1
|
Youn W, Yun M, Lee CJ, Schöll M. Cautions on utilizing plasma GFAP level as a biomarker for reactive astrocytes in neurodegenerative diseases. Mol Neurodegener 2025; 20:54. [PMID: 40346659 DOI: 10.1186/s13024-025-00846-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 04/17/2025] [Indexed: 05/11/2025] Open
Affiliation(s)
- Wongu Youn
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, Republic of Korea
| | - Mijin Yun
- Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, Republic of Korea.
| | - Michael Schöll
- Department of Psychiatry and Neurochemistry and the Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, 41345, Sweden.
- Dementia Research Centre, Queen Square Institute of Neurology, University College London, London, WC1E, UK.
- Department of Neuropsychiatry, Sahlgrenska University Hospital, Mölndal, 43141, Sweden.
| |
Collapse
|
2
|
Heneka MT, van der Flier WM, Jessen F, Hoozemanns J, Thal DR, Boche D, Brosseron F, Teunissen C, Zetterberg H, Jacobs AH, Edison P, Ramirez A, Cruchaga C, Lambert JC, Laza AR, Sanchez-Mut JV, Fischer A, Castro-Gomez S, Stein TD, Kleineidam L, Wagner M, Neher JJ, Cunningham C, Singhrao SK, Prinz M, Glass CK, Schlachetzki JCM, Butovsky O, Kleemann K, De Jaeger PL, Scheiblich H, Brown GC, Landreth G, Moutinho M, Grutzendler J, Gomez-Nicola D, McManus RM, Andreasson K, Ising C, Karabag D, Baker DJ, Liddelow SA, Verkhratsky A, Tansey M, Monsonego A, Aigner L, Dorothée G, Nave KA, Simons M, Constantin G, Rosenzweig N, Pascual A, Petzold GC, Kipnis J, Venegas C, Colonna M, Walter J, Tenner AJ, O'Banion MK, Steinert JR, Feinstein DL, Sastre M, Bhaskar K, Hong S, Schafer DP, Golde T, Ransohoff RM, Morgan D, Breitner J, Mancuso R, Riechers SP. Neuroinflammation in Alzheimer disease. Nat Rev Immunol 2025; 25:321-352. [PMID: 39653749 DOI: 10.1038/s41577-024-01104-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2024] [Indexed: 02/20/2025]
Abstract
Increasing evidence points to a pivotal role of immune processes in the pathogenesis of Alzheimer disease, which is the most prevalent neurodegenerative and dementia-causing disease of our time. Multiple lines of information provided by experimental, epidemiological, neuropathological and genetic studies suggest a pathological role for innate and adaptive immune activation in this disease. Here, we review the cell types and pathological mechanisms involved in disease development as well as the influence of genetics and lifestyle factors. Given the decade-long preclinical stage of Alzheimer disease, these mechanisms and their interactions are driving forces behind the spread and progression of the disease. The identification of treatment opportunities will require a precise understanding of the cells and mechanisms involved as well as a clear definition of their temporal and topographical nature. We will also discuss new therapeutic strategies for targeting neuroinflammation, which are now entering the clinic and showing promise for patients.
Collapse
Affiliation(s)
- Michael T Heneka
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette/Belvaux, Luxembourg.
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Frank Jessen
- Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
| | - Jeroen Hoozemanns
- Department of Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Dietmar Rudolf Thal
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium
- Laboratory for Neuropathology, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- Laboratory for Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), Leuven, Belgium
| | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | | | - Charlotte Teunissen
- Department of Laboratory Medicine, VUMC Amsterdam, Amsterdam, The Netherlands
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Andreas H Jacobs
- European Institute for Molecular Imaging, University of Münster, Münster, Germany
| | - Paul Edison
- Division of Neurology, Department of Brain Sciences, Imperial College London, London, UK
| | - Alfredo Ramirez
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
- Cluster of Excellence Cellular Stress Response in Aging-associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Carlos Cruchaga
- Department of Psychiatry, Washington School of Medicine in St. Louis, St. Louis, MO, USA
| | - Jean-Charles Lambert
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Agustin Ruiz Laza
- ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), Barcelona, Spain
| | - Jose Vicente Sanchez-Mut
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Alicante, Spain
| | - Andre Fischer
- Clinic for Psychiatry and Psychotherapy, University Medical Center, Georg-August-University Göttingen, Göttingen, Germany
- Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Centre for Neurodegenerative Disease (DZNE), Göttingen, Germany
| | - Sergio Castro-Gomez
- Center for Neurology, Clinic of Parkinson, Sleep and Movement Disorders, University Hospital Bonn, University of Bonn, Bonn, Germany
- Institute of Physiology II, University Hospital Bonn, University of Bonn, Bonn, Germany
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Thor D Stein
- Boston University Alzheimer's Disease Research Center and CTE Center, Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Luca Kleineidam
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurodegenerative Disease and Geriatric Psychiatry, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Michael Wagner
- Department of Neurodegenerative Disease and Geriatric Psychiatry, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Jonas J Neher
- Biomedical Center Munich, Biochemistry, Medical Faculty, LMU Munich, Munich, Germany
- Neuroimmunology and Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Colm Cunningham
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland
- Trinity College Institute of Neuroscience (TCIN), Trinity College Dublin, Dublin, Ireland
| | - Sim K Singhrao
- Brain and Behaviour Centre, Faculty of Clinical and Biomedical Sciences, School of Dentistry, University of Central Lancashire, Preston, UK
| | - Marco Prinz
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
- Signalling Research Centers BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Johannes C M Schlachetzki
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Oleg Butovsky
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kilian Kleemann
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Philip L De Jaeger
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Hannah Scheiblich
- Center for Neurology, Clinic of Parkinson, Sleep and Movement Disorders, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Guy C Brown
- Deparment of Biochemistry, University of Cambridge, Cambridge, UK
| | - Gary Landreth
- School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Miguel Moutinho
- School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Jaime Grutzendler
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Diego Gomez-Nicola
- School of Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Róisín M McManus
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Katrin Andreasson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Christina Ising
- Cluster of Excellence Cellular Stress Response in Aging-associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Deniz Karabag
- Cluster of Excellence Cellular Stress Response in Aging-associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Darren J Baker
- Department of Paediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Shane A Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York City, NY, USA
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York City, NY, USA
- Department of Ophthalmology, NYU Grossman School of Medicine, New York City, NY, USA
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Malu Tansey
- College of Medicine, University of Florida, Gainsville, FL, USA
| | - Alon Monsonego
- Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Guillaume Dorothée
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine (CRSA), Hôpital Saint-Antoine, Paris, France
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - Gabriela Constantin
- Section of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| | - Neta Rosenzweig
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alberto Pascual
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Gabor C Petzold
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Vascular Neurology, University of Bonn, Bonn, Germany
| | - Jonathan Kipnis
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Center for Brain Immunology and Glia (BIG), Washington University School of Medicine, St. Louis, MO, USA
| | - Carmen Venegas
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette/Belvaux, Luxembourg
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
- Instituto Biosanitario de Granada (ibs.Granada), Granada, Spain
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jochen Walter
- Center of Neurology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Andrea J Tenner
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, CA, USA
- Department of Neurobiology and Behaviour, University of California Irvine, Irvine, CA, USA
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Irvine, Irvine, CA, USA
| | - M Kerry O'Banion
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
- Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA
| | - Joern R Steinert
- Faculty of Medicine and Health Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| | - Douglas L Feinstein
- Department of NeuroAnesthesia, University of Illinois at Chicago, Chicago, IL, USA
| | - Magdalena Sastre
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Kiran Bhaskar
- Department of Molecular Genetics & Microbiology and Neurology, University of New Mexico, Albuquerque, NM, USA
| | - Soyon Hong
- UK Dementia Research Institute, Institute of Neurology, University College London, London, UK
| | - Dorothy P Schafer
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Todd Golde
- Department of Pharmacology and Chemical Biology, Emory Center for Neurodegenerative Disease, Emory University, Atlanta, GA, USA
- Department of Neurology, Emory Center for Neurodegenerative Disease, Emory University, Atlanta, GA, USA
| | | | - David Morgan
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - John Breitner
- Department of Psychiatry, McGill University Faculty of Medicine, Montreal, Québec, Canada
| | - Renzo Mancuso
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, University of Antwerp, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Sean-Patrick Riechers
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette/Belvaux, Luxembourg
| |
Collapse
|
3
|
Ye K, Han X, Tian M, Liu L, Gao X, Xia Q, Wang D. Analysis of human brain RNA-seq data reveals combined effects of 4 types of RNA modifications and 18 types of programmed cell death on Alzheimer's disease. J Transl Med 2025; 23:396. [PMID: 40181382 PMCID: PMC11969709 DOI: 10.1186/s12967-025-06324-6] [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: 11/30/2024] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND RNA modification plays a critical role in Alzheimer's disease (AD) by modulating the expression and function of AD-related genes, thereby affecting AD occurrence and progression. Programmed cell death is closely related to neuronal death and associated with neuronal loss and cognitive function changes in AD. However, the mechanism of their joint action on AD remains unknown and requires further exploration. METHODS We used the MSBB RNA-seq dataset to analyze the correlation between RNA modification, programmed cell death, and AD. We used combined studies of RNA modification and programmed cell death to distinguish subgroups of patients, and the results highlight the strong correlation between RNA modification-related programmed cell death and AD. A weighted gene co-expression network was constructed, and the pivotal roles of programmed cell death genes in key modules were identified. Finally, by combining unsupervised consensus clustering, gene co-expression networks, and machine learning algorithms, an RNA modification-related programmed cell death network was constructed, and the pivotal roles of programmed cell death genes in key modules were identified. An RNA modification-related programmed cell death risk score was calculated to predict the occurrence of AD. RESULTS RPCD-related genes classified patients into subgroups with distinct clinical characteristics. Nineteen key genes were identified and an RPCD risk score was constructed based on the key genes. This score can be used for the diagnosis of AD and the assessment of disease progression in patients. The diagnostic efficacy of the RPCD risk score and the key genes was validated in the ROSMAP, GEO, and ADNI datasets. CONCLUSION This study uncovered that RNA modification-related PCD is of significance for AD progression and early prediction, providing insights from a new perspective for the study of disease mechanisms in AD.
Collapse
Affiliation(s)
- Ke Ye
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Xinyu Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Mengjie Tian
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Lulu Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Xu Gao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
- Key Laboratory of Heilongjiang Province for Genetically Modified Animals, Harbin Medical University, Harbin, 150081, Heilongjiang, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, Heilongjiang, China
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin, 150081, Heilongjiang, China
| | - Qing Xia
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Dayong Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China.
- Key Laboratory of Heilongjiang Province for Genetically Modified Animals, Harbin Medical University, Harbin, 150081, Heilongjiang, China.
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, Heilongjiang, China.
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin, 150081, Heilongjiang, China.
| |
Collapse
|
4
|
Rocha FM, Roy A, Varshney M, Kumar A. Mapping reactive astrogliosis in Parkinson's brain with astroglial tracers BU99008 and Deprenyl: New insights from a multi-marker postmortem study. Alzheimers Dement 2025; 21:e14488. [PMID: 39936538 PMCID: PMC11848164 DOI: 10.1002/alz.14488] [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: 09/17/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 02/13/2025]
Abstract
BACKGROUND Despite significant astrocytic involvement in Parkinson's disease (PD), the knowledge regarding the role of reactive astrogliosis is still at the surface level; largely due to lack of specific biomarkers to track these processes. Novel astroglial PET-tracers BU99008 and Deprenyl, hold immense potential for visualizing reactive astrogliosis in PD. However, they have not been thoroughly investigated in PD. METHODS We employed a multi-marker approach and performed in vitro radioligand binding and autoradiography studies with 3H-BU99008 and 3H-Deprenyl together with astrocytic immunofluorescence and morphometric analyses in the frontal cortex, temporal cortex, caudate and putamen brain regions of PD (n = 4) and control (n = 7) cases. RESULTS AND DISCUSSION 3H-BU99008 and 3H-Deprenyl showed distinct binding behavior and displayed a diverse array of binding sites (single or multiple) in PD and control brains. Importantly, 3H-BU99008 and 3H-Deprenyl autoradiography studies captured pronounced reactive astrogliosis in PD brain regions, corroborated by marked changes in astrocytic markers, morphology, and cellular processes. HIGHLIGHTS Astroglial tracers BU99008 and Deprenyl displayed a range of binding sites with different levels of affinity and proportions (%) in healthy control (CN) and Parkinson's disease (PD) brains. Astroglial tracers BU99008 and Deprenyl showed a highly specific (permanent) high-affinity (HA) binding site in the nanomolar range, which might be consistent across different pathologies. Astroglial tracers BU99008 and Deprenyl highlighted distinct tracer binding behavior, indicating that they might be targeting different subpopulations or specific states of astrocytes in CN and PD brains. Astroglial tracers BU99008 and Deprenyl captured prominent reactive astrogliosis at the advanced/end stages of PD, substantiated by a significant increase in intercellular adhesion molecule 1 (ICAM-1)-positive reactive astrocytes and marked changes in astrocytic morphology and processes.
Collapse
Affiliation(s)
- Filipa M. Rocha
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and SocietyKarolinska InstitutetStockholmSweden
| | - Avishek Roy
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and SocietyKarolinska InstitutetStockholmSweden
| | - Mukesh Varshney
- Division of Clinical Microbiology, Department of Laboratory MedicineKarolinska InstitutetStockholmSweden
| | - Amit Kumar
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and SocietyKarolinska InstitutetStockholmSweden
| |
Collapse
|
5
|
Huang K, Hsiao I, Huang C, Huang C, Chang H, Huang S, Lin K, Ma M, Huang C, Chang C. The Taiwan-ADNI workflow toward integrating plasma p-tau217 into prediction models for the risk of Alzheimer's disease and tau burden. Alzheimers Dement 2025; 21:e14297. [PMID: 39777990 PMCID: PMC11772711 DOI: 10.1002/alz.14297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 01/11/2025]
Abstract
INTRODUCTION We integrated plasma biomarkers from the Taiwan Alzheimer's Disease Neuroimaging Initiative and propose a workflow to identify individuals showing amyloid-positive positron emission tomography (PET) with low/intermediate tau burden based on [18F]Florzolotau PET-based quantification. METHODS We assessed 361 participants across the Alzheimer's disease (AD) and non-AD continuum and measured plasma phosphorylated tau (p-tau)217, p-tau181, amyloid beta (Aβ)42/40 ratio, neurofilament light chain, and glial fibrillary acidic protein levels at two medical centers. We evaluated the diagnostic potential of these biomarkers. RESULTS Among all plasma biomarkers, p-tau217 had the highest consistency with amyloid PET results (area under the curve = 0.94), and a cutoff value could have reduced the number of confirmatory amyloid PET scans by 57.5%. In amyloid PET-positive cases intending to use anti-amyloid therapy, p-tau217 level, along with clinical parameters, had the highest predictive ability for low/intermediate tau burden. DISCUSSION A two-step workflow based on p-tau217 and confirmatory amyloid PET could accurately classify AD patients showing low/intermediate tau burden. HIGHLIGHTS The emergence of anti-amyloid therapy increases the need to accurately diagnose Alzheimer's disease (AD). The use of plasma biomarkers, especially phosphorylated tau 217 (p-tau217), can help in the diagnosis of AD. P-tau217 is a better predictor of amyloid positron emission tomography (PET) positivity than other core biomarkers. In amyloid PET-positive individuals, p-tau217 can predict tau burden. We propose a two-step workflow to identify AD cases suitable for treatment.
Collapse
Affiliation(s)
- Kuo‐Lun Huang
- Department of NeurologyLinkou Chang Gung Memorial HospitalChang Gung UniversityTaoyuanTaiwan
| | - Ing‐Tsung Hsiao
- Department of Medical Imaging and Radiological Sciences and Healthy Aging Research CenterChang Gung UniversityTaoyuanTaiwan
- Department of Nuclear MedicineLinkou Chang Gung Memorial HospitalChang Gung UniversityTaoyuanTaiwan
| | - Chi‐Wei Huang
- Department of NeurologyCognition and Aging CenterInstitute for Translational Research in BiomedicineKaohsiung Chang Gung Memorial HospitalChang Gung University College of MedicineKaohsiung CityTaiwan
| | - Chung‐Guei Huang
- Department of Medical Laboratory, Linkou Chang Gung Memorial Hospital, Department of Medical Biotechnology and Laboratory ScienceChang Gung UniversityTaoyuanTaiwan
| | - Hsin‐I Chang
- Department of NeurologyCognition and Aging CenterInstitute for Translational Research in BiomedicineKaohsiung Chang Gung Memorial HospitalChang Gung University College of MedicineKaohsiung CityTaiwan
| | - Shu‐Hua Huang
- Department of Nuclear Medicine, Kaohsiung Chang Gung Memorial HospitalChang Gung University College of MedicineKaohsiung CityTaiwan
| | - Kun‐Ju Lin
- Department of Nuclear MedicineLinkou Chang Gung Memorial HospitalChang Gung UniversityTaoyuanTaiwan
| | - Mi‐Chia Ma
- Department of Statistics, College of ManagementNational Cheng Kung UniversityTainanTaiwan
| | - Chin‐Chang Huang
- Department of NeurologyLinkou Chang Gung Memorial HospitalChang Gung UniversityTaoyuanTaiwan
| | - Chiung‐Chih Chang
- Department of NeurologyCognition and Aging CenterInstitute for Translational Research in BiomedicineKaohsiung Chang Gung Memorial HospitalChang Gung University College of MedicineKaohsiung CityTaiwan
- School of Medicine, College of MedicineNational Sun Yat‐sen UniversityKaohsiungTaiwan
| |
Collapse
|
6
|
Gogishvili D, Honey MIJ, Verberk IMW, Vermunt L, Hol EM, Teunissen CE, Abeln S. The GFAP proteoform puzzle: How to advance GFAP as a fluid biomarker in neurological diseases. J Neurochem 2025; 169:e16226. [PMID: 39289040 DOI: 10.1111/jnc.16226] [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: 06/05/2024] [Revised: 07/19/2024] [Accepted: 09/01/2024] [Indexed: 09/19/2024]
Abstract
Glial fibrillary acidic protein (GFAP) is a well-established biomarker of reactive astrogliosis in the central nervous system because of its elevated levels following brain injury and various neurological disorders. The advent of ultra-sensitive methods for measuring low-abundant proteins has significantly enhanced our understanding of GFAP levels in the serum or plasma of patients with diverse neurological diseases. Clinical studies have demonstrated that GFAP holds promise both as a diagnostic and prognostic biomarker, including but not limited to individuals with Alzheimer's disease. GFAP exhibits diverse forms and structures, herein referred to as its proteoform complexity, encompassing conformational dynamics, isoforms and post-translational modifications (PTMs). In this review, we explore how the proteoform complexity of GFAP influences its detection, which may affect the differential diagnostic performance of GFAP in different biological fluids and can provide valuable insights into underlying biological processes. Additionally, proteoforms are often disease-specific, and our review provides suggestions and highlights areas to focus on for the development of new assays for measuring GFAP, including isoforms, PTMs, discharge mechanisms, breakdown products, higher-order species and interacting partners. By addressing the knowledge gaps highlighted in this review, we aim to support the clinical translation and interpretation of GFAP in both CSF and blood and the development of reliable, reproducible and specific prognostic and diagnostic tests. To enhance disease pathology comprehension and optimise GFAP as a biomarker, a thorough understanding of detected proteoforms in biofluids is essential.
Collapse
Affiliation(s)
- Dea Gogishvili
- Bioinformatics, Computer Science Department, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- AI Technology for Life, Department of Computing and Information Sciences, Department of Biology, Utrecht University, Utrecht, The Netherlands
| | - Madison I J Honey
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam University Medical Centers, Amsterdam Neuroscience, Vrije Universiteit, Amsterdam, The Netherlands
| | - Inge M W Verberk
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam University Medical Centers, Amsterdam Neuroscience, Vrije Universiteit, Amsterdam, The Netherlands
| | - Lisa Vermunt
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam University Medical Centers, Amsterdam Neuroscience, Vrije Universiteit, Amsterdam, The Netherlands
| | - Elly M Hol
- Department of Translational Neuroscience, UMC Utrecht Brain Centre, University Medical Centre Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam University Medical Centers, Amsterdam Neuroscience, Vrije Universiteit, Amsterdam, The Netherlands
| | - Sanne Abeln
- Bioinformatics, Computer Science Department, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- AI Technology for Life, Department of Computing and Information Sciences, Department of Biology, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
7
|
Kovacech B, Cullen NC, Novak P, Hanes J, Kontsekova E, Katina S, Parrak V, Fresser M, Vanbrabant J, Feldman HH, Winblad B, Stoops E, Vanmechelen E, Zilka N. Post hoc analysis of ADAMANT, a phase 2 clinical trial of active tau immunotherapy with AADvac1 in patients with Alzheimer's disease, positive for plasma p-tau217. Alzheimers Res Ther 2024; 16:254. [PMID: 39580468 PMCID: PMC11585249 DOI: 10.1186/s13195-024-01620-7] [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: 09/13/2024] [Accepted: 11/11/2024] [Indexed: 11/25/2024]
Abstract
BACKGROUND The spread of tau pathology closely correlates with the disease course and cognitive decline in Alzheimer's disease (AD). Tau-targeting immunotherapies are being developed to stop the spread of tau pathology and thus halt disease progression. In this post hoc analysis of the ADAMANT clinical trial, we examined the performance of AADvac1, an active immunotherapy targeting the microtubule-binding region (MTBR) of tau, in a subgroup of participants with elevated plasma p-tau217, indicating AD-related neuropathological changes. METHODS ADAMANT was a 24-month, randomized, placebo-controlled, parallel-group, double-blinded, multicenter, phase 2 clinical trial in subjects with mild AD. The trial participants were randomized 3:2 to receive six doses of AADvac1 or placebo at 4-week intervals, followed by five booster doses at 14-week intervals. The primary outcome was safety. The secondary outcomes were the Clinical Dementia Rating-Sum of Boxes (CDR-SB), the Alzheimer's Disease Cooperative Study - Activities of Daily Living score for Mild Cognitive Impairment 18-item version (ADCS-ADL-MCI-18), and immunogenicity. Volumetric MRI, plasma neurofilament light (NfL), and glial fibrillary acidic protein (GFAP) were exploratory outcomes. The inclusion criterion for this post-hoc analysis was a baseline plasma p-tau217 level above the cutoff for AD. RESULTS Among 196 ADAMANT participants, 137 were positive for plasma p-tau217 (mean age 71.4 years, 59% women). AADvac1 was safe and well tolerated in this subgroup. AADvac1 reduced the rate of accumulation of log-plasma NfL by 56% and that of GFAP by 73%. The treatment differences in the CDR-SB and ADCS-ADL-MCI-18 scores favored AADvac1 but were not statistically significant. AADvac1 had no effect on whole-brain volume but nonsignificantly reduced the loss of brain cortical tissue in several regions. Importantly, the impact on the study outcomes was more pronounced in participants with higher anti-tau antibody levels. CONCLUSIONS These results suggest that AADvac1 tau immunotherapy can reduce plasma biomarkers of neurodegeneration and neuroinflammation. These findings and possible observations on brain atrophy and cognition are hypothesis-generating and warrant further evaluation in a larger clinical trial. TRIAL REGISTRATION EudraCT 2015-000630-30 (primary) and NCT02579252.
Collapse
Affiliation(s)
- Branislav Kovacech
- Axon Neuroscience R&D Services SE, Dvorakovo Nabr. 10, 81102, Bratislava, Slovakia.
| | - Nicholas C Cullen
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Clinical Research Centre, Jan Waldenströms Gata 35, 202 13, Malmö, Sweden
| | - Petr Novak
- Axon Neuroscience R&D Services SE, Dvorakovo Nabr. 10, 81102, Bratislava, Slovakia
| | - Jozef Hanes
- Axon Neuroscience R&D Services SE, Dvorakovo Nabr. 10, 81102, Bratislava, Slovakia
| | - Eva Kontsekova
- Axon Neuroscience R&D Services SE, Dvorakovo Nabr. 10, 81102 Bratislava, Slovakia and Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska Cesta 9, Bratislava, 84510, Slovakia
| | - Stanislav Katina
- Department of Mathematics and Statistics, Axon Neuroscience R&D Services SE, Bratislava, Slovakia, and (current) Masaryk University, Kotlářská 267/2, Brno, 611 37, Czech Republic
| | - Vojtech Parrak
- Axon Neuroscience R&D Services SE, Dvorakovo Nabr. 10, 81102, Bratislava, Slovakia
| | - Michal Fresser
- Axon Neuroscience SE, 4 Arch. Makariou & Kalogreon, 6016, Larnaca, Cyprus
| | | | - Howard H Feldman
- Department of Neurosciences, Alzheimer's Disease Cooperative Study, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Bengt Winblad
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, BioClinicum, 171 64, Solna, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, 141 86, Stockholm, Sweden
| | - Erik Stoops
- ADx NeuroSciences NV, Technologiepark 6, 9052, Ghent, Belgium
| | | | - Norbert Zilka
- Axon Neuroscience R&D Services SE, Dvorakovo Nabr. 10, 81102, Bratislava, Slovakia.
| |
Collapse
|
8
|
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
|
9
|
Katzdobler S, Nübling G, Klietz M, Fietzek UM, Palleis C, Bernhardt AM, Wegner F, Huber M, Rogozinski S, Schneider LS, Spruth EJ, Beyle A, Vogt IR, Brandt M, Hansen N, Glanz W, Brockmann K, Spottke A, Hoffmann DC, Peters O, Priller J, Wiltfang J, Düzel E, Schneider A, Falkenburger B, Klockgether T, Gasser T, Nuscher B, Haass C, Höglinger G, Levin J. GFAP and NfL as fluid biomarkers for clinical disease severity and disease progression in multiple system atrophy (MSA). J Neurol 2024; 271:6991-6999. [PMID: 39254698 PMCID: PMC11447157 DOI: 10.1007/s00415-024-12647-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/28/2024] [Accepted: 08/07/2024] [Indexed: 09/11/2024]
Abstract
BACKGROUND Multiple system atrophy (MSA), an atypical parkinsonian syndrome, is a rapidly progressive neurodegenerative disease with currently no established fluid biomarkers available. MSA is characterized by an oligodendroglial α-synucleinopathy, progressive neuronal cell loss and concomitant astrocytosis. Here, we investigate glial fibrillary acidic protein (GFAP) and neurofilament light chain (NfL) as fluid biomarkers for differential diagnosis, assessment of clinical disease severity and prediction of disease progression in MSA. METHODS GFAP and NfL levels were analyzed in plasma and CSF samples of 47 MSA patients as well as 24 Parkinson's disease (PD) and 25 healthy controls (HC) as reference cohorts. In MSA, biomarker levels were correlated to baseline and longitudinal clinical disease severity (UMSARS scores). RESULTS In MSA, GFAP levels in CSF and plasma predicted baseline clinical disease severity as indicated by UMSARS scores, while NfL levels predicted clinical disease progression as indicated by longitudinal changes in UMSARS scores. Cross-sectionally, NfL levels in CSF and plasma were significantly elevated in MSA compared to both PD and HC. Receiver operating curves (ROC) indicated high diagnostic accuracy of NfL for distinguishing MSA from PD (CSF: AUC = 0.97, 95% CI 0.90-1.00; plasma: AUC = 0.90, 95% CI 0.81-1.00). DISCUSSION In MSA, GFAP shows promise as novel biomarker for assessing current clinical disease severity, while NfL might serve as biomarker for prediction of disease progression and differential diagnosis of MSA against PD.
Collapse
Affiliation(s)
- Sabrina Katzdobler
- Department of Neurology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- Munich Cluster for Systems Neurology, SyNergy, Munich, Germany
- German Center for Neurodegenerative Diseases, DZNE, Munich, Germany
- Graduate School of Systemic Neurosciences (GSN), Munich, Germany
| | - Georg Nübling
- Department of Neurology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- German Center for Neurodegenerative Diseases, DZNE, Munich, Germany
| | - Martin Klietz
- Department of Neurology, Hanover Medical School, Hanover, Germany
| | - Urban M Fietzek
- Department of Neurology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- German Center for Neurodegenerative Diseases, DZNE, Munich, Germany
| | - Carla Palleis
- Department of Neurology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- Munich Cluster for Systems Neurology, SyNergy, Munich, Germany
- German Center for Neurodegenerative Diseases, DZNE, Munich, Germany
| | - Alexander M Bernhardt
- Department of Neurology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- German Center for Neurodegenerative Diseases, DZNE, Munich, Germany
- Clinical Mass Spectrometry Center Munich, Munich, Germany
| | - Florian Wegner
- Department of Neurology, Hanover Medical School, Hanover, Germany
| | - Meret Huber
- Department of Neurology, Hanover Medical School, Hanover, Germany
| | | | - Luisa-Sophie Schneider
- Department of psychiatry and neuroscience, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Eike Jakob Spruth
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Berlin, Germany
| | - Aline Beyle
- Department of Neurology, University of Bonn, Bonn, Germany
| | - Ina R Vogt
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Bonn, Germany
| | - Moritz Brandt
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
- Department of Neurology, Technische Universität Dresden, Dresden, Germany
| | - Niels Hansen
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, University of Goettingen, Goettingen, Germany
| | - Wenzel Glanz
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research, Otto-Von-Guericke University, Magdeburg, Germany
- Clinic for Neurology, Medical Faculty, University Hospital Magdeburg, Magdeburg, Germany
| | - Kathrin Brockmann
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department for Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Annika Spottke
- Department of Neurology, University of Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Bonn, Germany
| | - Daniel C Hoffmann
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Bonn, Germany
| | - Oliver Peters
- Department of psychiatry and neuroscience, Charité-Universitätsmedizin Berlin, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| | - Josef Priller
- Department of Psychiatry and Psychotherapy, School of Medicine and Health, Technical University of Munich, Munich, Germany
- Neuropsychiatry Unit and Laboratory of Molecular Psychiatry, Charité, Universitätsmedizin Berlin and DZNE, Berlin, Germany
- Centre for Clinical Brain Sciences, UK Dementia Research Institute at the University of Edinburgh, University of Edinburgh, Edinburgh, UK
| | - Jens Wiltfang
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, University of Goettingen, Goettingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany
- Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Emrah Düzel
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research, Otto-Von-Guericke University, Magdeburg, Germany
- Institute of Cognitive Neuroscience, University College London, London, UK
| | - Anja Schneider
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Bonn, Germany
- Dept. of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, University of Bonn Medical Center, Bonn, Germany
| | - Björn Falkenburger
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
- Department of Neurology, Technische Universität Dresden, Dresden, Germany
| | - Thomas Klockgether
- Department of Neurology, University of Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Bonn, Germany
| | - Thomas Gasser
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department for Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Brigitte Nuscher
- Chair of Metabolic Biochemistry, Faculty of Medicine, Biomedical Center (BMC), LMU Munich, Munich, Germany
| | - Christian Haass
- Munich Cluster for Systems Neurology, SyNergy, Munich, Germany
- German Center for Neurodegenerative Diseases, DZNE, Munich, Germany
- Chair of Metabolic Biochemistry, Faculty of Medicine, Biomedical Center (BMC), LMU Munich, Munich, Germany
| | - Günter Höglinger
- Department of Neurology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.
- Munich Cluster for Systems Neurology, SyNergy, Munich, Germany.
- German Center for Neurodegenerative Diseases, DZNE, Munich, Germany.
| | - Johannes Levin
- Department of Neurology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.
- Munich Cluster for Systems Neurology, SyNergy, Munich, Germany.
- German Center for Neurodegenerative Diseases, DZNE, Munich, Germany.
- Graduate School of Systemic Neurosciences (GSN), Munich, Germany.
- Clinical Mass Spectrometry Center Munich, Munich, Germany.
| |
Collapse
|
10
|
Johansson C, Thordardottir S, Laffita-Mesa J, Pannee J, Rodriguez-Vieitez E, Zetterberg H, Blennow K, Graff C. Gene-variant specific effects of plasma amyloid-β levels in Swedish autosomal dominant Alzheimer disease. Alzheimers Res Ther 2024; 16:207. [PMID: 39322953 PMCID: PMC11423518 DOI: 10.1186/s13195-024-01574-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: 05/24/2024] [Accepted: 09/11/2024] [Indexed: 09/27/2024]
Abstract
BACKGROUND Several blood-based biomarkers offer the opportunity of in vivo detection of brain pathology and neurodegeneration in Alzheimer disease with high specificity and sensitivity, but the performance of amyloid-β (Aβ) measurements remains under evaluation. Autosomal dominant Alzheimer disease (ADAD) with mutations in PSEN1, PSEN2 and APP can be studied as a model for sporadic Alzheimer disease. However, clarifying the genetic effects on the Aβ-levels in different matrices such as cerebrospinal fluid or plasma is crucial for generalizability and utility of data. We aimed to explore plasma Aβ concentrations over the Alzheimer disease continuum in a longitudinal cohort of genetic Alzheimer disease. METHODS 92 plasma samples were collected from at-risk individuals (n = 47) in a Swedish cohort of ADAD, including 18 mutation carriers (13 APPswe (p.KM670/671NL) MC), 5 PSEN1 (p.H163Y) MC) and 29 non-carriers (NC) as the reference group. Concentrations of Aβ1-38, Aβ1-40 and Aβ1-42 were analyzed in plasma using immunoprecipitation coupled to tandem liquid chromatography mass spectrometry (IP-LC-MS/MS). Cross-sectional and repeated-measures data analyses were investigated family-wise, applying non-parametric tests as well as mixed-effects models. RESULTS Cross-sectional analysis at baseline showed more than a 3-fold increase in all plasma Aβ peptides in APPswe MC, regardless of clinical status, compared to controls (p < 0.01). PSEN1 (p.H163Y) presymptomatic MC had a decrease of plasma Aβ1-38 compared to controls (p < 0.05). There was no difference in Aβ1-42/1-40 ratio between APPswe MC (PMC and SMC), PSEN1 MC (PMC) and controls at baseline. Notably, both cross-sectional data and repeated-measures analysis suggested that APPswe MC have a stable Aβ1-42/1-40 ratio with increasing age, in contrast to the decrease seen with aging in both controls and PSEN1 (p.H163Y) MC. CONCLUSION These data show very strong mutation-specific effects on Aβ profiles in blood, most likely due to a ubiquitous production outside of the CNS. Hence, analyses in an unselected clinical setting might unintentionally disclose genetic status. Furthermore, our findings suggest that the Aβ ratio might be a poor indicator of brain Aβ pathology in selected genetic cases. The very small sample size is a limitation that needs to be considered but reflects the scarcity of longitudinal in vivo data from genetic cohorts.
Collapse
Affiliation(s)
- Charlotte Johansson
- Department NVS, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Visionsgatan 4, Bioclinicum, Solna, J10:20, 171 64, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
| | - Steinunn Thordardottir
- Department NVS, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Visionsgatan 4, Bioclinicum, Solna, J10:20, 171 64, Sweden
| | - José Laffita-Mesa
- Department NVS, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Visionsgatan 4, Bioclinicum, Solna, J10:20, 171 64, Sweden
| | - Josef Pannee
- 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
| | - Elena Rodriguez-Vieitez
- Department NVS, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Visionsgatan 4, Bioclinicum, Solna, J10:20, 171 64, 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, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - 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
- Pitié-Salpêtrière Hospital, Paris Brain Institute, ICM, Sorbonne University, Paris, France
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, P.R. China
| | - Caroline Graff
- Department NVS, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Visionsgatan 4, Bioclinicum, Solna, J10:20, 171 64, Sweden.
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
11
|
Barsoum S, Latimer CS, Nolan AL, Barrett A, Chang K, Troncoso J, Keene CD, Benjamini D. Resiliency to Alzheimer's disease neuropathology can be distinguished from dementia using cortical astrogliosis imaging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.06.592719. [PMID: 38766087 PMCID: PMC11100587 DOI: 10.1101/2024.05.06.592719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Despite the presence of significant Alzheimer's disease (AD) pathology, characterized by amyloid β (Aβ) plaques and phosphorylated tau (pTau) tangles, some cognitively normal elderly individuals do not inevitably develop dementia. These findings give rise to the notion of cognitive 'resilience', suggesting maintained cognitive function despite the presence of AD neuropathology, highlighting the influence of factors beyond classical pathology. Cortical astroglial inflammation, a ubiquitous feature of symptomatic AD, shows a strong correlation with cognitive impairment severity, potentially contributing to the diversity of clinical presentations. However, noninvasively imaging neuroinflammation, particularly astrogliosis, using MRI remains a significant challenge. Here we sought to address this challenge and to leverage multidimensional (MD) MRI, a powerful approach that combines relaxation with diffusion MR contrasts, to map cortical astrogliosis in the human brain by accessing sub-voxel information. Our goal was to test whether MD-MRI can map astroglial pathology in the cerebral cortex, and if so, whether it can distinguish cognitive resiliency from dementia in the presence of hallmark AD neuropathological changes. We adopted a multimodal approach by integrating histological and MRI analyses using human postmortem brain samples. Ex vivo cerebral cortical tissue specimens derived from three groups comprised of non-demented individuals with significant AD pathology postmortem, individuals with both AD pathology and dementia, and non-demented individuals with minimal AD pathology postmortem as controls, underwent MRI at 7 T. We acquired and processed MD-MRI, diffusion tensor, and quantitative T 1 and T 2 MRI data, followed by histopathological processing on slices from the same tissue. By carefully co-registering MRI and microscopy data, we performed quantitative multimodal analyses, leveraging targeted immunostaining to assess MD-MRI sensitivity and specificity towards Aβ, pTau, and glial fibrillary acidic protein (GFAP), a marker for astrogliosis. Our findings reveal a distinct MD-MRI signature of cortical astrogliosis, enabling the creation of predictive maps for cognitive resilience amid AD neuropathological changes. Multiple linear regression linked histological values to MRI changes, revealing that the MD-MRI cortical astrogliosis biomarker was significantly associated with GFAP burden (standardized β=0.658, pFDR<0.0001), but not with Aβ (standardized β=0.009, p FDR =0.913) or pTau (standardized β=-0.196, p FDR =0.051). Conversely, none of the conventional MRI parameters showed significant associations with GFAP burden in the cortex. While the extent to which pathological glial activation contributes to neuronal damage and cognitive impairment in AD is uncertain, developing a noninvasive imaging method to see its affects holds promise from a mechanistic perspective and as a potential predictor of cognitive outcomes.
Collapse
|
12
|
Limberger C, Zimmer ER. Blood GFAP reflects astrocyte reactivity to Alzheimer's pathology in post-mortem brain tissue. Brain 2024; 147:1598-1600. [PMID: 38634672 DOI: 10.1093/brain/awae104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 03/26/2024] [Indexed: 04/19/2024] Open
Abstract
This scientific commentary refers to ‘Serum GFAP levels correlate with post-mortem brain atrophy and neurofibrillary tangles’ by Sánchez-Juan et al. (https://doi.org/10.1093/brain/awae035).
Collapse
Affiliation(s)
- Christian Limberger
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - 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 Pharmacology, UFRGS, Porto Alegre, Brazil
- McGill Centre for Studies in Aging, McGill University, Montreal, Canada
- Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
13
|
Sánchez-Juan P, Valeriano-Lorenzo E, Ruiz-González A, Pastor AB, Rodrigo Lara H, López-González F, Zea-Sevilla MA, Valentí M, Frades B, Ruiz P, Saiz L, Burgueño-García I, Calero M, del Ser T, Rábano A. Serum GFAP levels correlate with astrocyte reactivity, post-mortem brain atrophy and neurofibrillary tangles. Brain 2024; 147:1667-1679. [PMID: 38634687 PMCID: PMC11068326 DOI: 10.1093/brain/awae035] [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: 07/10/2023] [Revised: 12/28/2023] [Accepted: 01/21/2024] [Indexed: 04/19/2024] Open
Abstract
Glial fibrillary acidic protein (GFAP), a proxy of astrocyte reactivity, has been proposed as biomarker of Alzheimer's disease. However, there is limited information about the correlation between blood biomarkers and post-mortem neuropathology. In a single-centre prospective clinicopathological cohort of 139 dementia patients, for which the time-frame between GFAP level determination and neuropathological assessment was exceptionally short (on average 139 days), we analysed this biomarker, measured at three time points, in relation to proxies of disease progression such as cognitive decline and brain weight. Most importantly, we investigated the use of blood GFAP to detect the neuropathological hallmarks of Alzheimer's disease, while accounting for potential influences of the most frequent brain co-pathologies. The main findings demonstrated an association between serum GFAP level and post-mortem tau pathology (β = 12.85; P < 0.001) that was independent of amyloid deposits (β = 13.23; P = 0.02). A mediation analysis provided additional support for the role of astrocytic activation as a link between amyloid and tau pathology in Alzheimer's disease. Furthermore, a negative correlation was observed between pre-mortem serum GFAP and brain weight at post-mortem (r = -0.35; P < 0.001). This finding, together with evidence of a negative correlation with cognitive assessments (r = -0.27; P = 0.005), supports the role of GFAP as a biomarker for disease monitoring, even in the late phases of Alzheimer's disease. Moreover, the diagnostic performance of GFAP in advanced dementia patients was explored, and its discriminative power (area under the receiver operator characteristic curve at baseline = 0.91) in differentiating neuropathologically-confirmed Alzheimer's disease dementias from non-Alzheimer's disease dementias was determined, despite the challenging scenario of advanced age and frequent co-pathologies in these patients. Independently of Alzheimer's disease, serum GFAP levels were shown to be associated with two other pathologies targeting the temporal lobes-hippocampal sclerosis (β = 3.64; P = 0.03) and argyrophilic grain disease (β = -6.11; P = 0.02). Finally, serum GFAP levels were revealed to be correlated with astrocyte reactivity, using the brain GFAP-immunostained area as a proxy (ρ = 0.21; P = 0.02). Our results contribute to increasing evidence suggesting a role for blood GFAP as an Alzheimer's disease biomarker, and the findings offer mechanistic insights into the relationship between blood GFAP and Alzheimer's disease neuropathology, highlighting its ties with tau burden. Moreover, the data highlighting an independent association between serum GFAP levels and other neuropathological lesions provide information for clinicians to consider when interpreting test results. The longitudinal design and correlation with post-mortem data reinforce the robustness of our findings. However, studies correlating blood biomarkers and neuropathological assessments are still scant, and further research is needed to replicate and validate these results in diverse populations.
Collapse
Affiliation(s)
- Pascual Sánchez-Juan
- Alzheimer’s Centre Reina Sofia-CIEN Foundation-ISCIII, Research Platforms, 28031 Madrid, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28029 Madrid, Spain
| | | | - Alicia Ruiz-González
- Alzheimer’s Centre Reina Sofia-CIEN Foundation-ISCIII, Research Platforms, 28031 Madrid, Spain
| | - Ana Belén Pastor
- Alzheimer’s Centre Reina Sofia-CIEN Foundation-ISCIII, Research Platforms, 28031 Madrid, Spain
| | - Hector Rodrigo Lara
- Banco de Cerebros de la Región de Murcia, Neuropathology Department, 30120 Murcia, Spain
| | | | | | - Meritxell Valentí
- Alzheimer’s Centre Reina Sofia-CIEN Foundation-ISCIII, Research Platforms, 28031 Madrid, Spain
| | - Belen Frades
- Alzheimer’s Centre Reina Sofia-CIEN Foundation-ISCIII, Research Platforms, 28031 Madrid, Spain
| | - Paloma Ruiz
- Alzheimer’s Centre Reina Sofia-CIEN Foundation-ISCIII, Research Platforms, 28031 Madrid, Spain
| | - Laura Saiz
- Alzheimer’s Centre Reina Sofia-CIEN Foundation-ISCIII, Research Platforms, 28031 Madrid, Spain
| | - Iván Burgueño-García
- Alzheimer’s Centre Reina Sofia-CIEN Foundation-ISCIII, Research Platforms, 28031 Madrid, Spain
| | - Miguel Calero
- Alzheimer’s Centre Reina Sofia-CIEN Foundation-ISCIII, Research Platforms, 28031 Madrid, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28029 Madrid, Spain
- Chronic Disease Programme, Instituto de Salud Carlos III, Madrid, Spain
| | - Teodoro del Ser
- Alzheimer’s Centre Reina Sofia-CIEN Foundation-ISCIII, Research Platforms, 28031 Madrid, Spain
| | - Alberto Rábano
- Alzheimer’s Centre Reina Sofia-CIEN Foundation-ISCIII, Research Platforms, 28031 Madrid, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28029 Madrid, Spain
| |
Collapse
|
14
|
Nordberg A. Insights into the progression of genetic Alzheimer's disease from tau PET. Lancet Neurol 2024; 23:453-454. [PMID: 38631755 DOI: 10.1016/s1474-4422(24)00124-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 03/18/2024] [Indexed: 04/19/2024]
Affiliation(s)
- Agneta Nordberg
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Theme Inflammation and Aging, Karolinska University Hospital, Stockholm 14183, Sweden.
| |
Collapse
|
15
|
Jaisa-Aad M, Muñoz-Castro C, Healey MA, Hyman BT, Serrano-Pozo A. Characterization of monoamine oxidase-B (MAO-B) as a biomarker of reactive astrogliosis in Alzheimer's disease and related dementias. Acta Neuropathol 2024; 147:66. [PMID: 38568475 PMCID: PMC10991006 DOI: 10.1007/s00401-024-02712-2] [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: 08/20/2023] [Revised: 02/03/2024] [Accepted: 02/21/2024] [Indexed: 04/05/2024]
Abstract
Reactive astrogliosis accompanies the two neuropathological hallmarks of Alzheimer's disease (AD)-Aβ plaques and neurofibrillary tangles-and parallels neurodegeneration in AD and AD-related dementias (ADRD). Thus, there is growing interest in developing imaging and fluid biomarkers of reactive astrogliosis for AD/ADRD diagnosis and prognostication. Monoamine oxidase-B (MAO-B) is emerging as a target for PET imaging radiotracers of reactive astrogliosis. However, a thorough characterization of MAO-B expression in postmortem control and AD/ADRD brains is lacking. We sought to: (1) identify the primary cell type(s) expressing MAO-B in control and AD brains; (2) quantify MAO-B immunoreactivity in multiple brain regions of control and AD donors as a proxy for PET radiotracer uptake; (3) correlate MAO-B level with local AD neuropathological changes, reactive glia, and cortical atrophy; (4) determine whether the MAOB rs1799836 SNP genotype impacts MAO-B expression level; (5) compare MAO-B immunoreactivity across AD/ADRD, including Lewy body diseases (LBD) and frontotemporal lobar degenerations with tau (FTLD-Tau) and TDP-43 (FTLD-TDP). We found that MAO-B is mainly expressed by subpial and perivascular cortical astrocytes as well as by fibrous white matter astrocytes in control brains, whereas in AD brains, MAO-B is significantly upregulated by both cortical reactive astrocytes and white matter astrocytes across temporal, frontal, and occipital lobes. By contrast, MAO-B expression level was unchanged and lowest in cerebellum. Cortical MAO-B expression was independently associated with cortical atrophy and local measures of reactive astrocytes and microglia, and significantly increased in reactive astrocytes surrounding Thioflavin-S+ dense-core Aβ plaques. MAO-B expression was not affected by the MAOB rs1799836 SNP genotype. MAO-B expression was also significantly increased in the frontal cortex and white matter of donors with corticobasal degeneration, Pick's disease, and FTLD-TDP, but not in LBD or progressive supranuclear palsy. These findings support ongoing efforts to develop MAO-B-based PET radiotracers to image reactive astrogliosis in AD/ADRD.
Collapse
Affiliation(s)
- Methasit Jaisa-Aad
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th St., Charlestown, MA, 02129, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Clara Muñoz-Castro
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th St., Charlestown, MA, 02129, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Molly A Healey
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th St., Charlestown, MA, 02129, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th St., Charlestown, MA, 02129, USA
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, 02129, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Alberto Serrano-Pozo
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA.
- MassGeneral Institute for Neurodegenerative Disease, 114 16th St., Charlestown, MA, 02129, USA.
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, 02129, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
16
|
Chong JR, Chai YL, Yam ATY, Hilal S, Vrooman H, Venketasubramanian N, Blennow K, Zetterberg H, Ashton NJ, Chen CP, Lai MKP. Association of plasma GFAP with elevated brain amyloid is dependent on severity of white matter lesions in an Asian cognitively impaired cohort. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12576. [PMID: 38605996 PMCID: PMC11007806 DOI: 10.1002/dad2.12576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/25/2024] [Accepted: 03/01/2024] [Indexed: 04/13/2024]
Abstract
INTRODUCTION While elevated blood glial fibrillary acidic protein (GFAP) has been associated with brain amyloid pathology, whether this association occurs in populations with high cerebral small vessel disease (CSVD) concomitance remains unclear. METHODS Using a Singapore-based cohort of cognitively impaired subjects, we assessed associations between plasma GFAP and neuroimaging measures of brain amyloid and CSVD, including white matter hyperintensities (WMH). We also examined the diagnostic performance of plasma GFAP in detecting brain amyloid beta positivity (Aβ+). RESULTS When stratified by WMH status, elevated brain amyloid was associated with higher plasma GFAP only in the WMH- group (β = 0.383; P < 0.001). The diagnostic performance of plasma GFAP in identifying Aβ+ was significantly higher in the WMH- group (area under the curve [AUC] = 0.896) than in the WMH+ group (AUC = 0.712, P = 0.008). DISCUSSION The biomarker utility of plasma GFAP in detecting brain amyloid pathology is dependent on the severity of concomitant WMH. Highlight Glial fibrillary acidic protein (GFAP)'s association with brain amyloid is unclear in populations with high cerebral small vessel disease (CSVD).Plasma GFAP was measured in a cohort with CSVD and brain amyloid.Plasma GFAP was better in detecting amyloid in patients with low CSVD versus high CSVD.Biomarker utility of GFAP in detecting brain amyloid depends on the severity of CSVD.
Collapse
Affiliation(s)
- Joyce R. Chong
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
- Memory, Aging and Cognition CentreNational University Health SystemsKent RidgeSingapore
| | - Yuek Ling Chai
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
- Memory, Aging and Cognition CentreNational University Health SystemsKent RidgeSingapore
| | - Amelia T. Y. Yam
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
- Memory, Aging and Cognition CentreNational University Health SystemsKent RidgeSingapore
| | - Saima Hilal
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
- Memory, Aging and Cognition CentreNational University Health SystemsKent RidgeSingapore
- Saw Swee Hock School of Public HealthNational University of Singapore and National University Health SystemKent RidgeSingapore
- Department of Radiology and Nuclear MedicineErasmus Medical CenterRotterdamthe Netherlands
| | - Henri Vrooman
- Department of Radiology and Nuclear MedicineErasmus Medical CenterRotterdamthe Netherlands
| | | | - Kaj Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGöteborgSweden
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGöteborgSweden
- Department of Neurodegenerative DiseaseThe UCL Queen Square Institute of NeurologyLondonUK
| | - Nicholas J. Ashton
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGöteborgSweden
| | - Christopher P. Chen
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
- Memory, Aging and Cognition CentreNational University Health SystemsKent RidgeSingapore
| | - Mitchell K. P. Lai
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
- Memory, Aging and Cognition CentreNational University Health SystemsKent RidgeSingapore
| |
Collapse
|
17
|
Varma VR, An Y, Kac PR, Bilgel M, Moghekar A, Loeffler T, Amschl D, Troncoso J, Blennow K, Zetterberg H, Ashton NJ, Resnick SM, Thambisetty M. Longitudinal progression of blood biomarkers reveals a key role of astrocyte reactivity in preclinical Alzheimer's disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.25.24301779. [PMID: 38343809 PMCID: PMC10854357 DOI: 10.1101/2024.01.25.24301779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/26/2024]
Abstract
Defining the progression of blood biomarkers of Alzheimer's disease (AD) is essential for targeting treatments in patients most likely to benefit from early intervention. We delineated the temporal ordering of blood biomarkers a decade prior to the onset of AD symptoms in participants in the Baltimore Longitudinal Study of Aging. We show that increased astrocyte reactivity, assessed by elevated glial fibrillary acidic protein (GFAP) levels is an early event in the progression of blood biomarker changes in preclinical AD. In AD-converters who are initially cognitively unimpaired (N=158, 377 serial plasma samples), higher plasma GFAP levels are observed as early as 10-years prior to the onset of cognitive impairment due to incident AD compared to individuals who remain cognitively unimpaired (CU, N=160, 379 serial plasma samples). Plasma GFAP levels in AD-converters remain elevated 5-years prior to and coincident with the onset of cognitive impairment due to AD. In participants with neuropathologically confirmed AD, plasma GFAP levels are elevated relative to cognitively normal individuals and intermediate in those who remain cognitively unimpaired despite significant AD pathology (asymptomatic AD). Higher plasma GFAP levels at death are associated with greater severity of both neuritic plaques and neurofibrillary tangles. In the 5XFAD transgenic model of AD, we observed greater GFAP levels in the cortex and hippocampus of transgenic mice relative to wild-type prior to the development of cognitive impairment. Reactive astrocytosis, an established biological response to neuronal injury, may be an early initiator of AD pathogenesis and a promising therapeutic target.
Collapse
Affiliation(s)
- V R Varma
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, United States of America
| | - Y An
- Brain Aging and Behavior Section, Laboratory of Behavioral Neuroscience, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, MD, USA
| | - P R Kac
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - M Bilgel
- Brain Aging and Behavior Section, Laboratory of Behavioral Neuroscience, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, MD, USA
| | - A Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | - T Loeffler
- Scantox Neuro GmbH, Parkring 12, 8074, Grambach, Austria
| | - D Amschl
- Scantox Neuro GmbH, Parkring 12, 8074, Grambach, Austria
| | - J Troncoso
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - K 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
| | - H 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, 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
| | - N J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- 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
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - S M Resnick
- Brain Aging and Behavior Section, Laboratory of Behavioral Neuroscience, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, MD, USA
| | - M Thambisetty
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, United States of America
| |
Collapse
|
18
|
Rodriguez-Vieitez E, Kumar A, Malarte ML, Ioannou K, Rocha FM, Chiotis K. Imaging Neuroinflammation: Quantification of Astrocytosis in a Multitracer PET Approach. Methods Mol Biol 2024; 2785:195-218. [PMID: 38427196 DOI: 10.1007/978-1-0716-3774-6_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
The recent progress in the development of in vivo biomarkers is rapidly changing how neurodegenerative diseases are conceptualized and diagnosed and how clinical trials are designed today. Alzheimer's disease (AD) - the most common neurodegenerative disorder - is characterized by a complex neuropathology involving the deposition of extracellular amyloid-β (Aβ) plaques and intracellular neurofibrillary tangles (NFTs) of hyperphosphorylated tau proteins, accompanied by the activation of glial cells, i.e., astrocytes and microglia, and neuroinflammatory response, leading to neurodegeneration and cognitive dysfunction. An increasing diversity of positron emission tomography (PET) imaging radiotracers is available to selectively target the different pathophysiological processes of AD. Along with the success of Aβ PET and the more recent tau PET imaging, there is a great interest to develop PET tracers to image glial reactivity and neuroinflammation. While most research to date has focused on imaging microgliosis, there is an upsurge of interest in imaging reactive astrocytes in the AD continuum. There is increasing evidence that reactive astrocytes are morphologically and functionally heterogeneous, with different subtypes that express different markers and display various homeostatic or detrimental roles across disease stages. Therefore, multiple biomarkers are desirable to unravel the complex phenomenon of reactive astrocytosis. In the field of in vivo PET imaging in AD, the research concerning reactive astrocytes has predominantly focused on targeting monoamine oxidase B (MAO-B), most often using either 11C-deuterium-L-deprenyl (11C-DED) or 18F-SMBT-1 PET tracers. Additionally, imidazoline2 binding (I2BS) sites have been imaged using 11C-BU99008 PET. Recent studies in our group using 11C-DED PET imaging suggest that astrocytosis may be present from the early stages of disease development in AD. This chapter provides a detailed description of the practical approach used for the analysis of 11C-DED PET imaging data in a multitracer PET paradigm including 11C-Pittsburgh compound B (11C-PiB) and 18F-fluorodeoxyglucose (18F-FDG). The multitracer PET approach allows investigating the comparative regional and temporal patterns of in vivo brain astrocytosis, fibrillar Aβ deposition, glucose metabolism, and brain structural changes. It may also contribute to understanding the potential role of novel plasma biomarkers of reactive astrocytes, in particular the glial fibrillary acidic protein (GFAP), at different stages of disease progression. This chapter attempts to stimulate further research in the field, including the development of novel PET tracers that may allow visualizing different aspects of the complex astrocytic and microglial response in neurodegenerative diseases. Progress in the field will contribute to the incorporation of PET imaging of glial reactivity and neuroinflammation as biomarkers with clinical application and motivate further investigation on glial cells as therapeutic targets in AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Elena Rodriguez-Vieitez
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
| | - Amit Kumar
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Mona-Lisa Malarte
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Konstantinos Ioannou
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Filipa M Rocha
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Konstantinos Chiotis
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
19
|
Abstract
This paper provides an overview of the role of neuroinflammation in Alzheimer's disease and other neurodegenerative diseases, highlighting the potential of anti-inflammatory treatments to slow or prevent decline. This research focuses on the use of positron emission tomography (PET) imaging to visualize and quantify molecular brain changes in patients, specifically microglial activation and reactive astrogliosis. We discuss the development and application of several PET radioligands, including first-generation ligands like PK11195 and Ro5-4864, as well as second- and third-generation ligands such as [11C]PBR28, [18F]DPA-714, [18F]GE-180, and [11C]ER176. These ligands target the 18-kDa translocator protein (TSPO), which is overexpressed in activated microglia and upregulated in astrocytes. We also address the limitations of these ligands, such as low brain uptake, poor penetration of the blood-brain barrier, short half-life, and variable kinetic behavior. Furthermore, we demonstrate the impact of genetic polymorphisms on ligand binding.
Collapse
Affiliation(s)
- Maura Malpetti
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal and Gothenburg, Sweden
| | - Matthias Brendel
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
| |
Collapse
|