
Affiliation(s)
- Xiaoling Wang
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Xiaomin Zhang
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Jing Liu
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Jingjing Zhang
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Congcong Liu
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Yuting Cui
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Qiao Song
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Yuli Hou
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Yaqi Wang
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Qian Zhang
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Yingzhen Zhang
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Yujian Fan
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Jianping Jia
- National Clinical Research Center for Geriatric Disorders, 45 Changchun Street, Beijing, 100053, China.
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China.
- Beijing Key Laboratory of Geriatric Cognitive Disorders, 45 Changchun Street, Beijing, 100053, China.
- Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, 45 Changchun Street, Beijing, 100053, China.
- Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, 45 Changchun Street, Beijing, 100053, China.
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, 45 Changchun Street, Beijing, 100053, China.
| | - Peichang Wang
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Capital Medical University, 45 Changchun Street, Beijing, 100053, China.
- National Clinical Research Center for Geriatric Disorders, 45 Changchun Street, Beijing, 100053, China.
| |
Collapse
|
21
|
Massa F, Martinuzzo C, Gómez de San José N, Pelagotti V, Kreshpa W, Abu-Rumeileh S, Barba L, Mattioli P, Orso B, Brugnolo A, Girtler N, Vigo T, Arnaldi D, Serrati C, Uccelli A, Morbelli S, Chincarini A, Otto M, Pardini M. Cerebrospinal fluid NPTX2 changes and relationship with regional brain metabolism metrics across mild cognitive impairment due to Alzheimer's disease. J Neurol 2024; 271:1999-2009. [PMID: 38157030 DOI: 10.1007/s00415-023-12154-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/08/2023] [Accepted: 12/09/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND Neuronal pentraxin-2 (NPTX2), crucial for synaptic functioning, declines in cerebrospinal fluid (CSF) as cognition deteriorates. The variations of CSF NPTX2 across mild cognitive impairment (MCI) due to Alzheimer's disease (AD) and its association with brain metabolism remain elusive, albeit relevant for patient stratification and pathophysiological insights. METHODS We retrospectively analyzed 49 MCI-AD patients grouped by time until dementia (EMCI, n = 34 progressing within 2 years; LMCI, n = 15 progressing later/stable at follow-up). We analyzed demographic variables, cognitive status (MMSE score), and CSF NPTX2 levels using a commercial ELISA assay in EMCI, LMCI, and a control group of age-/sex-matched individuals with other non-dementing disorders (OND). Using [18F]FDG PET scans for voxel-based analysis, we explored correlations between regional brain metabolism metrics and CSF NPTX2 levels in MCI-AD patients, accounting for age. RESULTS Baseline and follow-up MMSE scores were lower in LMCI than EMCI (p value = 0.006 and p < 0.001). EMCI exhibited significantly higher CSF NPTX2 values than both LMCI (p = 0.028) and OND (p = 0.006). We found a significant positive correlation between NPTX2 values and metabolism of bilateral precuneus in MCI-AD patients (p < 0.005 at voxel level, p < 0.05 with family-wise error correction at the cluster level). CONCLUSIONS Higher CSF NPTX2 in EMCI compared to controls and LMCI suggests compensatory synaptic responses to initial AD pathology. Disease progression sees these mechanisms overwhelmed, lowering CSF NPTX2 approaching dementia. Positive CSF NPTX2 correlation with precuneus glucose metabolism links to AD-related metabolic changes across MCI course. These findings posit CSF NPTX2 as a promising biomarker for both AD staging and progression risk stratification.
Collapse
Affiliation(s)
- Federico Massa
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy.
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Caterina Martinuzzo
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | | | - Virginia Pelagotti
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Wendy Kreshpa
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Samir Abu-Rumeileh
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Lorenzo Barba
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Pietro Mattioli
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Beatrice Orso
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Andrea Brugnolo
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Nicola Girtler
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Tiziana Vigo
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Dario Arnaldi
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | | | - Antonio Uccelli
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Silvia Morbelli
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Health Science (DISSAL), University of Genoa, Genoa, Italy
| | - Andrea Chincarini
- National Institute of Nuclear Physics (INFN), Genoa Section, Genoa, Italy
| | - Markus Otto
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Matteo Pardini
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
22
|
Dong R, Yi N, Jiang D. Advances in single molecule arrays (SIMOA) for ultra-sensitive detection of biomolecules. Talanta 2024; 270:125529. [PMID: 38091745 DOI: 10.1016/j.talanta.2023.125529] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/25/2023] [Accepted: 12/05/2023] [Indexed: 01/27/2024]
Abstract
In the contemporary era of scientific and medical advancements, the accurate and ultra-sensitive detection of proteins, nucleic acids and metabolites plays a pivotal role in disease diagnosis and treatment monitoring. Single-molecule detection technologies play a great role in achieving this goal. In recent years, digital detection methods based on single molecule arrays (SIMOA) have brought groundbreaking contributions to the field of single-molecule detection. By confining the target molecules to femtoliter-sized containers, the SIMOA technology achieves detection sensitivity of attomolar. This review delves into the historical evolution and fundamentals of SIMOA technology, summarizes various approaches to optimize its performance, and describes the applications of SIMOA for the ultrasensitive detection of biomarkers for diseases such as cancer, COVID-19, and neurological disorders, as well as in DNA detection. Currently, some SIMOA technologies have been realized for high-throughput and multiplexed detection. It is believed that SIMOA technology will play a significant role in medical monitoring and disease prevention in the future.
Collapse
Affiliation(s)
- Renkai Dong
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Ning Yi
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Dechen Jiang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China.
| |
Collapse
|
23
|
Wu Y, Hu H, Liu W, Zhao Y, Xie F, Sun Z, Zhang L, Dong H, Wang X, Qian L. Hippocampal Lactate-Infusion Enhances Spatial Memory Correlated with Monocarboxylate Transporter 2 and Lactylation. Brain Sci 2024; 14:327. [PMID: 38671979 PMCID: PMC11048250 DOI: 10.3390/brainsci14040327] [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: 03/12/2024] [Revised: 03/24/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
Lactate has emerged as a key player in regulating neural functions and cognitive processes. Beyond its function as an energy substrate and signal molecule, recent research has revealed lactate to serve as an epigenetic regulator in the brain. However, the molecular mechanisms by which lactate regulates spatial memory and its role in the prevention of cognitive disorders remain unclear. Herein, we injected L-lactate (10 μmol/kg/d for 6 d) into the mouse's hippocampus, followed by the Morris water maze (MWM) test and molecular analyses. Improved spatial memory performances were observed in mice injected with lactate. Besides, lactate upregulated the expression of synaptic proteins post-synaptic density 95 (PSD95), synaptophysin (SYP), and growth associated protein 43 (GAP43) in hippocampal tissues and HT22 cells, suggesting a potential role in synaptic transmission and memory formation. The facilitative role of monocarboxylate transporter 2 (MCT2), a neuron-specific lactate transporter, in this process was confirmed, as MCT2 antagonists attenuated the lactate-induced upregulation of synaptic proteins. Moreover, lactate induced protein lactylation, a post-translational modification, which could be suppressed by MCT2 inhibition. RNA sequencing of lactated-injected hippocampal tissues revealed a comprehensive gene expression profile influenced by lactate, with significant changes in genes associated with transcriptional progress. These data demonstrate that hippocampal lactate injection enhances spatial memory in mice, potentially through the upregulation of synaptic proteins and induction of protein lactylation, with MCT2 playing a crucial role in these processes. Our findings shed light on the multi-faceted role of lactate in neural function and memory regulation, opening new avenues for therapeutic interventions targeting cognitive disorders.
Collapse
Affiliation(s)
- Yuhan Wu
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, China; (Y.W.); (H.H.); (W.L.); (Y.Z.); (F.X.); (Z.S.); (L.Z.); (H.D.)
| | - Hui Hu
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, China; (Y.W.); (H.H.); (W.L.); (Y.Z.); (F.X.); (Z.S.); (L.Z.); (H.D.)
| | - Weiwei Liu
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, China; (Y.W.); (H.H.); (W.L.); (Y.Z.); (F.X.); (Z.S.); (L.Z.); (H.D.)
- College of Public Health, North China University of Science and Technology, Tangshan 063210, China
| | - Yun Zhao
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, China; (Y.W.); (H.H.); (W.L.); (Y.Z.); (F.X.); (Z.S.); (L.Z.); (H.D.)
| | - Fang Xie
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, China; (Y.W.); (H.H.); (W.L.); (Y.Z.); (F.X.); (Z.S.); (L.Z.); (H.D.)
| | - Zhaowei Sun
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, China; (Y.W.); (H.H.); (W.L.); (Y.Z.); (F.X.); (Z.S.); (L.Z.); (H.D.)
| | - Ling Zhang
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, China; (Y.W.); (H.H.); (W.L.); (Y.Z.); (F.X.); (Z.S.); (L.Z.); (H.D.)
| | - Huafeng Dong
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, China; (Y.W.); (H.H.); (W.L.); (Y.Z.); (F.X.); (Z.S.); (L.Z.); (H.D.)
| | - Xue Wang
- Institute of Military Cognition and Brain Sciences, Beijing 100850, China
| | - Lingjia Qian
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, China; (Y.W.); (H.H.); (W.L.); (Y.Z.); (F.X.); (Z.S.); (L.Z.); (H.D.)
| |
Collapse
|
24
|
Saloner R, Staffaroni A, Dammer E, Johnson ECB, Paolillo E, Wise A, Heuer H, Forsberg L, Lago AL, Webb J, Vogel J, Santillo A, Hansson O, Kramer J, Miller B, Li J, Loureiro J, Sivasankaran R, Worringer K, Seyfried N, Yokoyama J, Seeley W, Spina S, Grinberg L, VandeVrede L, Ljubenkov P, Bayram E, Bozoki A, Brushaber D, Considine C, Day G, Dickerson B, Domoto-Reilly K, Faber K, Galasko D, Geschwind D, Ghoshal N, Graff-Radford N, Hales C, Honig L, Hsiung GY, Huey E, Kornak J, Kremers W, Lapid M, Lee S, Litvan I, McMillan C, Mendez M, Miyagawa T, Pantelyat A, Pascual B, Paulson H, Petrucelli L, Pressman P, Ramos E, Rascovsky K, Roberson E, Savica R, Snyder A, Sullivan AC, Tartaglia C, Vandebergh M, Boeve B, Rosen H, Rojas J, Boxer A, Casaletto K. Large-scale network analysis of the cerebrospinal fluid proteome identifies molecular signatures of frontotemporal lobar degeneration. RESEARCH SQUARE 2024:rs.3.rs-4103685. [PMID: 38585969 PMCID: PMC10996789 DOI: 10.21203/rs.3.rs-4103685/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
The pathophysiological mechanisms driving disease progression of frontotemporal lobar degeneration (FTLD) and corresponding biomarkers are not fully understood. We leveraged aptamer-based proteomics (> 4,000 proteins) to identify dysregulated communities of co-expressed cerebrospinal fluid proteins in 116 adults carrying autosomal dominant FTLD mutations (C9orf72, GRN, MAPT) compared to 39 noncarrier controls. Network analysis identified 31 protein co-expression modules. Proteomic signatures of genetic FTLD clinical severity included increased abundance of RNA splicing (particularly in C9orf72 and GRN) and extracellular matrix (particularly in MAPT) modules, as well as decreased abundance of synaptic/neuronal and autophagy modules. The generalizability of genetic FTLD proteomic signatures was tested and confirmed in independent cohorts of 1) sporadic progressive supranuclear palsy-Richardson syndrome and 2) frontotemporal dementia spectrum syndromes. Network-based proteomics hold promise for identifying replicable molecular pathways in adults living with FTLD. 'Hub' proteins driving co-expression of affected modules warrant further attention as candidate biomarkers and therapeutic targets.
Collapse
Affiliation(s)
| | | | | | | | | | - Amy Wise
- University of California, San Francisco
| | | | | | | | | | | | | | | | | | | | - Jingyao Li
- Novartis Institutes for Biomedical Research, Inc
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Suzee Lee
- University of California, San Francisco
| | | | - Corey McMillan
- Department of Neurology, University of Pennsylvania, Philadelphia, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Adam Boxer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | | |
Collapse
|
25
|
Cai H, Pang Y, Ren Z, Fu X, Jia L. Delivering synaptic protein mRNAs via extracellular vesicles ameliorates cognitive impairment in a mouse model of Alzheimer's disease. BMC Med 2024; 22:138. [PMID: 38528511 PMCID: PMC10964680 DOI: 10.1186/s12916-024-03359-2] [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: 07/06/2023] [Accepted: 03/15/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Synaptic dysfunction with reduced synaptic protein levels is a core feature of Alzheimer's disease (AD). Synaptic proteins play a central role in memory processing, learning, and AD pathogenesis. Evidence suggests that synaptic proteins in plasma neuronal-derived extracellular vesicles (EVs) are reduced in patients with AD. However, it remains unclear whether levels of synaptic proteins in EVs are associated with hippocampal atrophy of AD and whether upregulating the expression of these synaptic proteins has a beneficial effect on AD. METHODS In this study, we included 57 patients with AD and 56 healthy controls. We evaluated their brain atrophy through magnetic resonance imaging using the medial temporal lobe atrophy score. We measured the levels of four synaptic proteins, including synaptosome-associated protein 25 (SNAP25), growth-associated protein 43 (GAP43), neurogranin, and synaptotagmin 1 in both plasma neuronal-derived EVs and cerebrospinal fluid (CSF). We further examined the association of synaptic protein levels with brain atrophy. We also evaluated the levels of these synaptic proteins in the brains of 5×FAD mice. Then, we loaded rabies virus glycoprotein-engineered EVs with messenger RNAs (mRNAs) encoding GAP43 and SNAP25 and administered these EVs to 5×FAD mice. After treatment, synaptic proteins, dendritic density, and cognitive function were evaluated. RESULTS The results showed that GAP43, SNAP25, neurogranin, and synaptotagmin 1 were decreased in neuronal-derived EVs but increased in CSF in patients with AD, and the changes corresponded to the severity of brain atrophy. GAP43 and SNAP25 were decreased in the brains of 5×FAD mice. The engineered EVs efficiently and stably delivered these synaptic proteins to the brain, where synaptic protein levels were markedly upregulated. Upregulation of synaptic protein expression could ameliorate cognitive impairment in AD by promoting dendritic density. This marks the first successful delivery of synaptic protein mRNAs via EVs in AD mice, yielding remarkable therapeutic effects. CONCLUSIONS Synaptic proteins are closely related to AD processes. Delivery of synaptic protein mRNAs via EVs stands as a promising effective precision treatment strategy for AD, which significantly advances the current understanding of therapeutic approaches for the disease.
Collapse
Affiliation(s)
- Huimin Cai
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, 45 Changchun St., Beijing, 100053, China
- Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Yana Pang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, 45 Changchun St., Beijing, 100053, China
- Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Ziye Ren
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, 45 Changchun St., Beijing, 100053, China
- Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Xiaofeng Fu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, 45 Changchun St., Beijing, 100053, China
- Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Longfei Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, 45 Changchun St., Beijing, 100053, China.
- Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China.
| |
Collapse
|
26
|
Piel JHA, Bargemann L, Leypoldt F, Wandinger KP, Dargvainiene J. Serum NFL and tau, but not serum UCHL-1 and GFAP or CSF SNAP-25, NPTX2, or sTREM2, correlate with delirium in a 3-year retrospective analysis. Front Neurol 2024; 15:1356575. [PMID: 38566855 PMCID: PMC10985356 DOI: 10.3389/fneur.2024.1356575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 02/26/2024] [Indexed: 04/04/2024] Open
Abstract
Delirium represents a common terminal pathway of heterogeneous neurological conditions characterized by disturbances in consciousness and attention. Contemporary theories highlight the acute impairment of synaptic function and network connectivity, driven by neuroinflammation, oxidative stress, and neurotransmitter imbalances. However, established biomarkers are still missing. Innovative diagnostic techniques, such as single-molecule array analysis, enable the detection of biomarkers in blood at picomolar concentrations. This approach paves the way for deeper insights into delirium and potentially therapeutic targets for tailored medical treatments. In a retrospective 3-year study, we investigated seven biomarkers indicative of neuroaxonal damage [neurofilament light chain (NFL), ubiquitin carboxyl-terminal hydrolase (UCHL-1), and tau protein], microglial activation [glial fibrillary acidic protein (GFAP) and soluble triggering receptor expressed on myeloid cells 2 (sTREM2)], and synaptic dysfunction [synaptosomal-associated protein 25 (SNAP-25) and neuronal pentraxin 2 (NPTX2)]. The analysis of 71 patients with delirium, Alzheimer's disease (AD), and non-AD controls revealed that serum NFL levels are higher in delirium cases compared to both AD and non-AD. This suggests that elevated NFL levels in delirium are not exclusively the result of dementia-related damage. Serum tau levels were also elevated in delirium cases compared to controls. Conversely, cerebrospinal fluid (CSF) SNAP-25 showed higher levels in AD patients compared to controls only. These findings add to the increasing body of evidence suggesting that serum NFL could be a valuable biomarker of neuroaxonal damage in delirium research. Although SNAP-25 and NPTX2 did not exhibit significant differences in delirium, the exploration of synaptic biomarkers remains promising for enhancing our understanding of this condition.
Collapse
Affiliation(s)
| | - Leon Bargemann
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Frank Leypoldt
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel, Germany
- Institute of Clinical Chemistry, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Klaus-Peter Wandinger
- Institute of Clinical Chemistry, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Justina Dargvainiene
- Institute of Clinical Chemistry, University Medical Center Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
27
|
Lista S, Santos-Lozano A, Emanuele E, Mercuri NB, Gabelle A, López-Ortiz S, Martín-Hernández J, Maisto N, Imbimbo C, Caraci F, Imbimbo BP, Zetterberg H, Nisticò R. Monitoring synaptic pathology in Alzheimer's disease through fluid and PET imaging biomarkers: a comprehensive review and future perspectives. Mol Psychiatry 2024; 29:847-857. [PMID: 38228892 DOI: 10.1038/s41380-023-02376-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 12/04/2023] [Accepted: 12/12/2023] [Indexed: 01/18/2024]
Abstract
Alzheimer's disease (AD) is currently constrained by limited clinical treatment options. The initial pathophysiological event, which can be traced back to decades before the clinical symptoms become apparent, involves the excessive accumulation of amyloid-beta (Aβ), a peptide comprised of 40-42 amino acids, in extraneuronal plaques within the brain. Biochemical and histological studies have shown that overaccumulation of Aβ instigates an aberrant escalation in the phosphorylation and secretion of tau, a microtubule-binding axonal protein. The accumulation of hyperphosphorylated tau into intraneuronal neurofibrillary tangles is in turn correlated with microglial dysfunction and reactive astrocytosis, culminating in synaptic dysfunction and neurodegeneration. As neurodegeneration progresses, it gives rise to mild clinical symptoms of AD, which may eventually evolve into overt dementia. Synaptic loss in AD may develop even before tau alteration and in response to possible elevations in soluble oligomeric forms of Aβ associated with early AD. These findings largely rely on post-mortem autopsy examinations, which typically involve a limited number of patients. Over the past decade, a range of fluid biomarkers such as neurogranin, α-synuclein, visinin-like protein 1 (VILIP-1), neuronal pentraxin 2, and β-synuclein, along with positron emission tomography (PET) markers like synaptic vesicle glycoprotein 2A, have been developed. These advancements have facilitated the exploration of how synaptic markers in AD patients correlate with cognitive impairment. However, fluid biomarkers indicating synaptic loss have only been validated in cerebrospinal fluid (CSF), not in plasma, with the exception of VILIP-1. The most promising PET radiotracer, [11C]UCB-J, currently faces significant challenges hindering its widespread clinical use, primarily due to the necessity of a cyclotron. As such, additional research geared toward the exploration of synaptic pathology biomarkers is crucial. This will not only enable their extensive clinical application, but also refine the optimization process of AD pharmacological trials.
Collapse
Affiliation(s)
- Simone Lista
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012, Valladolid, Spain.
| | - Alejandro Santos-Lozano
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012, Valladolid, Spain
- Physical Activity and Health Research Group (PaHerg), Research Institute of the Hospital 12 de Octubre ('imas12'), 28041, Madrid, Spain
| | | | - Nicola B Mercuri
- Experimental Neurology Laboratory, IRCCS Santa Lucia Foundation, 00143, Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Audrey Gabelle
- CMRR, Memory Resources and Research Center, Montpellier University of Excellence i-site, 34295, Montpellier, France
| | - Susana López-Ortiz
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012, Valladolid, Spain
| | - Juan Martín-Hernández
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012, Valladolid, Spain
| | - Nunzia Maisto
- Laboratory of Pharmacology of Synaptic Plasticity, EBRI Rita Levi-Montalcini Foundation, 00143, Rome, Italy
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, 00185, Rome, Italy
| | - Camillo Imbimbo
- Department of Brain and Behavioral Sciences, University of Pavia, 27100, Pavia, Italy
| | - Filippo Caraci
- Department of Drug and Health Sciences, University of Catania, 95125, Catania, Italy
- Neuropharmacology and Translational Neurosciences Research Unit, Oasi Research Institute-IRCCS, 94018, Troina, Italy
| | - Bruno P Imbimbo
- Department of Research and Development, Chiesi Farmaceutici, 43122, Parma, Italy
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, 431 80, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 431 80, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, WC1N, London, UK
- UK Dementia Research Institute at UCL, WC1E 6BT, 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, 53726, WI, USA
| | - Robert Nisticò
- Laboratory of Pharmacology of Synaptic Plasticity, EBRI Rita Levi-Montalcini Foundation, 00143, Rome, Italy.
- School of Pharmacy, University of Rome "Tor Vergata", 00133, Rome, Italy.
| |
Collapse
|
28
|
Dakterzada F, Jové M, Huerto R, Carnes A, Sol J, Pamplona R, Piñol-Ripoll G. Cerebrospinal fluid neutral lipids predict progression from mild cognitive impairment to Alzheimer's disease. GeroScience 2024; 46:683-696. [PMID: 37999901 PMCID: PMC10828158 DOI: 10.1007/s11357-023-00989-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 10/13/2023] [Indexed: 11/25/2023] Open
Abstract
Genetic, metabolic, and clinical evidence links lipid dysregulation to an increased risk of Alzheimer's disease (AD). However, the role of lipids in the pathophysiological processes of AD and its clinical progression is unclear. We investigated the association between cerebrospinal fluid (CSF) lipidome and the pathological hallmarks of AD, progression from mild cognitive impairment (MCI) to AD, and the rate of cognitive decline in MCI patients. The CSF lipidome was analyzed by liquid chromatography coupled to mass spectrometry in an LC-ESI-QTOF-MS/MS platform for 209 participants: 91 AD, 92 MCI, and 26 control participants. The MCI patients were followed up for a median of 58 (± 12.5) months to evaluate their clinical progression to AD. Forty-eight (52.2%) MCI patients progressed to AD during follow-up. We found that higher CSF levels of hexacosanoic acid and ceramide Cer(d38:4) were associated with an increased risk of amyloid beta 42 (Aβ42) positivity in CSF, while levels of phosphatidylethanolamine PE(40:0) were associated with a reduced risk. Higher CSF levels of sphingomyelin SM(30:1) were positively associated with pathological levels of phosphorylated tau in CSF. Cholesteryl ester CE(11D3:1) and an unknown lipid were recognized as the most associated lipid species with MCI to AD progression. Furthermore, TG(O-52:2) was identified as the lipid most strongly associated with the rate of progression. Our results indicate the involvement of membrane and intracellular neutral lipids in the pathophysiological processes of AD and the progression from MCI to AD dementia. Therefore, CSF neutral lipids can be used as potential prognostic markers for AD.
Collapse
Affiliation(s)
- Farida Dakterzada
- Unitat Trastorns Cognitius, Cognition and Behaviour Study Group, Hospital Universitari Santa Maria, IRBLleida, Rovira Roure No 44. 25198, Lleida, Spain
| | - Mariona Jové
- Department of Experimental Medicine, University of Lleida, IRBLleida, Lleida, Spain
| | - Raquel Huerto
- Unitat Trastorns Cognitius, Cognition and Behaviour Study Group, Hospital Universitari Santa Maria, IRBLleida, Rovira Roure No 44. 25198, Lleida, Spain
| | - Anna Carnes
- Unitat Trastorns Cognitius, Cognition and Behaviour Study Group, Hospital Universitari Santa Maria, IRBLleida, Rovira Roure No 44. 25198, Lleida, Spain
| | - Joaquim Sol
- Department of Experimental Medicine, University of Lleida, IRBLleida, Lleida, Spain
- Institut Català de La Salut, Lleida, Spain
- Research Support Unit Lleida, Fundació Institut Universitari Per a La Recerca a L'Atenció Primària de Salut Jordi Gol I Gurina (IDIAPJGol), Lleida, Spain
| | - Reinald Pamplona
- Department of Experimental Medicine, University of Lleida, IRBLleida, Lleida, Spain
| | - Gerard Piñol-Ripoll
- Unitat Trastorns Cognitius, Cognition and Behaviour Study Group, Hospital Universitari Santa Maria, IRBLleida, Rovira Roure No 44. 25198, Lleida, Spain.
| |
Collapse
|
29
|
Vervuurt M, Schrader JM, de Kort AM, Kersten I, Wessels HJCT, Klijn CJM, Schreuder FHBM, Kuiperij HB, Gloerich J, Van Nostrand WE, Verbeek MM. Cerebrospinal fluid shotgun proteomics identifies distinct proteomic patterns in cerebral amyloid angiopathy rodent models and human patients. Acta Neuropathol Commun 2024; 12:6. [PMID: 38191511 PMCID: PMC10775534 DOI: 10.1186/s40478-023-01698-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 11/28/2023] [Indexed: 01/10/2024] Open
Abstract
Cerebral amyloid angiopathy (CAA) is a form of small vessel disease characterised by the progressive deposition of amyloid β protein in the cerebral vasculature, inducing symptoms including cognitive impairment and cerebral haemorrhages. Due to their accessibility and homogeneous disease phenotypes, animal models are advantageous platforms to study diseases like CAA. Untargeted proteomics studies of CAA rat models (e.g. rTg-DI) and CAA patients provide opportunities for the identification of novel biomarkers of CAA. We performed untargeted, data-independent acquisition proteomic shotgun analyses on the cerebrospinal fluid of rTg-DI rats and wild-type (WT) littermates. Rodents were analysed at 3 months (n = 6/10), 6 months (n = 8/8), and 12 months (n = 10/10) for rTg-DI and WT respectively. For humans, proteomic analyses were performed on CSF of sporadic CAA patients (sCAA) and control participants (n = 39/28). We show recurring patterns of differentially expressed (mostly increased) proteins in the rTg-DI rats compared to wild type rats, especially of proteases of the cathepsin protein family (CTSB, CTSD, CTSS), and their main inhibitor (CST3). In sCAA patients, decreased levels of synaptic proteins (e.g. including VGF, NPTX1, NRXN2) and several members of the granin family (SCG1, SCG2, SCG3, SCG5) compared to controls were discovered. Additionally, several serine protease inhibitors of the SERPIN protein family (including SERPINA3, SERPINC1 and SERPING1) were differentially expressed compared to controls. Fifteen proteins were significantly altered in both rTg-DI rats and sCAA patients, including (amongst others) SCG5 and SERPING1. These results identify specific groups of proteins likely involved in, or affected by, pathophysiological processes involved in CAA pathology such as protease and synapse function of rTg-DI rat models and sCAA patients, and may serve as candidate biomarkers for sCAA.
Collapse
Affiliation(s)
- Marc Vervuurt
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, 830 TML, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Joseph M Schrader
- Department of Biomedical and Pharmaceutical Sciences, George & Anne Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA
| | - Anna M de Kort
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, 830 TML, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Iris Kersten
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, 830 TML, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Hans J C T Wessels
- Department of Human Genetics, Translational Metabolic Laboratory, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Catharina J M Klijn
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, 830 TML, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Floris H B M Schreuder
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, 830 TML, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - H Bea Kuiperij
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, 830 TML, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Jolein Gloerich
- Department of Human Genetics, Translational Metabolic Laboratory, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - William E Van Nostrand
- Department of Biomedical and Pharmaceutical Sciences, George & Anne Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA
| | - Marcel M Verbeek
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, 830 TML, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
- Department of Human Genetics, Translational Metabolic Laboratory, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
30
|
Krishnamurthy K, Pradhan RK. Emerging perspectives of synaptic biomarkers in ALS and FTD. Front Mol Neurosci 2024; 16:1279999. [PMID: 38249293 PMCID: PMC10796791 DOI: 10.3389/fnmol.2023.1279999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 12/01/2023] [Indexed: 01/23/2024] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) and Frontotemporal Dementia (FTD) are debilitating neurodegenerative diseases with shared pathological features like transactive response DNA-binding protein of 43 kDa (TDP-43) inclusions and genetic mutations. Both diseases involve synaptic dysfunction, contributing to their clinical features. Synaptic biomarkers, representing proteins associated with synaptic function or structure, offer insights into disease mechanisms, progression, and treatment responses. These biomarkers can detect disease early, track its progression, and evaluate therapeutic efficacy. ALS is characterized by elevated neurofilament light chain (NfL) levels in cerebrospinal fluid (CSF) and blood, correlating with disease progression. TDP-43 is another key ALS biomarker, its mislocalization linked to synaptic dysfunction. In FTD, TDP-43 and tau proteins are studied as biomarkers. Synaptic biomarkers like neuronal pentraxins (NPs), including neuronal pentraxin 2 (NPTX2), and neuronal pentraxin receptor (NPTXR), offer insights into FTD pathology and cognitive decline. Advanced technologies, like machine learning (ML) and artificial intelligence (AI), aid biomarker discovery and drug development. Challenges in this research include technological limitations in detection, variability across patients, and translating findings from animal models. ML/AI can accelerate discovery by analyzing complex data and predicting disease outcomes. Synaptic biomarkers offer early disease detection, personalized treatment strategies, and insights into disease mechanisms. While challenges persist, technological advancements and interdisciplinary efforts promise to revolutionize the understanding and management of ALS and FTD. This review will explore the present comprehension of synaptic biomarkers in ALS and FTD and discuss their significance and emphasize the prospects and obstacles.
Collapse
Affiliation(s)
- Karrthik Krishnamurthy
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Thomas Jefferson University, Philadelphia, PA, United States
| | | |
Collapse
|
31
|
Franzmeier N, Dehsarvi A, Steward A, Biel D, Dewenter A, Roemer SN, Wagner F, Groß M, Brendel M, Moscoso A, Arunachalam P, Blennow K, Zetterberg H, Ewers M, Schöll M. Elevated CSF GAP-43 is associated with accelerated tau accumulation and spread in Alzheimer's disease. Nat Commun 2024; 15:202. [PMID: 38172114 PMCID: PMC10764818 DOI: 10.1038/s41467-023-44374-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 12/11/2023] [Indexed: 01/05/2024] Open
Abstract
In Alzheimer's disease, amyloid-beta (Aβ) triggers the trans-synaptic spread of tau pathology, and aberrant synaptic activity has been shown to promote tau spreading. Aβ induces aberrant synaptic activity, manifesting in increases in the presynaptic growth-associated protein 43 (GAP-43), which is closely involved in synaptic activity and plasticity. We therefore tested whether Aβ-related GAP-43 increases, as a marker of synaptic changes, drive tau spreading in 93 patients across the aging and Alzheimer's spectrum with available CSF GAP-43, amyloid-PET and longitudinal tau-PET assessments. We found that (1) higher GAP-43 was associated with faster Aβ-related tau accumulation, specifically in brain regions connected closest to subject-specific tau epicenters and (2) that higher GAP-43 strengthened the association between Aβ and connectivity-associated tau spread. This suggests that GAP-43-related synaptic changes are linked to faster Aβ-related tau spread across connected regions and that synapses could be key targets for preventing tau spreading in Alzheimer's disease.
Collapse
Affiliation(s)
- Nicolai Franzmeier
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
- University of Gothenburg, The Sahlgrenska Academy, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Mölndal and Gothenburg, Sweden.
| | - Amir Dehsarvi
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Anna Steward
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Davina Biel
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Anna Dewenter
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Sebastian Niclas Roemer
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Fabian Wagner
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Mattes Groß
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Matthias Brendel
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Alexis Moscoso
- University of Gothenburg, The Sahlgrenska Academy, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Mölndal and Gothenburg, Sweden
| | - Prithvi Arunachalam
- University of Gothenburg, The Sahlgrenska Academy, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Mölndal and Gothenburg, Sweden
| | - Kaj Blennow
- University of Gothenburg, The Sahlgrenska Academy, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Mölndal and Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- University of Gothenburg, The Sahlgrenska Academy, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Mölndal and Gothenburg, 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
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael Ewers
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Michael Schöll
- University of Gothenburg, The Sahlgrenska Academy, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Mölndal and Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Dementia Research Centre, Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
32
|
Duits FH, Nilsson J, Zetterberg H, Blennow K, van der Flier WM, Teunissen CE, Brinkmalm A. Serial Cerebrospinal Fluid Sampling Reveals Trajectories of Potential Synaptic Biomarkers in Early Stages of Alzheimer's Disease. J Alzheimers Dis 2024; 100:S103-S114. [PMID: 39121126 PMCID: PMC11492063 DOI: 10.3233/jad-240610] [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] [Accepted: 07/24/2024] [Indexed: 08/11/2024]
Abstract
Background Synaptic dysfunction is closely associated with cognitive function in Alzheimer's disease (AD), and is present already in an early stage of the disease. Objective Using serial cerebrospinal fluid (CSF) sampling, we aimed to investigate slopes of CSF synaptic proteins, and their relation with cognition along the AD continuum. Methods We included subjects with subjective cognitive decline (SCD) or mild cognitive impairment (MCI) (n = 50 amyloid-β+ [A +], n = 50 A-) and 50 patients with AD dementia from the Amsterdam dementia cohort, with CSF at two time points (median[IQR] 2.1[1.4-2.7] years). We analyzed 17 synaptic proteins and neurofilament light (NfL). Using linear mixed models we assessed trajectories of protein levels, and associations with cognitive decline (repeated Mini-Mental State Examination). We used Cox regression models to assess predictive value of protein levels for progression to AD dementia. Results At baseline most proteins showed increased levels in AD dementia compared to the other groups. In contrast NPTX2 levels were lower in AD dementia. Higher baseline levels of SNAP25, β-syn, and 14-3-3 proteins were associated with faster cognitive decline (St.B[SE] -0.27[0.12] to -0.61[0.12]). Longitudinal analyses showed that SYT1 and NPTX levels decreased over time in AD dementia (st.B[SE] -0.10[0.04] to -0.15[0.05]) and SCD/MCI-A+ (St.B[SE] -0.07[0.03] to -0.12[0.03]), but not in SCD/MCI-A- (pinteraction < 0.05). Increase over time in NfL levels was associated with faster cognitive decline in AD dementia (St.B[SE] -1.75[0.58]), but not in the other groups (pinteraction < 0.05). Conclusions CSF synaptic proteins showed different slopes over time, suggesting complex synaptic dynamics. High levels of especially SNAP-25 may have value for prediction of cognitive decline in early AD stages, while increase in NfL over time correlates better with cognitive decline in later stages.
Collapse
Affiliation(s)
- Flora H. Duits
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Department of Laboratory Medicine, Neurochemistry Lab, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Johanna Nilsson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, 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 and UK Dementia Research Institute, UCL Institute of Neurology, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - 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
| | - Wiesje M. van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Department of Epidemiology and Biostatistics, Amsterdam UMC, Amsterdam, The Netherlands
| | - Charlotte E. Teunissen
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Department of Laboratory Medicine, Neurochemistry Lab, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - 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
| |
Collapse
|
33
|
Taddei RN, Perbet R, Mate de Gerando A, Wiedmer AE, Sanchez-Mico M, Connors Stewart T, Gaona A, Melloni A, Amaral AC, Duff K, Frosch MP, Gómez-Isla T. Tau Oligomer-Containing Synapse Elimination by Microglia and Astrocytes in Alzheimer Disease. JAMA Neurol 2023; 80:1209-1221. [PMID: 37812432 PMCID: PMC10562992 DOI: 10.1001/jamaneurol.2023.3530] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/03/2023] [Indexed: 10/10/2023]
Abstract
Importance Factors associated with synapse loss beyond amyloid-β plaques and neurofibrillary tangles may more closely correlate with the emergence of cognitive deficits in Alzheimer disease (AD) and be relevant for early therapeutic intervention. Objective To investigate whether accumulation of tau oligomers in synapses is associated with excessive synapse elimination by microglia or astrocytes and with cognitive outcomes (dementia vs no dementia [hereinafter termed resilient]) of individuals with equal burdens of AD neuropathologic changes at autopsy. Design, Setting, and Participants This cross-sectional postmortem study included 40 human brains from the Massachusetts Alzheimer Disease Research Center Brain Bank with Braak III to IV stages of tau pathology but divergent antemortem cognition (dementia vs resilient) and cognitively normal controls with negligible AD neuropathologic changes. The visual cortex, a region without tau tangle deposition at Braak III to IV stages, was assessed after expansion microscopy to analyze spatial relationships of synapses with microglia and astrocytes. Participants were matched for age, sex, and apolipoprotein E status. Evidence of Lewy bodies, TDP-43 aggregates, or other lesions different from AD neuropathology were exclusion criteria. Tissue was collected from July 1998 to November 2020, and analyses were conducted from February 1, 2022, through May 31, 2023. Main Outcomes and Measures Amyloid-β plaques, tau neuropil thread burden, synapse density, tau oligomers in synapses, and internalization of tau oligomer-tagged synapses by microglia and astrocytes were quantitated. Analyses were performed using 1-way analysis of variance for parametric variables and the Kruskal-Wallis test for nonparametric variables; between-group differences were evaluated with Holm-Šídák tests. Results Of 40 included participants (mean [SD] age at death, 88 [8] years; 21 [52%] male), 19 had early-stage dementia with Braak stages III to IV, 13 had resilient brains with similar Braak stages III to IV, and 8 had no dementia (Braak stages 0-II). Brains with dementia but not resilient brains had substantial loss of presynaptic (43%), postsynaptic (33%), and colocalized mature synaptic elements (38%) compared with controls and significantly higher percentages of mature synapses internalized by IBA1-positive microglia (mean [SD], 13.3% [3.9%] in dementia vs 2.6% [1.9%] in resilient vs 0.9% [0.5%] in control; P < .001) and by GFAP-positive astrocytes (mean [SD], 17.2% [10.9%] in dementia vs 3.7% [4.0%] in resilient vs 2.7% [1.8%] in control; P = .001). In brains with dementia but not in resilient brains, tau oligomers more often colocalized with synapses, and the proportions of tau oligomer-containing synapses inside microglia (mean [SD] for presynapses, mean [SD], 7.4% [1.8%] in dementia vs 5.1% [1.9%] resilient vs 3.7% [0.8%] control; P = .006; and for postsynapses 11.6% [3.6%] dementia vs 6.8% [1.3%] resilient vs 7.4% [2.5%] control; P = .001) and astrocytes (mean [SD] for presynapses, 7.0% [2.1%] dementia vs 4.3% [2.2%] resilient vs 4.0% [0.7%] control; P = .001; and for postsynapses, 7.9% [2.2%] dementia vs 5.3% [1.8%] resilient vs 3.0% [1.5%] control; P < .001) were significantly increased compared with controls. Those changes in brains with dementia occurred in the absence of tau tangle deposition in visual cortex. Conclusion and Relevance The findings from this cross-sectional study suggest that microglia and astrocytes may excessively engulf synapses in brains of individuals with dementia and that the abnormal presence of tau oligomers in synapses may serve as signals for increased glial-mediated synapse elimination and early loss of brain function in AD.
Collapse
Affiliation(s)
- Raquel N. Taddei
- Neurology Department, Massachusetts General Hospital, Harvard University, Boston, Massachusetts
- Department of Neurology, Dementia Research Institute, University College London, United Kingdom
| | - Romain Perbet
- Neurology Department, Massachusetts General Hospital, Harvard University, Boston, Massachusetts
| | | | - Anne E. Wiedmer
- Neurology Department, Massachusetts General Hospital, Harvard University, Boston, Massachusetts
| | - Maria Sanchez-Mico
- Neurology Department, Massachusetts General Hospital, Harvard University, Boston, Massachusetts
| | - Theresa Connors Stewart
- C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Angelica Gaona
- C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Alexandra Melloni
- C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Ana C. Amaral
- Neurology Department, Massachusetts General Hospital, Harvard University, Boston, Massachusetts
| | - Karen Duff
- Department of Neurology, Dementia Research Institute, University College London, United Kingdom
| | - Matthew P. Frosch
- C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Teresa Gómez-Isla
- Neurology Department, Massachusetts General Hospital, Harvard University, Boston, Massachusetts
| |
Collapse
|
34
|
Oeckl P, Janelidze S, Halbgebauer S, Stomrud E, Palmqvist S, Otto M, Hansson O. Higher plasma β-synuclein indicates early synaptic degeneration in Alzheimer's disease. Alzheimers Dement 2023; 19:5095-5102. [PMID: 37186338 DOI: 10.1002/alz.13103] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 05/17/2023]
Abstract
INTRODUCTION β-Synuclein is an emerging synaptic blood biomarker for Alzheimer's disease (AD) but differences in β-synuclein levels in preclinical AD and its association with amyloid and tau pathology have not yet been studied. METHODS We measured plasma β-synuclein levels in cognitively unimpaired individuals with positive Aβ-PET (i.e., preclinical AD, N = 48) or negative Aβ-PET (N = 61), Aβ-positive patients with mild cognitive impairment (MCI, N = 36), and Aβ-positive AD dementia (N = 85). Amyloid (A) and tau (T) pathology were assessed by [18 F]flutemetamol and [18 F]RO948 PET. RESULTS Plasma β-synuclein levels were higher in preclinical AD and even higher in MCI and AD dementia. Stratification according to amyloid/tau pathology revealed higher β-synuclein in A+ T- and A+ T+ subjects compared with A- T- . Plasma β-synuclein levels were related to tau and Aβ pathology and associated with temporal cortical thinning and cognitive impairment. DISCUSSION Our data indicate that plasma β-synuclein might track synaptic dysfunction, even during the preclinical stages of AD. HIGHLIGHTS Plasma β-synuclein is already higher in preclinical AD. Plasma β-synuclein is higher in MCI and AD dementia than in preclinical AD. Aβ- and tau-PET SUVRs are associated with plasma β-synuclein levels. Plasma β-synuclein is already higher in tau-PET negative subjects. Plasma β-synuclein is related to temporal cortical atrophy and cognitive impairment.
Collapse
Affiliation(s)
- Patrick Oeckl
- German Center for Neurodegenerative Diseases e.V. (DZNE), Ulm, Germany
- Department of Neurology, Ulm University Hospital, Ulm, Germany
| | - Shorena Janelidze
- Department of Clinical Sciences Malmö, Clinical Memory Research Unit, Lund University, Malmö, Sweden
| | - Steffen Halbgebauer
- German Center for Neurodegenerative Diseases e.V. (DZNE), Ulm, Germany
- Department of Neurology, Ulm University Hospital, Ulm, Germany
| | - Erik Stomrud
- Department of Clinical Sciences Malmö, Clinical Memory Research Unit, Lund University, Malmö, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Sebastian Palmqvist
- Department of Clinical Sciences Malmö, Clinical Memory Research Unit, Lund University, Malmö, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Markus Otto
- Department of Neurology, Ulm University Hospital, Ulm, Germany
- University Clinic and Polyclinic for Neurology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Oskar Hansson
- Department of Clinical Sciences Malmö, Clinical Memory Research Unit, Lund University, Malmö, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
35
|
Grønning R, Jeppsson A, Hellström P, Laurell K, Farahmand D, Zetterberg H, Blennow K, Wikkelsø C, Tullberg M. Association between ventricular CSF biomarkers and outcome after shunt surgery in idiopathic normal pressure hydrocephalus. Fluids Barriers CNS 2023; 20:77. [PMID: 37880775 PMCID: PMC10601279 DOI: 10.1186/s12987-023-00475-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 10/06/2023] [Indexed: 10/27/2023] Open
Abstract
INTRODUCTION The relationship between neurochemical changes and outcome after shunt surgery in idiopathic normal pressure hydrocephalus (iNPH), a treatable dementia and gait disorder, is unclear. We used baseline ventricular CSF to explore associations to outcome, after shunting, of biomarkers selected to reflect a range of pathophysiological processes. METHODS In 119 consecutive patients with iNPH, the iNPH scale was used before and after shunt surgery to quantify outcome. Ventricular CSF was collected perioperatively and analyzed for biomarkers of astrogliosis, axonal, amyloid and tau pathology, and synaptic dysfunction: glial fibrillary acidic protein (GFAP), chitinase-3-like protein 1 (YKL40/CHI3L1), monocyte chemoattractant protein-1 (MCP-1) neurofilament light (NfL), amyloid beta 38 (Aβ38), Aβ40, Aβ42, amyloid beta 42/40 ratio (Aβ42/40), soluble amyloid precursor protein alfa (sAPPα), sAPPβ, total tau (T-tau), phosphorylated tau (P-tau), growth-associated protein 43 (GAP43), and neurogranin. RESULTS The neurogranin concentration was higher in improved (68%) compared to unimproved patients (median 365 ng/L (IQR 186-544) vs 330 (205-456); p = 0.046). A linear regression model controlled for age, sex and vascular risk factors including neurogranin, T-tau, and GFAP, resulted in adjusted R2 = 0.06, p = 0.047. The Aβ42/40 ratio was bimodally distributed across all samples, as well as in the subgroups of improved and unimproved patients but did not contribute to outcome prediction. The preoperative MMSE score was lower within the low Aβ ratio group (median 25, IQR 23-28) compared to the high subgroup (26, 24-29) (p = 0.028). The T-Tau x Aβ40/42 ratio and P-tau x Aβ40/42 ratio did not contribute to shunt response prediction. The prevalence of vascular risk factors did not affect shunt response. DISCUSSION A higher preoperative ventricular CSF level of neurogranin, which is a postsynaptic marker, may signal a favorable postoperative outcome. Concentrations of a panel of ventricular CSF biomarkers explained only 6% of the variability in outcome. Evidence of amyloid or tau pathology did not affect the outcome.
Collapse
Affiliation(s)
- Rebecca Grønning
- Hydrocephalus Research Unit, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Blå Stråket 7, Sahlgrenska University Hospital, 41345, Gothenburg, Sweden.
| | - Anna Jeppsson
- Hydrocephalus Research Unit, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Blå Stråket 7, Sahlgrenska University Hospital, 41345, Gothenburg, Sweden
| | - Per Hellström
- Hydrocephalus Research Unit, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Blå Stråket 7, Sahlgrenska University Hospital, 41345, Gothenburg, Sweden
| | - Katarina Laurell
- Department of Medical Sciences, Neurology, Uppsala University, Uppsala, Sweden
| | - Dan Farahmand
- Hydrocephalus Research Unit, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Blå Stråket 7, Sahlgrenska University Hospital, 41345, Gothenburg, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- 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, WI, USA
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Carsten Wikkelsø
- Hydrocephalus Research Unit, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Blå Stråket 7, Sahlgrenska University Hospital, 41345, Gothenburg, Sweden
| | - Mats Tullberg
- Hydrocephalus Research Unit, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Blå Stråket 7, Sahlgrenska University Hospital, 41345, Gothenburg, Sweden
| |
Collapse
|
36
|
Shaw LM, Galasko D. Assessments of the Utilities of CSF NPTX2 for Disease Progression in Cognitively Normal Individuals Who Progress to Clinical MCI and AD. Ann Neurol 2023; 94:618-619. [PMID: 37614206 DOI: 10.1002/ana.26768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 08/11/2023] [Indexed: 08/25/2023]
Affiliation(s)
- Leslie M Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Douglas Galasko
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
37
|
Soldan A, Oh S, Ryu T, Pettigrew C, Zhu Y, Moghekar A, Xiao MF, Pontone GM, Albert M, Na CH, Worley P. NPTX2 in Cerebrospinal Fluid Predicts the Progression From Normal Cognition to Mild Cognitive Impairment. Ann Neurol 2023; 94:620-631. [PMID: 37345460 PMCID: PMC10543570 DOI: 10.1002/ana.26725] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/31/2023] [Accepted: 06/04/2023] [Indexed: 06/23/2023]
Abstract
OBJECTIVE This study examined whether cerebrospinal fluid (CSF) baseline levels of the synaptic protein NPTX2 predict time to onset of symptoms of mild cognitive impairment (MCI), both alone and when accounting for traditional CSF Alzheimer's disease (AD) biomarker levels. Longitudinal NPTX2 levels were also examined. METHODS CSF was collected longitudinally from 269 cognitively normal BIOCARD Study participants (mean baseline age = 57.7 years; mean follow-up = 16.3 years; n = 77 progressed to MCI/dementia). NPTX2 levels were measured from 3 correlated peptides using quantitative parallel reaction monitoring mass spectrometry. Levels of Aβ42 /Aβ40 , p-tau181 , and t-tau were measured from the same CSF specimens using Lumipulse automated electrochemiluminescence assays. RESULTS In Cox regression models, lower baseline NPTX2 levels were associated with an earlier time to MCI symptom onset (hazard ratio [HR] = 0.76, SE = 0.09, p = 0.023). This association was significant for progression within 7 years (p = 0.036) and after 7 years from baseline (p = 0.001). Baseline NPTX2 levels improved prediction of time to MCI symptom onset after accounting for baseline AD biomarker levels (p < 0.01), and NPTX2 did not interact with the CSF AD biomarkers or APOE-ε4 genetic status. In linear mixed effects models, higher baseline p-tau181 and t-tau levels were associated with higher baseline levels of NPTX2 (both p < 0.001) and greater rates of NPTX2 declines over time. INTERPRETATION NPTX2 may be a valuable prognostic biomarker during preclinical AD that provides additive and independent prediction of MCI onset among individuals who are cognitively normal. We hypothesize that NPTX2-mediated circuit homeostasis confers resilience during the early phase of AD. ANN NEUROL 2023;94:620-631.
Collapse
Affiliation(s)
- Anja Soldan
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Sungtaek Oh
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Taekyung Ryu
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Corinne Pettigrew
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Yuxin Zhu
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Armstrong Institute for Patient Safety and Quality, Johns Hopkins Medicine, Baltimore, MD
| | - Abhay Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Mei-Fang Xiao
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Gregory M. Pontone
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Marilyn Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Chan-Hyun Na
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Paul Worley
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
38
|
Elmers J, Colzato LS, Akgün K, Ziemssen T, Beste C. Neurofilaments - Small proteins of physiological significance and predictive power for future neurodegeneration and cognitive decline across the life span. Ageing Res Rev 2023; 90:102037. [PMID: 37619618 DOI: 10.1016/j.arr.2023.102037] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/15/2023] [Accepted: 08/17/2023] [Indexed: 08/26/2023]
Abstract
Neurofilaments (NFs) are not only important for axonal integrity and nerve conduction in large myelinated axons but they are also thought to be crucial for receptor and synaptic functioning. Therefore, NFs may play a critical role in cognitive functions, as cognitive processes are known to depend on synaptic integrity and are modulated by dopaminergic signaling. Here, we present a theory-driven interdisciplinary approach that NFs may link inflammation, neurodegeneration, and cognitive functions. We base our hypothesis on a wealth of evidence suggesting a causal link between inflammation and neurodegeneration and between these two and cognitive decline (see Fig. 1), also taking dopaminergic signaling into account. We conclude that NFs may not only serve as biomarkers for inflammatory, neurodegenerative, and cognitive processes but also represent a potential mechanical hinge between them, moreover, they may even have predictive power regarding future cognitive decline. In addition, we advocate the use of both NFs and MRI parameters, as their synthesis offers the opportunity to individualize medical treatment by providing a comprehensive view of underlying disease activity in neurological diseases. Since our society will become significantly older in the upcoming years and decades, maintaining cognitive functions and healthy aging will play an important role. Thanks to technological advances in recent decades, NFs could serve as a rapid, noninvasive, and relatively inexpensive early warning system to identify individuals at increased risk for cognitive decline and could facilitate the management of cognitive dysfunctions across the lifespan.
Collapse
Affiliation(s)
- Julia Elmers
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Germany; Center of Clinical Neuroscience, Department of Neurology, University Hospital Carl Gustav Carus, TU Dresden, Germany
| | - Lorenza S Colzato
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Germany; Cognitive Psychology, Faculty of Psychology, Shandong Normal University, Jinan, China.
| | - Katja Akgün
- Center of Clinical Neuroscience, Department of Neurology, University Hospital Carl Gustav Carus, TU Dresden, Germany
| | - Tjalf Ziemssen
- Center of Clinical Neuroscience, Department of Neurology, University Hospital Carl Gustav Carus, TU Dresden, Germany
| | - Christian Beste
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Germany; Cognitive Psychology, Faculty of Psychology, Shandong Normal University, Jinan, China.
| |
Collapse
|
39
|
Daniilidou M, Eroli F, Alanko V, Goikolea J, Latorre-Leal M, Rodriguez-Rodriguez P, Griffiths WJ, Wang Y, Pacciarini M, Brinkmalm A, Zetterberg H, Blennow K, Rosenberg A, Bogdanovic N, Winblad B, Kivipelto M, Ibghi D, Cedazo-Minguez A, Maioli S, Matton A. Alzheimer's disease biomarker profiling in a memory clinic cohort without common comorbidities. Brain Commun 2023; 5:fcad228. [PMID: 37680670 PMCID: PMC10481253 DOI: 10.1093/braincomms/fcad228] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 05/17/2023] [Accepted: 08/24/2023] [Indexed: 09/09/2023] Open
Abstract
Alzheimer's disease is a multifactorial disorder with large heterogeneity. Comorbidities such as hypertension, hypercholesterolaemia and diabetes are known contributors to disease progression. However, less is known about their mechanistic contribution to Alzheimer's pathology and neurodegeneration. The aim of this study was to investigate the relationship of several biomarkers related to risk mechanisms in Alzheimer's disease with the well-established Alzheimer's disease markers in a memory clinic population without common comorbidities. We investigated 13 molecular markers representing key mechanisms underlying Alzheimer's disease pathogenesis in CSF from memory clinic patients without diagnosed hypertension, hypercholesterolaemia or diabetes nor other neurodegenerative disorders. An analysis of covariance was used to compare biomarker levels between clinical groups. Associations were analysed by linear regression. Two-step cluster analysis was used to determine patient clusters. Two key markers were analysed by immunofluorescence staining in the hippocampus of non-demented control and Alzheimer's disease individuals. CSF samples from a total of 90 participants were included in this study: 30 from patients with subjective cognitive decline (age 62.4 ± 4.38, female 60%), 30 with mild cognitive impairment (age 65.6 ± 7.48, female 50%) and 30 with Alzheimer's disease (age 68.2 ± 7.86, female 50%). Angiotensinogen, thioredoxin-1 and interleukin-15 had the most prominent associations with Alzheimer's disease pathology, synaptic and axonal damage markers. Synaptosomal-associated protein 25 kDa and neurofilament light chain were increased in mild cognitive impairment and Alzheimer's disease patients. Grouping biomarkers by biological function showed that inflammatory and survival components were associated with Alzheimer's disease pathology, synaptic dysfunction and axonal damage. Moreover, a vascular/metabolic component was associated with synaptic dysfunction. In the data-driven analysis, two patient clusters were identified: Cluster 1 had increased CSF markers of oxidative stress, vascular pathology and neuroinflammation and was characterized by elevated synaptic and axonal damage, compared with Cluster 2. Clinical groups were evenly distributed between the clusters. An analysis of post-mortem hippocampal tissue showed that compared with non-demented controls, angiotensinogen staining was higher in Alzheimer's disease and co-localized with phosphorylated-tau. The identification of biomarker-driven endophenotypes in cognitive disorder patients further highlights the biological heterogeneity of Alzheimer's disease and the importance of tailored prevention and treatment strategies.
Collapse
Affiliation(s)
- Makrina Daniilidou
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 64 Solna, Sweden
- Division of Clinical Geriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 83 Huddinge, Sweden
| | - Francesca Eroli
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 64 Solna, Sweden
| | - Vilma Alanko
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 64 Solna, Sweden
- Division of Clinical Geriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 83 Huddinge, Sweden
| | - Julen Goikolea
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 64 Solna, Sweden
| | - Maria Latorre-Leal
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 64 Solna, Sweden
| | - Patricia Rodriguez-Rodriguez
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 64 Solna, Sweden
| | | | - Yuqin Wang
- Swansea University Medical School, Swansea SA2 8PP, UK
| | | | - Ann Brinkmalm
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 413 90 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 90 Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 413 90 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 90 Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London WC1N3AR, UK
- UK Dementia Research Institute at UCL, London WC1N3AR, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 413 90 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 90 Mölndal, Sweden
| | - Anna Rosenberg
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, FI-70029 Kuopio, Finland
| | - Nenad Bogdanovic
- Theme Inflammation and Aging, Karolinska University Hospital, 141 83 Huddinge, Sweden
| | - Bengt Winblad
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 64 Solna, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, 141 83 Huddinge, Sweden
| | - Miia Kivipelto
- Division of Clinical Geriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 83 Huddinge, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, 141 83 Huddinge, Sweden
- Ageing Epidemiology Research Unit, School of Public Health, Imperial College London, London SW7 2AZ, UK
| | - Delphine Ibghi
- Neurodegeneration Cluster, Rare and Neurologic Disease Research Sanofi R&D, F-91380 Chilly-Mazarin, France
| | - Angel Cedazo-Minguez
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 64 Solna, Sweden
- Neurodegeneration Cluster, Rare and Neurologic Disease Research Sanofi R&D, F-91380 Chilly-Mazarin, France
| | - Silvia Maioli
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 64 Solna, Sweden
| | - Anna Matton
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 64 Solna, Sweden
- Division of Clinical Geriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 83 Huddinge, Sweden
- Ageing Epidemiology Research Unit, School of Public Health, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
40
|
Lista S, González-Domínguez R, López-Ortiz S, González-Domínguez Á, Menéndez H, Martín-Hernández J, Lucia A, Emanuele E, Centonze D, Imbimbo BP, Triaca V, Lionetto L, Simmaco M, Cuperlovic-Culf M, Mill J, Li L, Mapstone M, Santos-Lozano A, Nisticò R. Integrative metabolomics science in Alzheimer's disease: Relevance and future perspectives. Ageing Res Rev 2023; 89:101987. [PMID: 37343679 DOI: 10.1016/j.arr.2023.101987] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/12/2023] [Accepted: 06/15/2023] [Indexed: 06/23/2023]
Abstract
Alzheimer's disease (AD) is determined by various pathophysiological mechanisms starting 10-25 years before the onset of clinical symptoms. As multiple functionally interconnected molecular/cellular pathways appear disrupted in AD, the exploitation of high-throughput unbiased omics sciences is critical to elucidating the precise pathogenesis of AD. Among different omics, metabolomics is a fast-growing discipline allowing for the simultaneous detection and quantification of hundreds/thousands of perturbed metabolites in tissues or biofluids, reproducing the fluctuations of multiple networks affected by a disease. Here, we seek to critically depict the main metabolomics methodologies with the aim of identifying new potential AD biomarkers and further elucidating AD pathophysiological mechanisms. From a systems biology perspective, as metabolic alterations can occur before the development of clinical signs, metabolomics - coupled with existing accessible biomarkers used for AD screening and diagnosis - can support early disease diagnosis and help develop individualized treatment plans. Presently, the majority of metabolomic analyses emphasized that lipid metabolism is the most consistently altered pathway in AD pathogenesis. The possibility that metabolomics may reveal crucial steps in AD pathogenesis is undermined by the difficulty in discriminating between the causal or epiphenomenal or compensatory nature of metabolic findings.
Collapse
Affiliation(s)
- Simone Lista
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid, Spain.
| | - Raúl González-Domínguez
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, Universidad de Cádiz, Cádiz, Spain
| | - Susana López-Ortiz
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid, Spain
| | - Álvaro González-Domínguez
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, Universidad de Cádiz, Cádiz, Spain
| | - Héctor Menéndez
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid, Spain
| | - Juan Martín-Hernández
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid, Spain
| | - Alejandro Lucia
- Research Institute of the Hospital 12 de Octubre ('imas12'), Madrid, Spain; Faculty of Sport Sciences, European University of Madrid, Villaviciosa de Odón, Madrid, Spain; CIBER of Frailty and Healthy Ageing (CIBERFES), Madrid, Spain
| | | | - Diego Centonze
- Department of Systems Medicine, Tor Vergata University, Rome, Italy; Unit of Neurology, IRCCS Neuromed, Pozzilli, IS, Italy
| | - Bruno P Imbimbo
- Department of Research and Development, Chiesi Farmaceutici, Parma, Italy
| | - Viviana Triaca
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Rome, Italy
| | - Luana Lionetto
- Clinical Biochemistry, Mass Spectrometry Section, Sant'Andrea University Hospital, Rome, Italy; Department of Neuroscience, Mental Health and Sensory Organs, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Maurizio Simmaco
- Clinical Biochemistry, Mass Spectrometry Section, Sant'Andrea University Hospital, Rome, Italy; Department of Neuroscience, Mental Health and Sensory Organs, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Miroslava Cuperlovic-Culf
- Digital Technologies Research Center, National Research Council, Ottawa, Canada; Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Jericha Mill
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Lingjun Li
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA; School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Mark Mapstone
- Department of Neurology, Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
| | - Alejandro Santos-Lozano
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid, Spain; Research Institute of the Hospital 12 de Octubre ('imas12'), Madrid, Spain
| | - Robert Nisticò
- School of Pharmacy, University of Rome "Tor Vergata", Rome, Italy; Laboratory of Pharmacology of Synaptic Plasticity, EBRI Rita Levi-Montalcini Foundation, Rome, Italy
| |
Collapse
|
41
|
Gaetani L, Chiasserini D, Paolini Paoletti F, Bellomo G, Parnetti L. Required improvements for cerebrospinal fluid-based biomarker tests of Alzheimer's disease. Expert Rev Mol Diagn 2023; 23:1195-1207. [PMID: 37902844 DOI: 10.1080/14737159.2023.2276918] [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: 04/26/2023] [Accepted: 10/25/2023] [Indexed: 11/01/2023]
Abstract
INTRODUCTION Cerebrospinal fluid (CSF) biomarkers represent a well-established tool for diagnosing Alzheimer's disease (AD), independently from the clinical stage, by reflecting the presence of brain amyloidosis (A+) and tauopathy (T+). In front of this important achievement, so far, (i) CSF AD biomarkers have not yet been adopted for routine clinical use in all Centers dedicated to AD, mainly due to inter-lab variation and lack of internationally accepted cutoff values; (ii) we do need to add other biomarkers more suitable to correlate with the clinical stage and disease monitoring; (iii) we also need to detect the co-presence of other 'non-AD' pathologies. AREAS COVERED Efforts to establish standardized cutoff values based on large-scale multi-center studies are discussed. The influence of aging and comorbidities on CSF biomarker levels is also analyzed, and possible solutions are presented, i.e. complementing the A/T/(N) system with markers of axonal damage and synaptic derangement. EXPERT OPINION The first, mandatory need is to reach common cutoff values and defined (automated) methodologies for CSF AD biomarkers. To properly select subjects deserving CSF analysis, blood tests might represent the first-line approach. In those subjects undergoing CSF analysis, multiple biomarkers, able to give a comprehensive and personalized pathophysiological/prognostic information, should be included.
Collapse
Affiliation(s)
- Lorenzo Gaetani
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Davide Chiasserini
- Section of Physiology and Biochemistry, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Giovanni Bellomo
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Lucilla Parnetti
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
42
|
Nilsson J, Cousins KAQ, Gobom J, Portelius E, Chen-Plotkin A, Shaw LM, Grossman M, Irwin DJ, Trojanowski JQ, Zetterberg H, Blennow K, Brinkmalm A. Cerebrospinal fluid biomarker panel of synaptic dysfunction in Alzheimer's disease and other neurodegenerative disorders. Alzheimers Dement 2023; 19:1775-1784. [PMID: 36239248 PMCID: PMC10102247 DOI: 10.1002/alz.12809] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 08/21/2022] [Accepted: 09/02/2022] [Indexed: 11/11/2022]
Abstract
INTRODUCTION Synaptic degeneration is a key part of the pathophysiology of neurodegenerative diseases, and biomarkers reflecting the pathological alterations are greatly needed. METHOD Seventeen synaptic proteins were quantified in a pathology-confirmed cerebrospinal fluid cohort of patients with Alzheimer's disease (AD; n = 63), frontotemporal lobar degeneration (FTLD; n = 53), and Lewy body spectrum of disorders (LBD; n = 21), as well as healthy controls (HC; n = 48). RESULTS Comparisons revealed four distinct patterns: markers decreased across all neurodegenerative conditions compared to HC (the neuronal pentraxins), markers increased across all neurodegenerative conditions (14-3-3 zeta/delta), markers selectively increased in AD compared to other neurodegenerative conditions (neurogranin and beta-synuclein), and markers selectively decreased in LBD and FTLD compared to HC and AD (AP2B1 and syntaxin-1B). DISCUSSION Several of the synaptic proteins may serve as biomarkers for synaptic dysfunction in AD, LBD, and FTLD. Additionally, differential patterns of synaptic protein alterations seem to be present across neurodegenerative diseases. HIGHLIGHTS A panel of synaptic proteins were quantified in the cerebrospinal fluid using mass spectrometry. We compared Alzheimer's disease, frontotemporal degeneration, and Lewy body spectrum of disorders. Pathology was confirmed by autopsy or familial mutations. We discovered synaptic biomarkers for synaptic degeneration and cognitive decline. We found differential patterns of synaptic proteins across neurodegenerative diseases.
Collapse
Affiliation(s)
- Johanna Nilsson
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, SE-43180 Mölndal, Sweden
| | - Katheryn AQ Cousins
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Johan Gobom
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, SE-43180 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, SE-43180 Mölndal, Sweden
| | - Erik Portelius
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, SE-43180 Mölndal, Sweden
| | - Alice Chen-Plotkin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Leslie M Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Murray Grossman
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - David J. Irwin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, SE-43180 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, SE-43180 Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, United Kingdom
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, SE-43180 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, SE-43180 Mölndal, Sweden
| | - Ann Brinkmalm
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, SE-43180 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, SE-43180 Mölndal, Sweden
| |
Collapse
|
43
|
Wang R, Pang SC, Li JY, Li CL, Liu JM, Wang YM, Chen ML, Li YB. A review of the current research on in vivo and in vitro detection for alpha-synuclein: a biomarker of Parkinson's disease. Anal Bioanal Chem 2023; 415:1589-1605. [PMID: 36688984 DOI: 10.1007/s00216-023-04520-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/15/2022] [Accepted: 01/04/2023] [Indexed: 01/24/2023]
Abstract
Parkinson's disease is a health-threatening neurodegenerative disease of the elderly with clinical manifestations of motor and non-motor deficits such as tremor palsy and loss of smell. Alpha-synuclein (α-Syn) is the pathological basis of PD, it can abnormally aggregate into insoluble forms such as oligomers, fibrils, and plaques, causing degeneration of nigrostriatal dopaminergic neurons in the substantia nigra in the patient's brain and the formation of Lewy bodies (LBs) and Lewy neuritis (LN) inclusions. As a result, achieving α-Syn aggregate detection in the early stages of PD can effectively stop or delay the progression of the disease. In this paper, we provide a brief overview and analysis of the molecular structures and α-Syn in vivo and in vitro detection methods, such as mass spectrometry, antigen-antibody recognition, electrochemical sensors, and imaging techniques, intending to provide more technological support for detecting α-Syn early in the disease and intervening in the progression of Parkinson's disease.
Collapse
Affiliation(s)
- Rui Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China.,College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Shu-Chao Pang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Jing-Ya Li
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Chan-Lian Li
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Jun-Miao Liu
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yu-Ming Wang
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Mei-Ling Chen
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Yu-Bo Li
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
44
|
D’Amico R, Tomasello M, Impellizzeri D, Cordaro M, Siracusa R, Interdonato L, Abdelhameed AS, Fusco R, Calabrese V, Cuzzocrea S, Di Paola R. Mechanism of Action of Natural Compounds in Peripheral Multiorgan Dysfunction and Hippocampal Neuroinflammation Induced by Sepsis. Antioxidants (Basel) 2023; 12:antiox12030635. [PMID: 36978883 PMCID: PMC10045853 DOI: 10.3390/antiox12030635] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
Bacterial sepsis induces the production of excessive pro-inflammatory cytokines and oxidative stress, resulting in tissue injury and hyperinflammation. Patients recovering from sepsis have increased rates of central nervous system (CNS) morbidities, which are linked to long-term cognitive impairment, such as neurodegenerative pathologies. This paper focuses on the tissue injury and hyperinflammation observed in the acute phase of sepsis and on the development of long-term neuroinflammation associated with septicemia. Here we evaluate the effects of Coriolus versicolor administration as a novel approach to treat polymicrobial sepsis. Rats underwent cecal ligation and perforation (CLP), and Coriolus versicolor (200 mg/kg in saline) was administered daily by gavage. Survival was monitored, and tissues from vital organs that easily succumb to infection were harvested after 72 h to evaluate the histological changes. Twenty-eight days after CLP, behavioral analyses were performed, and serum and brain (hippocampus) samples were harvested at four weeks from surgery. Coriolus versicolor increased survival and reduced acute tissue injury. Indeed, it reduced the release of pro-inflammatory cytokines in the bloodstream, leading to a reduced chronic inflammation. In the hippocampus, Coriolus versicolor administration restored tight junction expressions, reduce cytokines accumulation and glia activation. It also reduced toll-like receptor 4 (TLR4) and neuronal nitric oxide synthase (nNOS) and the NLR family pyrin domain containing 3 (NLRP3) inflammasome components expression. Coriolus versicolor showed antioxidant activities, restoring glutathione (GSH) levels and catalase and superoxide dismutase (SOD) activities and reducing lipid peroxidation, nitrite and reactive oxygen species (ROS) levels. Importantly, Coriolus versicolor reduced amyloid precursor protein (APP), phosphorylated-Tau (p-Tau), pathologically phosphorylated tau (PHF1), phosphorylated tau (Ser202 and Thr205) (AT8), interferon-induced transmembrane protein 3 (IFITM3) expression, and β-amyloid accumulation induced by CLP. Indeed, Coriolus versicolor restored synaptic dysfunction and behavioral alterations. This research shows the effects of Coriolus versicolor administration on the long-term development of neuroinflammation and brain dysfunction induced by sepsis. Overall, our results demonstrated that Coriolus versicolor administration was able to counteract the degenerative process triggered by sepsis.
Collapse
Affiliation(s)
- Ramona D’Amico
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, n 31, 98166 Messina, Italy
| | - Mario Tomasello
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95124 Catania, Italy
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, n 31, 98166 Messina, Italy
| | - Marika Cordaro
- Department of Biomedical, Dental and Morphological and Functional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, n 31, 98166 Messina, Italy
| | - Livia Interdonato
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, n 31, 98166 Messina, Italy
| | - Ali Saber Abdelhameed
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 14451, Saudi Arabia
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, n 31, 98166 Messina, Italy
- Correspondence:
| | - Vittorio Calabrese
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95124 Catania, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, n 31, 98166 Messina, Italy
| | - Rosanna Di Paola
- Department of Vererinary Sciences, University of Messina, Viale Annunziata, 98168 Messina, Italy
| |
Collapse
|
45
|
Arvidsson Rådestig M, Skoog I, Skillbäck T, Zetterberg H, Kern J, Zettergren A, Andreasson U, Wetterberg H, Kern S, Blennow K. Cerebrospinal fluid biomarkers of axonal and synaptic degeneration in a population-based sample. Alzheimers Res Ther 2023; 15:44. [PMID: 36869347 PMCID: PMC9983206 DOI: 10.1186/s13195-023-01193-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/14/2023] [Indexed: 03/05/2023]
Abstract
BACKGROUND Neurofilament light (NfL) and neurogranin (Ng) are promising candidate AD biomarkers, reflecting axonal and synaptic damage, respectively. Since there is a need to understand the synaptic and axonal damage in preclinical Alzheimer's disease (AD), we aimed to determine the cerebrospinal fluid (CSF) levels of NfL and Ng in cognitively unimpaired elderly from the Gothenburg H70 Birth Cohort Studies classified according to the amyloid/tau/neurodegeneration (A/T/N) system. METHODS The sample consisted of 258 cognitively unimpaired older adults (age 70, 129 women and 129 men) from the Gothenburg Birth Cohort Studies. We compared CSF NfL and Ng concentrations in A/T/N groups using Student's T-test and ANCOVA. RESULTS CSF NfL concentration was higher in the A-T-N+ group (p=0.001) and the A-T+N+ group (p=0.006) compared with A-T-N-. CSF Ng concentration was higher in the A-T-N+, A-T+N+, A+T-N+, and A+T+N+ groups (p<0.0001) compared with A-T-N-. We found no difference in NfL or Ng concentration in A+ compared with A- (disregarding T- and N- status), whereas those with N+ had higher concentrations of NfL and Ng compared with N- (p<0.0001) (disregarding A- and T- status). CONCLUSIONS CSF NfL and Ng concentrations are increased in cognitively normal older adults with biomarker evidence of tau pathology and neurodegeneration.
Collapse
Affiliation(s)
- Maya Arvidsson Rådestig
- Department of Neuropsychiatric Epidemiology Unit, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Ingmar Skoog
- Department of Neuropsychiatric Epidemiology Unit, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Department of Psychiatry, Cognition and Old Age Psychiatry, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Tobias Skillbäck
- Department of Neuropsychiatric Epidemiology Unit, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden. .,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK.,UK Dementia Research Institute at UCL, London, WC1N 3BG, UK.,Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Jürgen Kern
- Department of Neuropsychiatric Epidemiology Unit, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Anna Zettergren
- Department of Neuropsychiatric Epidemiology Unit, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Ulf Andreasson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Hanna Wetterberg
- Department of Neuropsychiatric Epidemiology Unit, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Silke Kern
- Department of Neuropsychiatric Epidemiology Unit, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Department of Psychiatry, Cognition and Old Age Psychiatry, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| |
Collapse
|
46
|
Ribarič S. Detecting Early Cognitive Decline in Alzheimer's Disease with Brain Synaptic Structural and Functional Evaluation. Biomedicines 2023; 11:355. [PMID: 36830892 PMCID: PMC9952956 DOI: 10.3390/biomedicines11020355] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/22/2023] [Accepted: 01/24/2023] [Indexed: 01/28/2023] Open
Abstract
Early cognitive decline in patients with Alzheimer's (AD) is associated with quantifiable structural and functional connectivity changes in the brain. AD dysregulation of Aβ and tau metabolism progressively disrupt normal synaptic function, leading to loss of synapses, decreased hippocampal synaptic density and early hippocampal atrophy. Advances in brain imaging techniques in living patients have enabled the transition from clinical signs and symptoms-based AD diagnosis to biomarkers-based diagnosis, with functional brain imaging techniques, quantitative EEG, and body fluids sampling. The hippocampus has a central role in semantic and episodic memory processing. This cognitive function is critically dependent on normal intrahippocampal connections and normal hippocampal functional connectivity with many cortical regions, including the perirhinal and the entorhinal cortex, parahippocampal cortex, association regions in the temporal and parietal lobes, and prefrontal cortex. Therefore, decreased hippocampal synaptic density is reflected in the altered functional connectivity of intrinsic brain networks (aka large-scale networks), including the parietal memory, default mode, and salience networks. This narrative review discusses recent critical issues related to detecting AD-associated early cognitive decline with brain synaptic structural and functional markers in high-risk or neuropsychologically diagnosed patients with subjective cognitive impairment or mild cognitive impairment.
Collapse
Affiliation(s)
- Samo Ribarič
- Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Zaloška 4, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
47
|
Barba L, Abu Rumeileh S, Bellomo G, Paolini Paoletti F, Halbgebauer S, Oeckl P, Steinacker P, Massa F, Gaetani L, Parnetti L, Otto M. Cerebrospinal fluid β-synuclein as a synaptic biomarker for preclinical Alzheimer's disease. J Neurol Neurosurg Psychiatry 2023; 94:83-86. [PMID: 35944974 DOI: 10.1136/jnnp-2022-329124] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 07/12/2022] [Indexed: 02/02/2023]
Abstract
INTRODUCTION β-synuclein (β-syn) is a presynaptic protein, whose cerebrospinal fluid (CSF) levels are increased in patients with Alzheimer's diseases (AD) showing mild cognitive impairment (MCI) and dementia (dem). Here, we aimed to investigate CSF β-syn in subjects at different AD stages, including preclinical AD (pre-AD), and to compare its behaviour with another synaptic biomarker, α-synuclein (α-syn), and two biomarkers of neuro-axonal damage, namely neurofilament light chain protein (NfL) and total tau protein (t-tau). METHODS We measured β-syn, α-syn, t-tau and NfL in CSF of 75 patients with AD (pre-AD n=17, MCI-AD n=28, dem-AD n=30) and 35 controls (subjective memory complaints, SMC-Ctrl n=13, non-degenerative neurological disorders, Dis-Ctrl n=22). RESULTS CSF β-syn, α-syn, t-tau were significantly elevated in pre-AD patients compared with controls (p<0.0001, p=0.02 and p=0.0001, respectively), while NfL only increased in dem-AD (p=0.001). Pre-AD cases showed lower t-tau concentrations than MCI-AD (p=0.04) and dem-AD (p=0.01). CSF β-syn had the best diagnostic performance for the discrimination of pre-AD subjects from all controls (area under the curve, AUC=0.97) and from SMC-Ctrl subjects (AUC=0.99). DISCUSSION CSF β-syn increases in the whole AD continuum since the preclinical stage and represents a promising biomarker of synaptic damage in AD.
Collapse
Affiliation(s)
- Lorenzo Barba
- Department of Neurology, University Hospital Halle, Halle (Saale), Germany.,Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Samir Abu Rumeileh
- Department of Neurology, University Hospital Halle, Halle (Saale), Germany
| | - Giovanni Bellomo
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | | | - Patrick Oeckl
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Petra Steinacker
- Department of Neurology, University Hospital Halle, Halle (Saale), Germany
| | - Federico Massa
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Lorenzo Gaetani
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Lucilla Parnetti
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Markus Otto
- Department of Neurology, University Hospital Halle, Halle (Saale), Germany .,Department of Neurology, University of Ulm, Ulm, Germany
| |
Collapse
|
48
|
Rådestig MA, Skoog J, Zetterberg H, Skillbäck T, Zettergren A, Sterner TR, Fässberg MM, Sacuiu S, Waern M, Wetterberg H, Blennow K, Skoog I, Kern S. Subtle Differences in Cognition in 70-Year-Olds with Elevated Cerebrospinal Fluid Neurofilament Light and Neurogranin: A H70 Cross-Sectional Study. J Alzheimers Dis 2023; 91:291-303. [PMID: 36617786 PMCID: PMC9881027 DOI: 10.3233/jad-220452] [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] [Accepted: 10/19/2022] [Indexed: 01/05/2023]
Abstract
BACKGROUND Most research on cerebrospinal fluid (CSF) neurofilament light protein (NfL) as a marker for neurodegeneration and neurogranin (Ng) for synaptic dysfunction has largely focused on clinical cohorts rather than population-based samples. OBJECTIVE We hypothesized that increased CSF levels of NfL and Ng are associated with subtle cognitive deficits in cognitively unimpaired (CU) older adults. METHODS The sample was derived from the Gothenburg H70 Birth Cohort Studies and comprised 258 CU 70-year-olds, with a Clinical Dementia Rating score of zero. All participants underwent extensive cognitive testing. CSF levels of NfL and Ng, as well as amyloid β1 - 42, total tau, and phosphorylated tau, were measured. RESULTS Participants with high CSF NfL performed worse in one memory-based test (Immediate recall, p = 0.013) and a language test (FAS, p = 0.016). Individuals with high CSF Ng performed worse on the memory-based test Supra Span (p = 0.035). When stratified according to CSF tau and Aβ42 concentrations, participants with high NfL and increased tau performed worse on a memory test than participants normal tau concentrations (Delayed recall, p = 0.003). In participants with high NfL, those with pathologic Aβ42 concentrations performed worse on the Delayed recall memory (p = 0.044). In the high Ng group, participants with pathological Aβ42 concentrations had lower MMSE scores (p = 0.027). However, in regression analysis we found no linear correlations between CSF NfL or CSF Ng in relation to cognitive tests when controlled for important co-variates. CONCLUSION Markers of neurodegeneration and synaptic pathology might be associated with subtle signs of cognitive decline in a population-based sample of 70-year-olds.
Collapse
Affiliation(s)
- Maya Arvidsson Rådestig
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Johan Skoog
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychology, University of Gothenburg, Gothenburg, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Psychiatry/Cognition and Old Age Psychiatry Clinic, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- UCL Institute of Neurology, Queen Square, London, UK
- The UK Dementia Research Institute, UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | - Tobias Skillbäck
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Anna Zettergren
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Therese Rydberg Sterner
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Madeleine Mellqvist Fässberg
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Simona Sacuiu
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Psychiatry/Cognition and Old Age Psychiatry Clinic, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
- Memory Disorders Clinic, Theme Inflammation and Aging, Karolinska University Hospital, Region Stockholm, Stockholm, Sweden
- Department of Neurobiology, Care Sciences and Society (NVS), Clinical Geriatric, Karolinska Institute, Stockholm, Sweden
| | - Margda Waern
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Psychosis Clinic, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Hanna Wetterberg
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Ingmar Skoog
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Psychiatry/Cognition and Old Age Psychiatry Clinic, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Silke Kern
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Psychiatry/Cognition and Old Age Psychiatry Clinic, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| |
Collapse
|
49
|
van den Berg E, Nilsson J, Kersten I, Brinkmalm G, de Kort AM, Klijn CJ, Schreuder FH, Jäkel L, Gobom J, Portelius E, Zetterberg H, Brinkmalm A, Blennow K, Kuiperij HB, Verbeek MM. Cerebrospinal Fluid Panel of Synaptic Proteins in Cerebral Amyloid Angiopathy and Alzheimer's Disease. J Alzheimers Dis 2023; 92:467-475. [PMID: 36776062 PMCID: PMC10041443 DOI: 10.3233/jad-220977] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2023] [Indexed: 02/10/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) and cerebral amyloid angiopathy (CAA) share pathogenic pathways related to amyloid-β deposition. Whereas AD is known to affect synaptic function, such an association for CAA remains yet unknown. OBJECTIVE We therefore aimed to investigate synaptic dysfunction in CAA. METHODS Multiple reaction monitoring mass spectrometry was used to quantify cerebrospinal fluid (CSF) concentrations of 15 synaptic proteins in CAA and AD patients, and age- and sex-matched cognitively unimpaired controls. RESULTS We included 25 patients with CAA, 49 patients with AD, and 25 controls. Only neuronal pentraxin-2 levels were decreased in the CSF of CAA patients compared with controls (p = 0.04). CSF concentrations of 12 other synaptic proteins were all increased in AD compared with CAA or controls (all p≤0.01) and were unchanged between CAA and controls. Synaptic protein concentrations in the subgroup of CAA patients positive for AD biomarkers (CAA/ATN+; n = 6) were similar to AD patients, while levels in CAA/ATN- (n = 19) were comparable with those in controls. A regression model including all synaptic proteins differentiated CAA from AD at high accuracy levels (area under the curve 0.987). CONCLUSION In contrast to AD, synaptic CSF biomarkers were found to be largely unchanged in CAA. Moreover, concomitant AD pathology in CAA is associated with abnormal synaptic protein levels. Impaired synaptic function in AD was confirmed in this independent cohort. Our findings support an apparent differential involvement of synaptic dysfunction in CAA and AD and may reflect distinct pathological mechanisms.
Collapse
Affiliation(s)
- Emma van den Berg
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Johanna Nilsson
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Iris Kersten
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gunnar Brinkmalm
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Anna M. de Kort
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Catharina J.M. Klijn
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Floris H.B.M. Schreuder
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lieke Jäkel
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Johan Gobom
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Erik Portelius
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, United Kingdom
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Ann Brinkmalm
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - H. Bea Kuiperij
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marcel M. Verbeek
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, TheNetherlands
| |
Collapse
|
50
|
Malpetti M, Jones PS, Cope TE, Holland N, Naessens M, Rouse MA, Rittman T, Savulich G, Whiteside DJ, Street D, Fryer TD, Hong YT, Milicevic Sephton S, Aigbirhio FI, O′Brien JT, Rowe JB. Synaptic Loss in Frontotemporal Dementia Revealed by [ 11 C]UCB-J Positron Emission Tomography. Ann Neurol 2023; 93:142-154. [PMID: 36321699 PMCID: PMC10099663 DOI: 10.1002/ana.26543] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 09/28/2022] [Accepted: 10/30/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Synaptic loss is an early feature of neurodegenerative disease models, and is severe in post mortem clinical studies, including frontotemporal dementia. Positron emission tomography (PET) with radiotracers that bind to synaptic vesicle glycoprotein 2A enables quantification of synaptic density in vivo. This study used [11 C]UCB-J PET in participants with behavioral variant frontotemporal dementia (bvFTD), testing the hypothesis that synaptic loss is severe and related to clinical severity. METHODS Eleven participants with clinically probable bvFTD and 25 age- and sex-matched healthy controls were included. Participants underwent dynamic [11 C]UCB-J PET, structural magnetic resonance imaging, and a neuropsychological battery, including the revised Addenbrooke Cognitive Examination, and INECO frontal screening. General linear models compared [11 C]UCB-J binding potential maps and gray matter volume between groups, and assessed associations between synaptic density and clinical severity in patients. Analyses were also performed using partial volume corrected [11 C]UCB-J binding potential from regions of interest (ROIs). RESULTS Patients with bvFTD showed severe synaptic loss compared to controls. [11 C]UCB-J binding was reduced bilaterally in medial and dorsolateral frontal regions, inferior frontal gyri, anterior and posterior cingulate gyrus, insular cortex, and medial temporal lobe. Synaptic loss in the frontal and cingulate regions correlated significantly with cognitive impairments. Synaptic loss was more severe than atrophy. Results from ROI-based analyses mirrored the voxelwise results. INTERPRETATION In accordance with preclinical models, and human postmortem evidence, there is widespread frontotemporal loss of synapses in symptomatic bvFTD, in proportion to severity. [11 C]UCB-J PET could support translational studies and experimental medicine strategies for new disease-modifying treatments for neurodegeneration. ANN NEUROL 2023;93:142-154.
Collapse
Affiliation(s)
- Maura Malpetti
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Cambridge University Hospitals National Health Service Foundation TrustCambridgeUK
| | - P. Simon Jones
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | - Thomas E. Cope
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Cambridge University Hospitals National Health Service Foundation TrustCambridgeUK
- Medical Research Council Cognition and Brain Sciences UnitUniversity of CambridgeUK
| | - Negin Holland
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Cambridge University Hospitals National Health Service Foundation TrustCambridgeUK
| | - Michelle Naessens
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | - Matthew A. Rouse
- Medical Research Council Cognition and Brain Sciences UnitUniversity of CambridgeUK
| | - Timothy Rittman
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Cambridge University Hospitals National Health Service Foundation TrustCambridgeUK
| | | | - David J. Whiteside
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Cambridge University Hospitals National Health Service Foundation TrustCambridgeUK
| | - Duncan Street
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Cambridge University Hospitals National Health Service Foundation TrustCambridgeUK
| | - Tim D. Fryer
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Wolfson Brain Imaging CentreUniversity of CambridgeCambridgeUK
| | - Young T. Hong
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Wolfson Brain Imaging CentreUniversity of CambridgeCambridgeUK
| | - Selena Milicevic Sephton
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Wolfson Brain Imaging CentreUniversity of CambridgeCambridgeUK
| | - Franklin I. Aigbirhio
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Wolfson Brain Imaging CentreUniversity of CambridgeCambridgeUK
| | - John T. O′Brien
- Cambridge University Hospitals National Health Service Foundation TrustCambridgeUK
- Department of PsychiatryUniversity of CambridgeCambridgeUK
| | - James B. Rowe
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Cambridge University Hospitals National Health Service Foundation TrustCambridgeUK
- Medical Research Council Cognition and Brain Sciences UnitUniversity of CambridgeUK
| |
Collapse
|