1
|
Wang G, Han Y, Peng K, Jiang Z, Wang T, Zheng Q, Li W, Xu H, Ji F, Li Z. Astragalin-functionalized ultrasmall nanoparticles modulate the complement pathway to inhibit microglial synaptic phagocytosis for reducing anesthetic neurotoxicity. Mater Today Bio 2025; 32:101714. [PMID: 40230643 PMCID: PMC11994406 DOI: 10.1016/j.mtbio.2025.101714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 03/10/2025] [Accepted: 03/28/2025] [Indexed: 04/16/2025] Open
Abstract
Synaptic impairment is identified as a primary pathology in sevoflurane-induced neurotoxicity, contributing to neurobehavioral and neurodevelopmental deficits. Synaptic loss in neurons occurs through microglia-mediated synaptic phagocytosis via the complement pathway. Astragalin, a natural flavonoid compound, exhibits diverse bioactivities, such as anti-tumor, anti-complement, and anti-inflammatory effects. Herein, astragalin-functionalized Cu 2-x Se nanoparticles (CSPA NPs) can effectively inhibit the complement pathway, mitigating microglia-mediated synaptic phagocytosis and promoting synaptic restoration to repair sevoflurane-induced neurotoxicity. They efficiently target and reduce microglial activation and phagocytosis. By downregulating sortilin, CSPA NPs increase progranulin expression, promoting TFEB cytoplasmic translocation to decrease lysosomal activity and microglial phagocytosis. Furthermore, CSPA NPs decrease complement C1q and C3 levels, inhibiting microglial synaptic engulfment and ameliorating cognition dysfunction in sevoflurane-treated mice. This study illustrates that CSPA NPs inhibit microglial synaptic elimination via the complement pathway, alleviating sevoflurane-induced neurotoxicity and providing insights into treating complement pathway-related diseases.
Collapse
Affiliation(s)
- Gang Wang
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, Jiangsu, China
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College, Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Yaobao Han
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College, Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Ke Peng
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, Jiangsu, China
| | - Zhilin Jiang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College, Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Tingting Wang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College, Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Qing Zheng
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College, Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Wenting Li
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, Jiangsu, China
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College, Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Hanbing Xu
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, Jiangsu, China
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College, Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Fuhai Ji
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, Jiangsu, China
| | - Zhen Li
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College, Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| |
Collapse
|
2
|
Niedowicz DM, Wang W, Prajapati P, Zhong Y, Fister S, Rogers CB, Sompol P, Powell DK, Patel I, Norris CM, Saatman KE, Nelson PT. Nicorandil treatment improves survival and spatial learning in aged granulin knockout mice. Brain Pathol 2025; 35:e13312. [PMID: 39438022 PMCID: PMC11961209 DOI: 10.1111/bpa.13312] [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: 05/29/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024] Open
Abstract
Mutations in the human granulin (GRN) gene are associated with multiple diseases, including dementia disorders such as frontotemporal dementia (FTD) and limbic-predominant age-related TDP-43 encephalopathy (LATE). We studied a Grn knockout (Grn-KO) mouse model in order to evaluate a potential therapeutic strategy for these diseases using nicorandil, a commercially available agonist for the ABCC9/Abcc9-encoded regulatory subunit of the "K+ATP" channel that is well-tolerated in humans. Aged (13 months) Grn-KO and wild-type (WT) mice were treated as controls or with nicorandil (15 mg/kg/day) in drinking water for 7 months, then tested for neurobehavioral performance, neuropathology, and gene expression. Mortality was significantly higher for aged Grn-KO mice (particularly females), but there was a conspicuous improvement in survival for both sexes treated with nicorandil. Grn-KO mice performed worse on some cognitive tests than WT mice, but Morris Water Maze performance was improved with nicorandil treatment. Neuropathologically, Grn-KO mice had significantly increased levels of glial fibrillary acidic protein (GFAP)-immunoreactive astrocytosis but not ionized calcium binding adaptor molecule 1 (IBA-1)-immunoreactive microgliosis, indicating cell-specific inflammation in the brain. Expression of several astrocyte-enriched genes, including Gfap, were also elevated in the Grn-KO brain. Nicorandil treatment was associated with a subtle shift in a subset of detected brain transcript levels, mostly related to attenuated inflammatory markers. Nicorandil treatment improved survival outcomes, cognition, and inflammation in aged Grn-KO mice.
Collapse
Affiliation(s)
- Dana M. Niedowicz
- Sanders Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
| | - Wang‐Xia Wang
- Sanders Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
- Department of PathologyUniversity of KentuckyLexingtonKentuckyUSA
| | - Paresh Prajapati
- Sanders Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
| | - Yu Zhong
- Sanders Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
| | - Shuling Fister
- Sanders Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
| | - Colin B. Rogers
- Sanders Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
| | - Pradoldej Sompol
- Sanders Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
- Department of Pharmacology and Nutritional SciencesUniversity of KentuckyLexingtonKentuckyUSA
| | - David K. Powell
- Department of NeuroscienceUniversity of KentuckyLexingtonKentuckyUSA
| | - Indumati Patel
- Sanders Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
| | - Christopher M. Norris
- Sanders Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
- Department of Pharmacology and Nutritional SciencesUniversity of KentuckyLexingtonKentuckyUSA
| | | | - Peter T. Nelson
- Sanders Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
- Department of PathologyUniversity of KentuckyLexingtonKentuckyUSA
| |
Collapse
|
3
|
Pottier C, Küçükali F, Baker M, Batzler A, Jenkins GD, van Blitterswijk M, Vicente CT, De Coster W, Wynants S, Van de Walle P, Ross OA, Murray ME, Faura J, Haggarty SJ, van Rooij JG, Mol MO, Hsiung GYR, Graff C, Öijerstedt L, Neumann M, Asmann Y, McDonnell SK, Baheti S, Josephs KA, Whitwell JL, Bieniek KF, Forsberg L, Heuer H, Lago AL, Geier EG, Yokoyama JS, Oddi AP, Flanagan M, Mao Q, Hodges JR, Kwok JB, Domoto-Reilly K, Synofzik M, Wilke C, Onyike C, Dickerson BC, Evers BM, Dugger BN, Munoz DG, Keith J, Zinman L, Rogaeva E, Suh E, Gefen T, Geula C, Weintraub S, Diehl-Schmid J, Farlow MR, Edbauer D, Woodruff BK, Caselli RJ, Donker Kaat LL, Huey ED, Reiman EM, Mead S, King A, Roeber S, Nana AL, Ertekin-Taner N, Knopman DS, Petersen RC, Petrucelli L, Uitti RJ, Wszolek ZK, Ramos EM, Grinberg LT, Tempini MLG, Rosen HJ, Spina S, Piguet O, Grossman M, Trojanowski JQ, Keene CD, Jin LW, Prudlo J, Geschwind DH, Rissman RA, Cruchaga C, Ghetti B, Halliday GM, Beach TG, Serrano GE, Arzberger T, Herms J, Boxer AL, Honig LS, Vonsattel JP, Lopez OL, Kofler J, White CL, Gearing M, Glass J, Rohrer JD, Irwin DJ, Lee EB, et alPottier C, Küçükali F, Baker M, Batzler A, Jenkins GD, van Blitterswijk M, Vicente CT, De Coster W, Wynants S, Van de Walle P, Ross OA, Murray ME, Faura J, Haggarty SJ, van Rooij JG, Mol MO, Hsiung GYR, Graff C, Öijerstedt L, Neumann M, Asmann Y, McDonnell SK, Baheti S, Josephs KA, Whitwell JL, Bieniek KF, Forsberg L, Heuer H, Lago AL, Geier EG, Yokoyama JS, Oddi AP, Flanagan M, Mao Q, Hodges JR, Kwok JB, Domoto-Reilly K, Synofzik M, Wilke C, Onyike C, Dickerson BC, Evers BM, Dugger BN, Munoz DG, Keith J, Zinman L, Rogaeva E, Suh E, Gefen T, Geula C, Weintraub S, Diehl-Schmid J, Farlow MR, Edbauer D, Woodruff BK, Caselli RJ, Donker Kaat LL, Huey ED, Reiman EM, Mead S, King A, Roeber S, Nana AL, Ertekin-Taner N, Knopman DS, Petersen RC, Petrucelli L, Uitti RJ, Wszolek ZK, Ramos EM, Grinberg LT, Tempini MLG, Rosen HJ, Spina S, Piguet O, Grossman M, Trojanowski JQ, Keene CD, Jin LW, Prudlo J, Geschwind DH, Rissman RA, Cruchaga C, Ghetti B, Halliday GM, Beach TG, Serrano GE, Arzberger T, Herms J, Boxer AL, Honig LS, Vonsattel JP, Lopez OL, Kofler J, White CL, Gearing M, Glass J, Rohrer JD, Irwin DJ, Lee EB, Van Deerlin V, Castellani R, Mesulam MM, Tartaglia MC, Finger EC, Troakes C, Al-Sarraj S, Dalgard CL, Miller BL, Seelaar H, Graff-Radford NR, Boeve BF, Mackenzie IR, van Swieten JC, Seeley WW, Sleegers K, Dickson DW, Biernacka JM, Rademakers R. Deciphering distinct genetic risk factors for FTLD-TDP pathological subtypes via whole-genome sequencing. Nat Commun 2025; 16:3914. [PMID: 40280976 PMCID: PMC12032271 DOI: 10.1038/s41467-025-59216-0] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 04/15/2025] [Indexed: 04/29/2025] Open
Abstract
Frontotemporal lobar degeneration with neuronal inclusions of the TAR DNA-binding protein 43 (FTLD-TDP) is a fatal neurodegenerative disorder with only a limited number of risk loci identified. We report our comprehensive genome-wide association study as part of the International FTLD-TDP Whole-Genome Sequencing Consortium, including 985 patients and 3,153 controls compiled from 26 institutions/brain banks in North America, Europe and Australia, and meta-analysis with the Dementia-seq cohort. We confirm UNC13A as the strongest overall FTLD-TDP risk factor and identify TNIP1 as a novel FTLD-TDP risk factor. In subgroup analyzes, we further identify genome-wide significant loci specific to each of the three main FTLD-TDP pathological subtypes (A, B and C), as well as enrichment of risk loci in distinct tissues, brain regions, and neuronal subtypes, suggesting distinct disease aetiologies in each of the subtypes. Rare variant analysis confirmed TBK1 and identified C3AR1, SMG8, VIPR1, RBPJL, L3MBTL1 and ANO9, as novel subtype-specific FTLD-TDP risk genes, further highlighting the role of innate and adaptive immunity and notch signaling pathway in FTLD-TDP, with potential diagnostic and novel therapeutic implications.
Collapse
Affiliation(s)
- Cyril Pottier
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium.
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA.
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, MO, USA.
| | - Fahri Küçükali
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
| | - Matt Baker
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Anthony Batzler
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Gregory D Jenkins
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | - Cristina T Vicente
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
| | - Wouter De Coster
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
| | - Sarah Wynants
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
| | - Pieter Van de Walle
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Júlia Faura
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
| | - Stephen J Haggarty
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Merel O Mol
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ging-Yuek R Hsiung
- Department of Medicine, Division of Neurology, University of British Columbia, Vancouver, BC, Canada
| | - Caroline Graff
- Division of Neurogeriatrics, Karolinska Institutet, Solna, Sweden
- Unit for Hereditary Dementias, Karolinska University Hospital, Solna, Sweden
| | - Linn Öijerstedt
- Division of Neurogeriatrics, Karolinska Institutet, Solna, Sweden
- Unit for Hereditary Dementias, Karolinska University Hospital, Solna, Sweden
| | - Manuela Neumann
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Neuropathology, University of Tübingen, Tübingen, Germany
| | - Yan Asmann
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, USA
| | | | - Saurabh Baheti
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | | - Kevin F Bieniek
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Leah Forsberg
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Hilary Heuer
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Argentina Lario Lago
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Ethan G Geier
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Jennifer S Yokoyama
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Alexis P Oddi
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Margaret Flanagan
- University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Qinwen Mao
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - John R Hodges
- Central Clinical School and Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
| | - John B Kwok
- University of Sydney, Sydney, NSW, Australia
- NeuRA, University of New South Wales, Randwick, NSW, Australia
| | | | - Matthis Synofzik
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Division Translational Genomics of Neurodegenerative Diseases, Center for Neurology and Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Carlo Wilke
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Division Translational Genomics of Neurodegenerative Diseases, Center for Neurology and Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Chiadi Onyike
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA
| | | | - Bret M Evers
- Division of Neuropathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Brittany N Dugger
- Department of Pathology and Laboratory Medicine, University of California, Davis Medical Center, Sacramento, CA, USA
| | - David G Munoz
- St. Michael's Hospital, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Julia Keith
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Lorne Zinman
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Ekaterina Rogaeva
- Krembil Discovery Tower, Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, ON, Canada
| | - EunRan Suh
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Tamar Gefen
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University, Chicago, IL, USA
| | - Changiz Geula
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University, Chicago, IL, USA
| | - Sandra Weintraub
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University, Chicago, IL, USA
| | - Janine Diehl-Schmid
- Department of Psychiatry and Psychotherapy, Technical University of Munich, Munich, Germany
- kbo-Inn-Salzach-Klinikum, Clinical Center for Psychiatry, Psychotherapy, Psychosomatic Medicine, Geriatrics and Neurology, Wasserburg/Inn, Germany
| | - Martin R Farlow
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Dieter Edbauer
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | | | | | - Laura L Donker Kaat
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Edward D Huey
- Department of Psychiatry and Human Behavior, Brown Alpert Medical School, Brown University, Providence, RI, USA
| | | | - Simon Mead
- MRC Prion Unit at University College London, Institute of Prion Diseases, London, UK
| | - Andrew King
- Department of Basic and Clinical Neuroscience, London Neurodegenerative Diseases Brain Bank, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Department of Clinical Neuropathology, King's College Hospital NHS Foundation Trust, London, UK
| | - Sigrun Roeber
- Centre for Neuropathology and Prion Research, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Alissa L Nana
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Nilufer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
| | | | | | | | - Ryan J Uitti
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
| | | | - Eliana Marisa Ramos
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Lea T Grinberg
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Maria Luisa Gorno Tempini
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Howard J Rosen
- Department of Pathology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Salvatore Spina
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Olivier Piguet
- School of Psychology and Brain and Mind Centre, University of Sydney, Sydney, SWA, Australia
| | - Murray Grossman
- Department of Neurology, Penn Frontotemporal Degeneration Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - C Dirk Keene
- University of Washington BioRepository and Integrated Neuropathology (BRaIN) lab, Harborview Medical Center, Seattle, WA, USA
| | - Lee-Way Jin
- M.I.N.D. Institute Laboratory, University of California, Davis Medical Center, Sacramento, CA, USA
| | - Johannes Prudlo
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Neurology, Rostock University Medical Center, Rostock, Germany
| | - Daniel H Geschwind
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Robert A Rissman
- Alzheimer's Therapeutic Research Institute, Keck School of Medicine of the University of Southern California, San Diego, CA, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Thomas G Beach
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Geidy E Serrano
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Thomas Arzberger
- Centre for Neuropathology and Prion Research, Ludwig-Maximilians-University of Munich, Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Jochen Herms
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Centre for Neuropathology and Prion Research, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Adam L Boxer
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Lawrence S Honig
- Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Jean P Vonsattel
- Department of Pathology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Oscar L Lopez
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Julia Kofler
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Charles L White
- Division of Neuropathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Marla Gearing
- Department of Pathology and Laboratory Medicine and Department of Neurology, Emory University, Atlanta, GA, USA
| | - Jonathan Glass
- Department of Pathology and Laboratory Medicine and Department of Neurology, Emory University, Atlanta, GA, USA
| | - Jonathan D Rohrer
- Department of Neurodegenerative Disease, Dementia Research Centre, University College London, Queen Square Institute of Neurology, London, UK
| | - David J Irwin
- Department of Neurology, Penn Frontotemporal Degeneration Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Edward B Lee
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Vivianna Van Deerlin
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Rudolph Castellani
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Marsel M Mesulam
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University, Chicago, IL, USA
| | - Maria C Tartaglia
- Krembil Discovery Tower, Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, ON, Canada
| | - Elizabeth C Finger
- Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Claire Troakes
- Department of Basic and Clinical Neuroscience, London Neurodegenerative Diseases Brain Bank, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Safa Al-Sarraj
- Department of Basic and Clinical Neuroscience, London Neurodegenerative Diseases Brain Bank, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- King's College Hospital NHS Foundation Trust, London, UK
| | - Clifton L Dalgard
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Bruce L Miller
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Harro Seelaar
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | | | - Ian Ra Mackenzie
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - John C van Swieten
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - William W Seeley
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Kristel Sleegers
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
| | | | - Joanna M Biernacka
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA
| | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium.
| |
Collapse
|
4
|
Lee GB, Yang C, Hu F, Hao L. Evaluating sample normalization methods for MS-based multi-omics and the application to a neurodegenerative mouse model. Analyst 2025; 150:1271-1279. [PMID: 39995368 PMCID: PMC11851094 DOI: 10.1039/d4an01573h] [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: 12/25/2024] [Accepted: 02/13/2025] [Indexed: 02/26/2025]
Abstract
Mass spectrometry (MS)-based omics methods have transformed biomedical research with accurate and high-throughput analysis of diverse molecules in biological systems. Recent technological advances also enabled multi-omics to be achieved from the same sample or on a single analytical platform. Sample normalization is a critical step in MS-omics studies but is usually conducted independently for each omics experiment. To bridge this technical gap, we evaluated different sample normalization methods suitable for analyzing proteins, lipids, and metabolites from the same sample for multi-omics analysis. We found that normalizing samples based on tissue weight or protein concentration before or after extraction generated distinct quantitative results. Normalizing samples first by tissue weight before extraction and then by protein concentration after extraction resulted in the lowest sample variation to reveal true biological differences. We then applied this two-step normalization method to investigate multi-omics profiles of mouse brains lacking the GRN gene. Loss-of-function mutations in the GRN gene lead to the deficiency of the progranulin protein and eventually cause neurodegeneration. Comparing the proteomics, lipidomics, and metabolomics profiles of GRN KO and WT mouse brains revealed molecular changes and pathways related to lysosomal dysfunction and neuroinflammation. In summary, we demonstrated the importance of selecting an appropriate normalization method during multi-omics sample preparation. Our normalization method is applicable to all tissue-based multi-omics studies, ensuring reliable and accurate biomolecule quantification for biological comparisons.
Collapse
Affiliation(s)
- Gwang Bin Lee
- Department of Chemistry & Biochemistry, University of Maryland, College Park, MD, 20742, USA.
| | - Cha Yang
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Fenghua Hu
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Ling Hao
- Department of Chemistry & Biochemistry, University of Maryland, College Park, MD, 20742, USA.
| |
Collapse
|
5
|
Yang Y, Liu X, Xu Y, Xing F, Yuan Z, Chen Y, Wu X, Wang C, Liang G. Coumarin Excimer Nanotube for Long-Time Lysosome Tracking. NANO LETTERS 2025. [PMID: 40033666 DOI: 10.1021/acs.nanolett.5c00041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Lysosome abnormality closely relates to a variety of diseases; thus, its long-time tracking could benefit accurate disease diagnosis. Current long-time lysosome imaging probes are not biocompatible enough, while compatible peptide-based probes are easily degraded by the abundant proteinases in lysosome. Herein, we rationally design a d-amino acid-containing peptide Cys(StBu)-d-Glu-Lys(coumarin)-d-Glu-CBT (Cou-D/L-CBT) which is subjected to intracellular GSH-initiated CBT-Cys click reaction and assembles into nanotubes in acidic lysosome. In vitro experiments showed that, under reduction environment and at pH 4.8, Cou-D/L-CBT assembled into nanotubes with an outer diameter of 156 nm, accompanied by "turn-on" coumarin excimer fluorescence at 550 nm. Cell experiments indicated that while Cou-D/L-CBT provided 29 h of lysosome fluorescence imaging, control probe Cou-L-CBT sustained only 6 h. We expect that our Cou-D/L-CBT could be applied for in vitro sensitive diagnosis of lysosome-related diseases in the clinic in the near future.
Collapse
Affiliation(s)
- Yang Yang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing 211189, China
| | - Xiaoyang Liu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing 211189, China
| | - Ying Xu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing 211189, China
| | - Fan Xing
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing 211189, China
| | - Zihan Yuan
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing 211189, China
| | - Yuxuan Chen
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing 210009, China
| | - Xiaofeng Wu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing 211189, China
| | - Cailian Wang
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing 210009, China
| | - Gaolin Liang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing 211189, China
| |
Collapse
|
6
|
Belbasis L, Morris S, van Duijn C, Bennett D, Walters R. Mendelian randomization identifies proteins involved in neurodegenerative diseases. Brain 2025:awaf018. [PMID: 40037332 DOI: 10.1093/brain/awaf018] [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: 02/24/2024] [Revised: 10/26/2024] [Accepted: 12/20/2024] [Indexed: 03/06/2025] Open
Abstract
Proteins are involved in multiple biological functions. High-throughput technologies have allowed the measurement of thousands of proteins in population biobanks. In this study, we aimed to identify proteins related to Alzheimer's disease, Parkinson's disease, multiple sclerosis and amyotrophic lateral sclerosis by leveraging large-scale genetic and proteomic data. We performed a two-sample cis Mendelian randomization study by selecting instrumental variables for the abundance of >2700 proteins measured by either Olink or SomaScan platforms in plasma from the UK Biobank and the deCODE Health Study. We also used the latest publicly available genome-wide association studies for the neurodegenerative diseases of interest. The potentially causal effect of proteins on neurodegenerative diseases was estimated based on the Wald ratio. We tested 13 377 protein-disease associations, identifying 169 associations that were statistically significant (5% false discovery rate). Evidence of co-localization between plasma protein abundance and disease risk (posterior probability > 0.80) was identified for 61 protein-disease pairs, leading to 50 unique protein-disease associations. Notably, 23 of 50 protein-disease associations corresponded to genetic loci not previously reported by genome-wide association studies. The two-sample Mendelian randomization and co-localization analysis also showed that APOE abundance in plasma was associated with three subcortical volumes (hippocampus, amygdala and nucleus accumbens) and white matter hyper-intensities, whereas PILRA and PILRB abundance in plasma was associated with caudate nucleus volume. Our study provided a comprehensive assessment of the effect of the human proteome that is currently measurable through two different platforms on neurodegenerative diseases. The newly associated proteins indicated the involvement of complement (C1S and C1R), microglia (SIRPA, SIGLEC9 and PRSS8) and lysosomes (CLN5) in Alzheimer's disease; the interleukin-6 pathway (CTF1) in Parkinson's disease; lysosomes (TPP1), blood-brain barrier integrity (MFAP2) and astrocytes (TNFSF13) in amyotrophic lateral sclerosis; and blood-brain barrier integrity (VEGFB), oligodendrocytes (PARP1), node of Ranvier and dorsal root ganglion (NCS1, FLRT3 and CDH15) and the innate immune system (CR1, AHSG and WARS) in multiple sclerosis. Our study demonstrates how harnessing large-scale genomic and proteomic data can yield new insights into the role of the plasma proteome in the pathogenesis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Lazaros Belbasis
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Sam Morris
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Cornelia van Duijn
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Derrick Bennett
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Robin Walters
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| |
Collapse
|
7
|
Rambarack N, Fodder K, Murthy M, Toomey C, de Silva R, Heutink P, Humphrey J, Raj T, Lashley T, Bettencourt C. DNA methylation as a contributor to dysregulation of STX6 and other frontotemporal lobar degeneration genetic risk-associated loci. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.21.634065. [PMID: 39975316 PMCID: PMC11838521 DOI: 10.1101/2025.01.21.634065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Frontotemporal Lobar Degeneration (FTLD) represents a spectrum of clinically, genetically, and pathologically heterogeneous neurodegenerative disorders characterised by progressive atrophy of the frontal and temporal lobes of the brain. The two major FTLD pathological subgroups are FTLD-TDP and FTLD-tau. While the majority of FTLD cases are sporadic, heterogeneity also exists within the familial cases, typically involving mutations in MAPT, GRN or C9orf72, which is not fully explained by known genetic mechanisms. We sought to address this gap by investigating the effect of epigenetic modifications, specifically DNA methylation variation, on genes associated with FTLD genetic risk in different FTLD subtypes. We compiled a list of genes associated with genetic risk of FTLD using text-mining databases and literature searches. Frontal cortex DNA methylation profiles were derived from three FTLD datasets containing different subgroups of FTLD-TDP and FTLD-tau: FTLD1m (N = 23) containing FTLD-TDP type A C9orf72 mutation carriers and TDP Type C sporadic cases, FTLD2m (N = 48) containing FTLD-Tau MAPT mutation carriers, FTLD-TDP Type A GRN mutation carriers, and FTLD-TDP Type B C9orf72 mutation carriers and FTLD3m (N = 163) progressive supranuclear palsy (PSP) cases, and corresponding controls. To investigate the downstream effects of DNA methylation further, we then leveraged transcriptomic and proteomic datasets for FTLD cases and controls to examine gene and protein expression levels. Our analysis revealed shared promoter region hypomethylation in STX6 across FTLD-TDP and FTLD-tau subtypes, though the largest effect size was observed in the PSP cases compared to controls (delta-beta = -32%, adjusted-p value=0.002). We also observed dysregulation of the STX6 gene and protein expression across FTLD subtypes. Additionally, we performed a detailed examination of MAPT, GRN and C9orf72 in subtypes with and without the presence of the genetic mutations and observed nominally significant differentially methylated CpGs in variable positions across the genes, often with unique patterns and downstream consequences in gene/protein expression in mutation carriers. We highlight the contribution of DNA methylation at different gene regions in regulating the expression of genes previously associated with genetic risk of FTLD, including STX6. We analysed the relationship of subtypes and presence of mutations with this epigenetic mechanism to increase our understanding of how these mechanisms interact in FTLD.
Collapse
Affiliation(s)
- Naiomi Rambarack
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Katherine Fodder
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Megha Murthy
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Christina Toomey
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- The Francis Crick Institute, London, UK
| | - Rohan de Silva
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London, UK
| | - Peter Heutink
- German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Jack Humphrey
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Towfique Raj
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Tammaryn Lashley
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Conceição Bettencourt
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| |
Collapse
|
8
|
Luettel DM, Terluk MR, Roh J, Weinreb NJ, Kartha RV. Emerging biomarkers in Gaucher disease. Adv Clin Chem 2025; 124:1-56. [PMID: 39818434 DOI: 10.1016/bs.acc.2024.11.001] [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: 01/18/2025]
Abstract
Gaucher disease (GD) is a rare lysosomal disorder characterized by the accumulation of glycosphingolipids in macrophages resulting from glucocerebrosidase (GCase) deficiency. The accumulation of toxic substrates, which causes the hallmark symptoms of GD, is dependent on the extent of enzyme dysfunction. Accordingly, three distinct subtypes have been recognized, with type 1 GD (GD1) as the common and milder form, while types 2 (GD2) and 3 (GD3) are categorized as neuronopathic and severe. Manifestations variably include hepatosplenomegaly, anemia, thrombocytopenia, easy bruising, inflammation, bone pain and other skeletal pathologies, abnormal eye movements and neuropathy. Although the molecular basis of GD is relatively well understood, currently used biomarkers are nonspecific and inadequate for making finer distinctions between subtypes and in evaluating changes in disease status and guiding therapy. Thus, there is continued effort to investigate and identify potential biomarkers to improve GD diagnosis, monitoring and potential identification of novel therapeutic targets. Here, we provide a comprehensive review of emerging biomarkers in GD that can enhance current understanding and improve quality of life through better testing, disease management and treatment.
Collapse
Affiliation(s)
- Danielle M Luettel
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, United States
| | - Marcia R Terluk
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, United States
| | - Jaehyeok Roh
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, United States
| | - Neal J Weinreb
- Department of Human Genetics, Leonard Miller School of Medicine of University of Miami, Miami, FL, United States
| | - Reena V Kartha
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, United States.
| |
Collapse
|
9
|
Du H, Yang C, Nana AL, Seeley WW, Smolka M, Hu F. WITHDRAWN: Progranulin inhibits phospholipase sPLA2-IIA to control neuroinflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.06.535844. [PMID: 37066328 PMCID: PMC10104136 DOI: 10.1101/2023.04.06.535844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
The authors have withdrawn this manuscript because more work is needed to fully define the role of sPLA2-IIA. Therefore, the authors do not wish this work to be cited as reference for the project. If you have any questions, please contact the corresponding author.
Collapse
|
10
|
Borrego-Ecija S, Juncà-Parella J, Vandebergh M, Pérez Millan A, Balasa M, Llado A, Bouzigues A, Russell LL, Foster PH, Ferry-Bolder E, Van Swieten JC, Jiskoot LC, Seelaar H, Laforce R, Graff C, Galimberti D, Vandenberghe R, de Mendonça A, Tiraboschi P, Santana I, Gerhard A, Levin J, Sorbi S, Otto M, Pasquier F, Ducharme S, Butler C, Le Ber I, Finger E, Tartaglia MC, Masellis M, Rowe JB, Synofzik M, Moreno F, Borroni B, Rademakers R, Rohrer JD, Sánchez-Valle R. Association of Initial Side of Brain Atrophy With Clinical Features and Disease Progression in Patients With GRN Frontotemporal Dementia. Neurology 2024; 103:e209944. [PMID: 39527772 PMCID: PMC11558542 DOI: 10.1212/wnl.0000000000209944] [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: 07/15/2024] [Accepted: 10/02/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Pathogenic variants in the GRN gene cause frontotemporal dementia (FTD-GRN) with marked brain asymmetry. This study aims to assess whether the disease progression of FTD-GRN depends on the initial side of the atrophy. We also investigated the potential use of brain asymmetry as a biomarker of the disease. METHODS Retrospective examination of data from the prospective Genetic Frontotemporal Initiative (GENFI) cohort study that recruits individuals who carry or were at risk of carrying a pathogenic variant causing FTD. GENFI participants underwent a standardized clinical and neuropsychological assessment, MRI, and a blood sample test yearly. We generated an asymmetry index for brain MRI to characterize brain asymmetry in participants with or at risk of FTD-GRN. Depending on the side of the asymmetry, we classified symptomatic GRN patients as right-GRN or left-GRN and compared their clinical features and disease progression. We generated generalized additive models to study how the asymmetry index evolves in carriers and noncarriers and compare its models with others created with volumetric values and plasma neurofilament light chain. RESULTS A total of 399 participants (mean age 49.7 years, 59% female) were included (63 symptomatic carriers, 177 presymptomatic carriers, and 159 noncarriers). Symptomatic carriers showed higher brain asymmetry (11.6) than noncarriers (1.0, p < 0.001) and presymptomatic carriers (1.0, p < 0.001), making it possible to classify most of them as right-GRN (n = 21) or left-GRN (n = 36). Patients with right-GRN showed more disease severity at baseline (β = 6.9, 95% CI 2.4-11.0, p = 0.003) but a lower deterioration by year (β = -1.5, 95% CI -2.7 to -0.31, p = 0.015) than patients with left-GRN. Brain asymmetry could be found in GRN carriers 10.4 years before the onset of the symptoms (standard difference 0.85, CI 0.01-1.68). DISCUSSION FTD-GRN affects the brain hemispheres asymmetrically and causes 2 anatomical asymmetry patterns depending on the side of the disease onset. We demonstrated that these 2 anatomical asymmetry patterns present different symptoms, severity at the time of the first visit, and different disease courses. Our results also suggest brain asymmetry as a possible biomarker of conversion in GRN carriers.
Collapse
Affiliation(s)
| | | | - Marijne Vandebergh
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Agnès Pérez Millan
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Mircea Balasa
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Albert Llado
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Arabella Bouzigues
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Lucy Louise Russell
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Phoebe H. Foster
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Eve Ferry-Bolder
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - John C. Van Swieten
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Lize Corrine Jiskoot
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Harro Seelaar
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Robert Laforce
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Caroline Graff
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Daniela Galimberti
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Rik Vandenberghe
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Alexandre de Mendonça
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Pietro Tiraboschi
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Isabel Santana
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Alexander Gerhard
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Johannes Levin
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Sandro Sorbi
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Markus Otto
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Florence Pasquier
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Simon Ducharme
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Christopher Butler
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Isabelle Le Ber
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Elizabeth Finger
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Maria Carmela Tartaglia
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Mario Masellis
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - James B. Rowe
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Matthis Synofzik
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Fermin Moreno
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Barbara Borroni
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Rosa Rademakers
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | - Jonathan Daniel Rohrer
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| | | | - for the Genetic Frontotemporal Initiative (GENFI)
- From the Alzheimer's Disease and Other Cognitive Disorders Unit (S.B.-E., J.J.-P., A.P.M., M.B., A.L., R.S.-V.), Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Spain; VIB Center for Molecular Neurology (M.V., R.R.); Department of Biomedical Sciences (M.V., R.R.), University of Antwerp, Belgium; Dementia Research Centre (A.B., L.L.R., P.H.F., E.F.-B., J.D.R.), Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (J.C.V.S., L.C.J., H.S.), Erasmus Medical Centre, Rotterdam, Netherlands; Clinique Interdisciplinaire de Mémoire (R.L.), Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Canada; Division of Neurogeriatrics, Bioclinicum (C.G.), Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Karolinska Institutet; Unit for Hereditary Dementias (C.G.), Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden; Department of Biomedical (D.G.), Surgical and Dental Sciences, University of Milan; Fondazione Ca' Granda (D.G.), IRCCS Ospedale Policlinico, Milan, Italy; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven; Neurology Service (R.V.), University Hospitals Leuven; Leuven Brain Institute (R.V.), KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione IRCCS Istituto Neurologico Carlo Besta (P.T.), Milano, Italy; Neurology Service (I.S.), Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra; Center for Neuroscience and Cell Biology (I.S.), Faculty of Medicine, University of Coimbra, Portugal; Division of Psychology Communication and Human Neuroscience (A.G.), Wolfson Molecular Imaging Centre, University of Manchester, United Kingdom; Department of Nuclear Medicine (A.G.), Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen; Department of Geriatric Medicine (A.G.), Klinikum Hochsauerland, Arnsberg; Department of Neurology (J.L.), Ludwig-Maximilians Universität München; German Center for Neurodegenerative Diseases (DZNE) (J.L.); Munich Cluster of Systems Neurology (SyNergy) (J.L.), Munich, Germany; Department of Neurofarba (S.S.), University of Florence; IRCCS Fondazione Don Carlo Gnocchi (S.S.), Florence, Italy; Department of Neurology (M.O.), University of Ulm, Germany; Univ Lille (F.P.), France; Department of Psychiatry (S.D.), McGill University Health Centre, McConnell Brain Imaging Centre (S.D.), Montreal Neurological Institute, McGill University, Montreal, Québec, Canada; Medical Sciences Division (C.B.), Nuffield Department of Clinical Neurosciences, University of Oxford, Department of Brain Sciences (C.B.), Imperial College London, United Kingdom; Sorbonne Université (I.L.B.), Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225; Centre de référence des démences rares ou précoces (I.L.B.), IM2A, Département de Neurologie; Département de Neurologie (I.L.B.), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London; Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), Ontario; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute, University of Toronto, Canada; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust (J.B.R.), University of Cambridge, United Kingdom; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Germany; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neurology Unit (B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; and Department of Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL
| |
Collapse
|
11
|
Ali NH, Al-Kuraishy HM, Al-Gareeb AI, Alnaaim SA, Hetta HF, Saad HM, Batiha GES. A Mutual Nexus Between Epilepsy and α-Synuclein: A Puzzle Pathway. Mol Neurobiol 2024; 61:10198-10215. [PMID: 38703341 DOI: 10.1007/s12035-024-04204-6] [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: 07/01/2023] [Accepted: 04/12/2024] [Indexed: 05/06/2024]
Abstract
Alpha-synuclein (α-Syn) is a specific neuronal protein that regulates neurotransmitter release and trafficking of synaptic vesicles. Exosome-associated α-Syn which is specific to the central nervous system (CNS) is involved in the pathogenesis of epilepsy. Therefore, this review aimed to elucidate the possible link between α-Syn and epilepsy, and how it affects the pathophysiology of epilepsy. A neurodegenerative protein such as α-Syn is implicated in the pathogenesis of epilepsy. Evidence from preclinical and clinical studies revealed that upregulation of α-Syn induces progressive neuronal dysfunctions through induction of oxidative stress, neuroinflammation, and inhibition of autophagy in a vicious cycle with subsequent development of severe epilepsy. In addition, accumulation of α-Syn in epilepsy could be secondary to the different cellular alterations including oxidative stress, neuroinflammation, reduction of brain-derived neurotrophic factor (BDNF) and progranulin (PGN), and failure of the autophagy pathway. However, the mechanism of α-Syn-induced-epileptogenesis is not well elucidated. Therefore, α-Syn could be a secondary consequence of epilepsy. Preclinical and clinical studies are warranted to confirm this causal relationship.
Collapse
Affiliation(s)
- Naif H Ali
- Department of Internal Medicine, Medical College, Najran University, Najran, Kingdom of Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, M.B.Ch.B, FRCP, P.O. Box 14132, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Jabir Ibn Hayyan Medical University, Al-Ameer Qu, P.O. Box 13, Kufa, Najaf, Iraq
| | - Saud A Alnaaim
- Clinical Neurosciences Department, College of Medicine, King Faisal University, Hofuf, Saudi Arabia
| | - Helal F Hetta
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut, 71515, Egypt
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Matrouh, 51744, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt.
| |
Collapse
|
12
|
Smith DM, Aggarwal G, Niehoff ML, Jones SA, Banerjee S, Farr SA, Nguyen AD. Biochemical, Biomarker, and Behavioral Characterization of the Grn R493X Mouse Model of Frontotemporal Dementia. Mol Neurobiol 2024; 61:9708-9722. [PMID: 38696065 PMCID: PMC11496013 DOI: 10.1007/s12035-024-04190-9] [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: 06/07/2023] [Accepted: 04/17/2024] [Indexed: 05/14/2024]
Abstract
Heterozygous loss-of-function mutations in the progranulin gene (GRN) are a major cause of frontotemporal dementia due to progranulin haploinsufficiency; complete deficiency of progranulin causes neuronal ceroid lipofuscinosis. Several progranulin-deficient mouse models have been generated, including both knockout mice and knockin mice harboring a common patient mutation (R493X). However, the GrnR493X mouse model has not been characterized completely. Additionally, while homozygous GrnR493X and Grn knockout mice have been extensively studied, data from heterozygous mice is still limited. Here, we performed more in-depth characterization of heterozygous and homozygous GrnR493X knockin mice, which includes biochemical assessments, behavioral studies, and analysis of fluid biomarkers. In the brains of homozygous GrnR493X mice, we found increased phosphorylated TDP-43 along with increased expression of lysosomal genes, markers of microgliosis and astrogliosis, pro-inflammatory cytokines, and complement factors. Heterozygous GrnR493X mice did not have increased TDP-43 phosphorylation but did exhibit limited increases in lysosomal and inflammatory gene expression. Behavioral studies found social and emotional deficits in GrnR493X mice that mirror those observed in Grn knockout mouse models, as well as impairment in memory and executive function. Overall, the GrnR493X knockin mouse model closely phenocopies Grn knockout models. Lastly, in contrast to homozygous knockin mice, heterozygous GrnR493X mice do not have elevated levels of fluid biomarkers previously identified in humans, including neurofilament light chain (NfL) and glial fibrillary acidic protein (GFAP) in both plasma and CSF. These results may help to inform pre-clinical studies that use this Grn knockin mouse model and other Grn knockout models.
Collapse
Affiliation(s)
- Denise M Smith
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, USA
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, USA
- Institute for Translational Neuroscience, Saint Louis University, St. Louis, USA
| | - Geetika Aggarwal
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, USA
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, USA
- Institute for Translational Neuroscience, Saint Louis University, St. Louis, USA
| | - Michael L Niehoff
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, USA
- Veterans Affairs Medical Center, St. Louis, USA
| | - Spencer A Jones
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, USA
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, USA
- Institute for Translational Neuroscience, Saint Louis University, St. Louis, USA
| | - Subhashis Banerjee
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, USA
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, USA
- Institute for Translational Neuroscience, Saint Louis University, St. Louis, USA
| | - Susan A Farr
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, USA
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, USA
- Institute for Translational Neuroscience, Saint Louis University, St. Louis, USA
- Veterans Affairs Medical Center, St. Louis, USA
| | - Andrew D Nguyen
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, USA.
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, USA.
- Institute for Translational Neuroscience, Saint Louis University, St. Louis, USA.
| |
Collapse
|
13
|
Poniatowski ŁA, Joniec-Maciejak I, Wawer A, Sznejder-Pachołek A, Machaj E, Ziętal K, Mirowska-Guzel D. Dose-Ranging Effects of the Intracerebral Administration of Atsttrin in Experimental Model of Parkinson's Disease Induced by 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in Mice. Mol Neurobiol 2024; 61:9432-9458. [PMID: 38642286 PMCID: PMC11496375 DOI: 10.1007/s12035-024-04161-0] [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: 02/03/2024] [Accepted: 04/02/2024] [Indexed: 04/22/2024]
Abstract
Parkinson's disease is one of the most common neurodegenerative disorders characterized by a multitude of motor and non-motor clinical symptoms resulting from the progressive and long-lasting abnormal loss of nigrostriatal dopaminergic neurons. Currently, the available treatments for patients with Parkinson's disease are limited and exert only symptomatic effects, without adequate signs of delaying or stopping the progression of the disease. Atsttrin constitutes the bioengineered protein which ultrastructure is based on the polypeptide chain frame of the progranulin (PGRN), which exerts anti-inflammatory effects through the inhibition of TNFα. The conducted preclinical studies suggest that the therapeutic implementation of Atsttrin may be potentially effective in the treatment of neurodegenerative diseases that are associated with the occurrence of neuroinflammatory processes. The aim of the proposed study was to investigate the effect of direct bilateral intracerebral administration of Atsttrin using stereotactic methods in the preclinical C57BL/6 mouse model of Parkinson's disease inducted by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) intoxication. The analysis of the dose dependency effects of the increasing doses of Atsttrin has covered a number of parameters and markers regarding neurodegenerative processes and inflammatory responses including IL-1α, TNFα, IL-6, TH, and TG2 mRNA expressions. Accordingly, the evaluation of the changes in the neurochemical profile included DA, DOPAC, 3-MT, HVA, NA, MHPG, 5-HT, and 5-HIAA concentration levels. The intracerebral administration of Atsttrin into the striatum effectively attenuated the neuroinflammatory reaction in evaluated neuroanatomical structures. Furthermore, the partial restoration of monoamine content and its metabolic turnover were observed. In this case, taking into account the previously described pharmacokinetic profile and extrapolated bioavailability as well as the stability characteristics of Atsttrin, an attempt was made to describe as precisely as possible the quantitative and qualitative effects of increasing doses of the compound within the brain tissue microenvironment in the presented preclinical model of the disease. Collectively, this findings demonstrated that the intracerebral administration of Atsttrin may represent a potential novel therapeutic method for the treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Łukasz A Poniatowski
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
- Department of Neurosurgery, Dietrich-Bonhoeffer-Klinikum, Salvador-Allende-Straße 30, 17036, Neubrandenburg, Germany
| | - Ilona Joniec-Maciejak
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland.
| | - Adriana Wawer
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
| | - Anna Sznejder-Pachołek
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
| | - Ewa Machaj
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
| | - Katarzyna Ziętal
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
| | - Dagmara Mirowska-Guzel
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
| |
Collapse
|
14
|
Lin Y, Zhao X, Liou B, Fannin V, Zhang W, Setchell KDR, Wang X, Pan D, Grabowski GA, Liu CJ, Sun Y. Intrinsic link between PGRN and Gba1 D409V mutation dosage in potentiating Gaucher disease. Hum Mol Genet 2024; 33:1771-1788. [PMID: 39101473 PMCID: PMC11458007 DOI: 10.1093/hmg/ddae113] [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: 03/28/2024] [Revised: 07/08/2024] [Accepted: 07/25/2024] [Indexed: 08/06/2024] Open
Abstract
Gaucher disease (GD) is caused by biallelic GBA1/Gba1 mutations that encode defective glucocerebrosidase (GCase). Progranulin (PGRN, encoded by GRN/Grn) is a modifier of GCase, but the interplay between PGRN and GCase, specifically GBA1/Gba1 mutations, contributing to GD severity is unclear. Mouse models were developed with various dosages of Gba1 D409V mutation against the PGRN deficiency (Grn-/-) [Grn-/-;Gba1D409V/WT (PG9Vwt), Grn-/-;Gba1D409V/D409V (PG9V), Grn-/-;Gba1D409V/Null (PG9VN)]. Disease progression in those mouse models was characterized by biochemical, pathological, transcriptomic, and neurobehavioral analyses. Compared to PG9Vwt, Grn-/-;Gba1WT/Null and Grn-/- mice that had a higher level of GCase activity and undetectable pathologies, homozygous or hemizygous D409V in PG9V or PG9VN, respectively, resulted in profound inflammation and neurodegeneration. PG9VN mice exhibited much earlier onset, shorter life span, tissue fibrosis, and more severe phenotypes than PG9V mice. Glycosphingolipid accumulation, inflammatory responses, lysosomal-autophagy dysfunction, microgliosis, retinal gliosis, as well as α-Synuclein increases were much more pronounced in PG9VN mice. Neurodegeneration in PG9VN was characterized by activated microglial phagocytosis of impaired neurons and programmed cell death due to necrosis and, possibly, pyroptosis. Brain transcriptomic analyses revealed the intrinsic relationship between D409V dosage, and the degree of altered gene expression related to lysosome dysfunction, microgliosis, and neurodegeneration in GD, suggesting the disease severity is dependent on a GCase activity threshold related to Gba1 D409V dosage and loss of PGRN. These findings contribute to a deeper understanding of GD pathogenesis by elucidating additional underlying mechanisms of interplay between PGRN and Gba1 mutation dosage in modulating GCase function and disease severity in GD and GBA1-associated neurodegenerative diseases.
Collapse
Affiliation(s)
- Yi Lin
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, United States
| | - Xiangli Zhao
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, 789 Howard Avenue, New Haven, CT 06519, United States
| | - Benjamin Liou
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, United States
| | - Venette Fannin
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, United States
| | - Wujuan Zhang
- Department of Pathology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, United States
| | - Kenneth D R Setchell
- Department of Pathology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH 45229, United States
| | - Xiaohong Wang
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, United States
| | - Dao Pan
- Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH 45229, United States
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, United States
| | - Gregory A Grabowski
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH 45229, United States
| | - Chuan-ju Liu
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, 789 Howard Avenue, New Haven, CT 06519, United States
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, 301 East 17th Street, New York, NY 10003, United States
| | - Ying Sun
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH 45229, United States
| |
Collapse
|
15
|
Yan D, Zhang Y, Huang Y, Ouyang W. Progranulin Facilitates Corneal Repair Through Dual Mechanisms of Inflammation Suppression and Regeneration Promotion. Inflammation 2024; 47:1648-1666. [PMID: 38460093 DOI: 10.1007/s10753-024-01999-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 02/12/2024] [Accepted: 02/28/2024] [Indexed: 03/11/2024]
Abstract
The cornea serves as a vital protective barrier for the eye; however, it is prone to injury and damage that can disrupt corneal epithelium and nerves, triggering inflammation. Therefore, understanding the biological effects and molecular mechanisms involved in corneal wound healing and identifying drugs targeting these pathways is crucial for researchers in this field. This study aimed to investigate the therapeutic potential of progranulin (PGRN) in treating corneal injuries. Our findings demonstrated that PGRN significantly enhanced corneal wound repair by accelerating corneal re-epithelialization and re-innervation. In vitro experiments with cultured epithelial cells and trigeminal ganglion cells further revealed that PGRN stimulated corneal epithelial cell proliferation and promoted axon growth in trigeminal ganglion cells. Through RNA-sequencing (RNA-seq) analysis and other experimental techniques, we discovered that PGRN exerted its healing effects modulating Wnt signaling pathway, which played a critical role in repairing epithelial cells and promoting axon regeneration in trigeminal neurons. Importantly, our study highlighted the anti-inflammatory properties of PGRN by inhibiting the NF-κB signaling pathway, leading to decreased infiltration of macrophages. In conclusion, our findings underscored the potential of PGRN in facilitating corneal wound healing by promoting corneal epithelial cell proliferation, trigeminal ganglion cell axon regeneration, and suppressing ocular inflammation. These results suggest that PGRN could potentially expedite the healing process and improve visual outcomes in patients with corneal injuries.
Collapse
Affiliation(s)
- Dan Yan
- Department of Ophthalmology, the Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Guizhou, Xiamen, China
- Xiamen University affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, China
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Yunuo Zhang
- Department of Ophthalmology, the Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Guizhou, Xiamen, China
- Xiamen University affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, China
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Yuhan Huang
- Department of Ophthalmology, the Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Guizhou, Xiamen, China
- Xiamen University affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, China
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Weijie Ouyang
- Department of Ophthalmology, the Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Guizhou, Xiamen, China.
- Xiamen University affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, China.
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.
| |
Collapse
|
16
|
Lee GB, Mazli WNAB, Hao L. Multiomics Evaluation of Human iPSCs and iPSC-Derived Neurons. J Proteome Res 2024; 23:3149-3160. [PMID: 38415376 PMCID: PMC11799864 DOI: 10.1021/acs.jproteome.3c00790] [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] [Indexed: 02/29/2024]
Abstract
Human induced pluripotent stem cells (iPSCs) can be differentiated into neurons, providing living human neurons to model brain diseases. However, it is unclear how different types of molecules work together to regulate stem cell and neuron biology in healthy and disease states. In this study, we conducted integrated proteomics, lipidomics, and metabolomics analyses with confident identification, accurate quantification, and reproducible measurements to compare the molecular profiles of human iPSCs and iPSC-derived neurons. Proteins, lipids, and metabolites related to mitosis, DNA replication, pluripotency, glycosphingolipids, and energy metabolism were highly enriched in iPSCs, whereas synaptic proteins, neurotransmitters, polyunsaturated fatty acids, cardiolipins, and axon guidance pathways were highly enriched in neurons. Mutations in the GRN gene lead to the deficiency of the progranulin (PGRN) protein, which has been associated with various neurodegenerative diseases. Using this multiomics platform, we evaluated the impact of PGRN deficiency on iPSCs and neurons at the whole-cell level. Proteomics, lipidomics, and metabolomics analyses implicated PGRN's roles in neuroinflammation, purine metabolism, and neurite outgrowth, revealing commonly altered pathways related to neuron projection, synaptic dysfunction, and brain metabolism. Multiomics data sets also pointed toward the same hypothesis that neurons seem to be more susceptible to PGRN loss compared to iPSCs, consistent with the neurological symptoms and cognitive impairment from patients carrying inherited GRN mutations.
Collapse
Affiliation(s)
- Gwang Bin Lee
- Department of Chemistry, The George Washington University, 800 22nd St. NW, Washington, D.C. 20052, United States
| | - Wan Nur Atiqah Binti Mazli
- Department of Chemistry, The George Washington University, 800 22nd St. NW, Washington, D.C. 20052, United States
| | - Ling Hao
- Department of Chemistry, The George Washington University, 800 22nd St. NW, Washington, D.C. 20052, United States
| |
Collapse
|
17
|
Huq A, Thompson B, Winship I. Clinical application of whole genome sequencing in young onset dementia: challenges and opportunities. Expert Rev Mol Diagn 2024; 24:659-675. [PMID: 39135326 DOI: 10.1080/14737159.2024.2388765] [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: 04/25/2024] [Accepted: 08/01/2024] [Indexed: 08/30/2024]
Abstract
INTRODUCTION Young onset dementia (YOD) by its nature is difficult to diagnose. Despite involvement of multidisciplinary neurogenetics services, patients with YOD and their families face significant diagnostic delays. Genetic testing for people with YOD currently involves a staggered, iterative approach. There is currently no optimal single genetic investigation that simultaneously identifies the different genetic variants resulting in YOD. AREAS COVERED This review discusses the advances in clinical genomic testing for people with YOD. Whole genome sequencing (WGS) can be employed as a 'one stop shop' genomic test for YOD. In addition to single nucleotide variants, WGS can reliably detect structural variants, short tandem repeat expansions, mitochondrial genetic variants as well as capture single nucleotide polymorphisms for the calculation of polygenic risk scores. EXPERT OPINION WGS, when used as the initial genetic test, can enhance the likelihood of a precision diagnosis and curtail the time taken to reach this. Finding a clinical diagnosis using WGS can reduce invasive and expensive investigations and could be cost effective. These advances need to be balanced against the limitations of the technology and the genetic counseling needs for these vulnerable patients and their families.
Collapse
Affiliation(s)
- Aamira Huq
- Department of Genomic Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
- Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - Bryony Thompson
- Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
- Department of Pathology, University of Melbourne, Parkville, Victoria, Australia
| | - Ingrid Winship
- Department of Genomic Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
- Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
18
|
Prajapati SK, Pathak A, Samaiya PK. Alzheimer's disease: from early pathogenesis to novel therapeutic approaches. Metab Brain Dis 2024; 39:1231-1254. [PMID: 39046584 DOI: 10.1007/s11011-024-01389-6] [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: 12/06/2023] [Accepted: 07/15/2024] [Indexed: 07/25/2024]
Abstract
The mainstay behind Alzheimer's disease (AD) remains unknown due to the elusive pathophysiology of the disease. Beta-amyloid and phosphorylated Tau is still widely incorporated in various research studies while studying AD. However, they are not sufficient. Therefore, many scientists and researchers have dug into AD studies to deliver many innovations in this field. Many novel biomarkers, such as phosphoglycerate-dehydrogenase, clusterin, microRNA, and a new peptide ratio (Aβ37/Aβ42) in cerebral-spinal fluid, plasma glial-fibrillary-acidic-protein, and lipid peroxidation biomarkers, are mushrooming. They are helping scientists find breakthroughs and substantiating their research on the early detection of AD. Neurovascular unit dysfunction in AD is a significant discovery that can help us understand the relationship between neuronal activity and cerebral blood flow. These new biomarkers are promising and can take these AD studies to another level. There have also been big steps forward in diagnosing and finding AD. One example is self-administered-gerocognitive-examination, which is less expensive and better at finding AD early on than mini-mental-state-examination. Quantum brain sensors and electrochemical biosensors are innovations in the detection field that must be explored and incorporated into the studies. Finally, novel innovations in AD studies like nanotheranostics are the future of AD treatment, which can not only diagnose and detect AD but also offer treatment. Non-pharmacological strategies to treat AD have also yielded interesting results. Our literature review spans from 1957 to 2022, capturing research and trends in the field over six decades. This review article is an update not only on the recent advances in the search for credible biomarkers but also on the newer detection techniques and therapeutic approaches targeting AD.
Collapse
Affiliation(s)
- Santosh Kumar Prajapati
- Bhavdiya Institute of Pharmaceutical Sciences and Research, Ayodhya, UP, India
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, 33613, USA
| | - Arjit Pathak
- Department of Pharmacy Shri G.S. Institute of Technology and Science, Indore, 452003, Madhya Pradesh, India
| | - Puneet K Samaiya
- Department of Pharmacy Shri G.S. Institute of Technology and Science, Indore, 452003, Madhya Pradesh, India.
| |
Collapse
|
19
|
Hällqvist J, Bartl M, Dakna M, Schade S, Garagnani P, Bacalini MG, Pirazzini C, Bhatia K, Schreglmann S, Xylaki M, Weber S, Ernst M, Muntean ML, Sixel-Döring F, Franceschi C, Doykov I, Śpiewak J, Vinette H, Trenkwalder C, Heywood WE, Mills K, Mollenhauer B. Plasma proteomics identify biomarkers predicting Parkinson's disease up to 7 years before symptom onset. Nat Commun 2024; 15:4759. [PMID: 38890280 PMCID: PMC11189460 DOI: 10.1038/s41467-024-48961-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 05/20/2024] [Indexed: 06/20/2024] Open
Abstract
Parkinson's disease is increasingly prevalent. It progresses from the pre-motor stage (characterised by non-motor symptoms like REM sleep behaviour disorder), to the disabling motor stage. We need objective biomarkers for early/pre-motor disease stages to be able to intervene and slow the underlying neurodegenerative process. Here, we validate a targeted multiplexed mass spectrometry assay for blood samples from recently diagnosed motor Parkinson's patients (n = 99), pre-motor individuals with isolated REM sleep behaviour disorder (two cohorts: n = 18 and n = 54 longitudinally), and healthy controls (n = 36). Our machine-learning model accurately identifies all Parkinson patients and classifies 79% of the pre-motor individuals up to 7 years before motor onset by analysing the expression of eight proteins-Granulin precursor, Mannan-binding-lectin-serine-peptidase-2, Endoplasmatic-reticulum-chaperone-BiP, Prostaglaindin-H2-D-isomaerase, Interceullular-adhesion-molecule-1, Complement C3, Dickkopf-WNT-signalling pathway-inhibitor-3, and Plasma-protease-C1-inhibitor. Many of these biomarkers correlate with symptom severity. This specific blood panel indicates molecular events in early stages and could help identify at-risk participants for clinical trials aimed at slowing/preventing motor Parkinson's disease.
Collapse
Affiliation(s)
- Jenny Hällqvist
- UCL Institute of Child Health and Great Ormond Street Hospital, London, UK.
- UCL Queen Square Institute of Neurology, Clinical and Movement Neurosciences, London, UK.
| | - Michael Bartl
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany.
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Goettingen, Goettingen, Germany.
| | - Mohammed Dakna
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
| | | | - Paolo Garagnani
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | | | - Chiara Pirazzini
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Kailash Bhatia
- National Hospital for Neurology & Neurosurgery, Queen Square, WC1N3BG, London, UK
| | | | - Mary Xylaki
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
| | - Sandrina Weber
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
| | - 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
| | - Claudio Franceschi
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Ivan Doykov
- UCL Institute of Child Health and Great Ormond Street Hospital, London, UK
| | - Justyna Śpiewak
- UCL Institute of Child Health and Great Ormond Street Hospital, London, UK
| | - Héloїse Vinette
- UCL Institute of Child Health and Great Ormond Street Hospital, London, UK
- UCL: Food, Microbiomes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Claudia Trenkwalder
- Paracelsus-Elena-Klinik, Kassel, Germany
- Department of Neurosurgery, University Medical Center Goettingen, Goettingen, Germany
| | - Wendy E Heywood
- UCL Institute of Child Health and Great Ormond Street Hospital, London, UK
| | - Kevin Mills
- UCL Queen Square Institute of Neurology, Clinical and Movement Neurosciences, London, UK
| | - Brit Mollenhauer
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
- Paracelsus-Elena-Klinik, Kassel, Germany
| |
Collapse
|
20
|
Cook AK, Greathouse KM, Manuel PN, Cooper NH, Eberhardt JM, Freeman CD, Weber AJ, Herskowitz JH, Arrant AE. Dendritic spine head diameter is reduced in the prefrontal cortex of progranulin haploinsufficient mice. Mol Brain 2024; 17:33. [PMID: 38840181 PMCID: PMC11155153 DOI: 10.1186/s13041-024-01095-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 04/26/2024] [Indexed: 06/07/2024] Open
Abstract
Loss-of-function mutations in the progranulin (GRN) gene are an autosomal dominant cause of Frontotemporal Dementia (FTD). These mutations typically result in haploinsufficiency of the progranulin protein. Grn+/- mice provide a model for progranulin haploinsufficiency and develop FTD-like behavioral abnormalities by 9-10 months of age. In previous work, we demonstrated that Grn+/- mice develop a low dominance phenotype in the tube test that is associated with reduced dendritic arborization of layer II/III pyramidal neurons in the prelimbic region of the medial prefrontal cortex (mPFC), a region key for social dominance behavior in the tube test assay. In this study, we investigated whether progranulin haploinsufficiency induced changes in dendritic spine density and morphology. Individual layer II/III pyramidal neurons in the prelimbic mPFC of 9-10 month old wild-type or Grn+/- mice were targeted for iontophoretic microinjection of fluorescent dye, followed by high-resolution confocal microscopy and 3D reconstruction for morphometry analysis. Dendritic spine density in Grn+/- mice was comparable to wild-type littermates, but the apical dendrites in Grn+/- mice had a shift in the proportion of spine types, with fewer stubby spines and more thin spines. Additionally, apical dendrites of Grn+/- mice had longer spines and smaller thin spine head diameter in comparison to wild-type littermates. These changes in spine morphology may contribute to altered circuit-level activity and social dominance deficits in Grn+/- mice.
Collapse
Affiliation(s)
- Anna K Cook
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kelsey M Greathouse
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Phaedra N Manuel
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Noelle H Cooper
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Juliana M Eberhardt
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Cameron D Freeman
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Audrey J Weber
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jeremy H Herskowitz
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Andrew E Arrant
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
21
|
Liu J, Li Y, Liu Y, Yu R, Yin Y, Lai X, Xu B, Cao J. Elevated serum level of progranulin is associated with increased mortality in critically ill patients with candidemia. Microbes Infect 2024; 26:105302. [PMID: 38246573 DOI: 10.1016/j.micinf.2024.105302] [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: 10/03/2023] [Revised: 01/03/2024] [Accepted: 01/17/2024] [Indexed: 01/23/2024]
Abstract
Candidemia is a severe disease with high mortality in both intensive care unit (ICU) and non-ICU settings. Considering that progranulin (PGRN) is a potential therapeutic target for the candidemia caused by Candida albicans, we determined the serum level of PGRN after candidemia and evaluated its association with mortality. A retrospective discovery cohort (62 patients) and a validation cohort (70 patients) were enrolled. Blood was collected on day of first blood culture positivity for C. albicans, and serum PGRN levels were then measured. In the discovery cohort, all serum PGRN studied were expressed at higher levels in candidemia patients than in bacteremia patients and healthy volunteers, non-survivors presented with significantly higher serum PGRN concentrations when compared with survivors. Serum PGRN concentration was associated with 30-day mortality and patients at a higher risk of death showed higher serum PGRN levels. These results were confirmed in the independent validation cohort. Interestingly, in vitro study demonstrated that macrophages, neutrophils and lymphocytes may be the major source of PGRN production after C. albicans infection instead of epithelial cells. Our findings highlight that serum PGRN appears as a biomarker in candidemia patients and as a promising tool for mortality risk stratification in managing candidemia.
Collapse
Affiliation(s)
- Jiayu Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Yue Li
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuhan Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Renlin Yu
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yibing Yin
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Xiaofei Lai
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Banglao Xu
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China.
| | - Ju Cao
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
22
|
Astuti Y, Raymant M, Quaranta V, Clarke K, Abudula M, Smith O, Bellomo G, Chandran-Gorner V, Nourse C, Halloran C, Ghaneh P, Palmer D, Jones RP, Campbell F, Pollard JW, Morton JP, Mielgo A, Schmid MC. Efferocytosis reprograms the tumor microenvironment to promote pancreatic cancer liver metastasis. NATURE CANCER 2024; 5:774-790. [PMID: 38355776 PMCID: PMC11136665 DOI: 10.1038/s43018-024-00731-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 01/18/2024] [Indexed: 02/16/2024]
Abstract
Pancreatic ductal adenocarcinoma is a highly metastatic disease and macrophages support liver metastases. Efferocytosis, or engulfment of apoptotic cells by macrophages, is an essential process in tissue homeostasis and wound healing, but its role in metastasis is less well understood. Here, we found that the colonization of the hepatic metastatic site is accompanied by low-grade tissue injury and that efferocytosis-mediated clearance of parenchymal dead cells promotes macrophage reprogramming and liver metastasis. Mechanistically, progranulin expression in macrophages is necessary for efficient efferocytosis by controlling lysosomal acidification via cystic fibrosis transmembrane conductance regulator and the degradation of lysosomal cargo, resulting in LXRα/RXRα-mediated macrophage conversion and upregulation of arginase 1. Pharmacological blockade of efferocytosis or macrophage-specific genetic depletion of progranulin impairs macrophage conversion, improves CD8+ T cell functions, and reduces liver metastasis. Our findings reveal how hard-wired functions of macrophages in tissue repair contribute to liver metastasis and identify potential targets for prevention of pancreatic ductal adenocarcinoma liver metastasis.
Collapse
Affiliation(s)
- Yuliana Astuti
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Meirion Raymant
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Valeria Quaranta
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Kim Clarke
- Computational Biology Facility, University of Liverpool, Liverpool, UK
| | - Maidinaimu Abudula
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Olivia Smith
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Gaia Bellomo
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | | | - Craig Nourse
- Cancer Research UK Scotland Institute, Glasgow, UK
| | - Christopher Halloran
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Paula Ghaneh
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Daniel Palmer
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Robert P Jones
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Fiona Campbell
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | | | - Jennifer P Morton
- Cancer Research UK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Ainhoa Mielgo
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Michael C Schmid
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK.
| |
Collapse
|
23
|
Sevigny J, Uspenskaya O, Heckman LD, Wong LC, Hatch DA, Tewari A, Vandenberghe R, Irwin DJ, Saracino D, Le Ber I, Ahmed R, Rohrer JD, Boxer AL, Boland S, Sheehan P, Brandes A, Burstein SR, Shykind BM, Kamalakaran S, Daniels CW, David Litwack E, Mahoney E, Velaga J, McNamara I, Sondergaard P, Sajjad SA, Kobayashi YM, Abeliovich A, Hefti F. Progranulin AAV gene therapy for frontotemporal dementia: translational studies and phase 1/2 trial interim results. Nat Med 2024; 30:1406-1415. [PMID: 38745011 PMCID: PMC11108785 DOI: 10.1038/s41591-024-02973-0] [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: 07/03/2023] [Accepted: 04/03/2024] [Indexed: 05/16/2024]
Abstract
GRN mutations cause progranulin haploinsufficiency, which eventually leads to frontotemporal dementia (FTD-GRN). PR006 is an investigational gene therapy delivering the granulin gene (GRN) using an adeno-associated virus serotype 9 (AAV9) vector. In non-clinical studies, PR006 transduced neurons derived from induced pluripotent stem cells of patients with FTD-GRN, resulted in progranulin expression and improvement of lipofuscin, lysosomal and neuroinflammation pathologies in Grn-knockout mice, and was well tolerated except for minimal, asymptomatic dorsal root ganglionopathy in non-human primates. We initiated a first-in-human phase 1/2 open-label trial. Here we report results of a pre-specified interim analysis triggered with the last treated patient of the low-dose cohort (n = 6) reaching the 12-month follow-up timepoint. We also include preliminary data from the mid-dose cohort (n = 7). Primary endpoints were safety, immunogenicity and change in progranulin levels in cerebrospinal fluid (CSF) and blood. Secondary endpoints were Clinical Dementia Rating (CDR) plus National Alzheimer's Disease Coordinating Center (NACC) Frontotemporal Lobar Degeneration (FTLD) rating scale and levels of neurofilament light chain (NfL). One-time administration of PR006 into the cisterna magna was generally safe and well tolerated. All patients developed treatment-emergent anti-AAV9 antibodies in the CSF, but none developed anti-progranulin antibodies. CSF pleocytosis was the most common PR006-related adverse event. Twelve serious adverse events occurred, mostly unrelated to PR006. Deep vein thrombosis developed in three patients. There was one death (unrelated) occurring 18 months after treatment. CSF progranulin increased after PR006 treatment in all patients; blood progranulin increased in most patients but only transiently. NfL levels transiently increased after PR006 treatment, likely reflecting dorsal root ganglia toxicity. Progression rates, based on the CDR scale, were within the broad ranges reported for patients with FTD. These data provide preliminary insights into the safety and bioactivity of PR006. Longer follow-up and additional studies are needed to confirm the safety and potential efficacy of PR006. ClinicalTrials.gov identifier: NCT04408625 .
Collapse
Affiliation(s)
- Jeffrey Sevigny
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA.
| | - Olga Uspenskaya
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Laura Dean Heckman
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Li Chin Wong
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Daniel A Hatch
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Ambika Tewari
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Rik Vandenberghe
- Neurology Service, University Hospitals Leuven, Leuven, Belgium and Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - David J Irwin
- Department of Neurology, Penn Frontotemporal Degeneration Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Dario Saracino
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau, ICM, Inserm, CNRS UMR 7225 APHP - Hôpital Pitié-Salpêtrière, Paris, France
| | - Isabelle Le Ber
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau, ICM, Inserm, CNRS UMR 7225 APHP - Hôpital Pitié-Salpêtrière, Paris, France
| | - Rebekah Ahmed
- Department of Neurology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Jonathan D Rohrer
- Department of Neurodegenerative Disease, Dementia Research Center, UCL Queen Square Institute of Neurology, London, UK
| | - Adam L Boxer
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
| | - Sebastian Boland
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Patricia Sheehan
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Alissa Brandes
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Suzanne R Burstein
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Benjamin M Shykind
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Sitharthan Kamalakaran
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Carter W Daniels
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - E David Litwack
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Erin Mahoney
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Jenny Velaga
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Ilan McNamara
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Patricia Sondergaard
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Syed A Sajjad
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Yvonne M Kobayashi
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Asa Abeliovich
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Franz Hefti
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| |
Collapse
|
24
|
Swift IJ, Rademakers R, Finch N, Baker M, Ghidoni R, Benussi L, Binetti G, Rossi G, Synofzik M, Wilke C, Mengel D, Graff C, Takada LT, Sánchez-Valle R, Antonell A, Galimberti D, Fenoglio C, Serpente M, Arcaro M, Schreiber S, Vielhaber S, Arndt P, Santana I, Almeida MR, Moreno F, Barandiaran M, Gabilondo A, Stubert J, Gómez-Tortosa E, Agüero P, Sainz MJ, Gohda T, Murakoshi M, Kamei N, Kittel-Schneider S, Reif A, Weigl J, Jian J, Liu C, Serrero G, Greither T, Theil G, Lohmann E, Gazzina S, Bagnoli S, Coppola G, Bruni A, Quante M, Kiess W, Hiemisch A, Jurkutat A, Block MS, Carlson AM, Bråthen G, Sando SB, Grøntvedt GR, Lauridsen C, Heslegrave A, Heller C, Abel E, Gómez-Núñez A, Puey R, Arighi A, Rotondo E, Jiskoot LC, Meeter LHH, Durães J, Lima M, Tábuas-Pereira M, Lemos J, Boeve B, Petersen RC, Dickson DW, Graff-Radford NR, LeBer I, Sellami L, Lamari F, Clot F, Borroni B, Cantoni V, Rivolta J, Lleó A, Fortea J, Alcolea D, Illán-Gala I, Andres-Cerezo L, Van Damme P, Clarimon J, Steinacker P, Feneberg E, Otto M, van der Ende EL, van Swieten JC, Seelaar H, Zetterberg H, Sogorb-Esteve A, Rohrer JD. A systematic review of progranulin concentrations in biofluids in over 7,000 people-assessing the pathogenicity of GRN mutations and other influencing factors. Alzheimers Res Ther 2024; 16:66. [PMID: 38539243 PMCID: PMC10976725 DOI: 10.1186/s13195-024-01420-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 02/25/2024] [Indexed: 07/13/2024]
Abstract
BACKGROUND Pathogenic heterozygous mutations in the progranulin gene (GRN) are a key cause of frontotemporal dementia (FTD), leading to significantly reduced biofluid concentrations of the progranulin protein (PGRN). This has led to a number of ongoing therapeutic trials aiming to treat this form of FTD by increasing PGRN levels in mutation carriers. However, we currently lack a complete understanding of factors that affect PGRN levels and potential variation in measurement methods. Here, we aimed to address this gap in knowledge by systematically reviewing published literature on biofluid PGRN concentrations. METHODS Published data including biofluid PGRN concentration, age, sex, diagnosis and GRN mutation were collected for 7071 individuals from 75 publications. The majority of analyses (72%) had focused on plasma PGRN concentrations, with many of these (56%) measured with a single assay type (Adipogen) and so the influence of mutation type, age at onset, sex, and diagnosis were investigated in this subset of the data. RESULTS We established a plasma PGRN concentration cut-off between pathogenic mutation carriers and non-carriers of 74.8 ng/mL using the Adipogen assay based on 3301 individuals, with a CSF concentration cut-off of 3.43 ng/mL. Plasma PGRN concentration varied by GRN mutation type as well as by clinical diagnosis in those without a GRN mutation. Plasma PGRN concentration was significantly higher in women than men in GRN mutation carriers (p = 0.007) with a trend in non-carriers (p = 0.062), and there was a significant but weak positive correlation with age in both GRN mutation carriers and non-carriers. No significant association was seen with weight or with TMEM106B rs1990622 genotype. However, higher plasma PGRN levels were seen in those with the GRN rs5848 CC genotype in both GRN mutation carriers and non-carriers. CONCLUSIONS These results further support the usefulness of PGRN concentration for the identification of the large majority of pathogenic mutations in the GRN gene. Furthermore, these results highlight the importance of considering additional factors, such as mutation type, sex and age when interpreting PGRN concentrations. This will be particularly important as we enter the era of trials for progranulin-associated FTD.
Collapse
Grants
- 2019-02248 JPND GENFI-PROX grant
- 2019-02248 JPND GENFI-PROX grant
- 2019-02248 JPND GENFI-PROX grant
- 2019-02248 JPND GENFI-PROX grant
- 2019-02248 JPND GENFI-PROX grant
- 2019-02248 JPND GENFI-PROX grant
- MR/M008525/1 MRC Clinician Scientist Fellowship
- MR/M008525/1 MRC Clinician Scientist Fellowship
- 2013/017584 FAPESP grant number
- 01ED2008A EU Joint Programme-Neurodegenerative Diseases networks Genfi-Prox
- 01ED2001 bPride
- FTLDc 01GI1007A, Moodmarker 01EW200 German Federal Ministry of Education and Research
- MIRIADE 860197, FAIR-PARK II 633190 the EU
- SFB1279 German Research Foundation/DFG
- D.3830 The foundation of the state Baden-Württemberg
- D.5009 Boehringer Ingelheim Ulm University BioCenter and the Thierry Latran Foundation
- #ALFGBG-71320 Swedish State Support for Clinical Research
- #201809-2016862 Alzheimer Drug Discovery Foundation (ADDF), USA
- #ADSF-21-831376-C, #ADSF-21-831381-C, #ADSF-21-831377-C AD Strategic Fund and the Alzheimer's Association
- #2019-02397, #2022-01018 Swedish Research Council
- #FO2022-0270 the Bluefield Project, the Olav Thon Foundation, the Erling-Persson Family Foundation, Stiftelsen för Gamla Tjänarinnor, Hjärnfonden, Sweden
- JPND2021-00694) the European Union Joint Programme - Neurodegenerative Disease Research
- UKDRI-1003 UK Dementia Research Institute at UCL
- ARUK-RADF2021A-003 Alzheimer's Research UK
- BRC149/NS/MH NIHR Rare Disease Translational Research Collaboration
- MR/M023664/1 MRC UK GENFI grant
- P30 AG062677 NIA NIH HHS
- Alzheimer’s Association
- Clinician Scientist programme "PRECISE.net" funded by the Else Kröner-Fresenius-Stiftung
- the European Union’s Horizon 2020 research and innovation programme under the Marie Skłodowska-Curie grant agreement No 860197 (MIRIADE)
- the European Union Joint Programme – Neurodegenerative Disease Research
- National Institute for Health and Care Research University College London Hospitals Biomedical Research Centre
- Alzheimer’s Research UK
- Bluefield Project
Collapse
Affiliation(s)
- Imogen J Swift
- Department of Neurodegenerative Disease, Dementia Research Institute, UCL Institute of Neurology, Queen Square, London, UK
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - NiCole Finch
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Matt Baker
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Roberta Ghidoni
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni Di Dio Fatebenefratelli, Brescia, Italy
| | - Luisa Benussi
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni Di Dio Fatebenefratelli, Brescia, Italy
| | - Giuliano Binetti
- MAC-Memory Clinic and Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni Di Dio Fatebenefratelli, Brescia, Italy
| | - Giacomina Rossi
- Unit of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Matthis Synofzik
- Division Translational Genomics of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- German Center of Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Carlo Wilke
- Division Translational Genomics of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- German Center of Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - David Mengel
- Division Translational Genomics of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- German Center of Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Caroline Graff
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, BioclinicumKarolinska Institutet, Solna, Sweden
- Unit for Hereditary Dementias, Theme Inflammation and Aging, Karolinska University Hospital, Solna, Sweden
| | - Leonel T Takada
- Department of Neurology, Hospital das Clinicas, University of São Paulo Medical School, São Paulo, Brazil
| | - Raquel Sánchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, FRCB-IDIBAPS, Institut de Neurociències, Neurology Service, Hospital Clínic de Barcelona, Universitat de Barcelona (UB), 08036, Barcelona, Spain
| | - Anna Antonell
- Alzheimer's Disease and Other Cognitive Disorders Unit, FRCB-IDIBAPS, Institut de Neurociències, Neurology Service, Hospital Clínic de Barcelona, Universitat de Barcelona (UB), 08036, Barcelona, Spain
| | - Daniela Galimberti
- Dept. of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
- Neurodegerative Diseases Center, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Chiara Fenoglio
- Dept. of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
- Neurodegerative Diseases Center, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Maria Serpente
- Neurodegerative Diseases Center, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Marina Arcaro
- Neurodegerative Diseases Center, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefanie Schreiber
- Department of Neurology, Otto Von Guericke University, Magdeburg, Germany
| | - Stefan Vielhaber
- Department of Neurology, Otto Von Guericke University, Magdeburg, Germany
| | - Philipp Arndt
- Department of Neurology, Otto Von Guericke University, Magdeburg, Germany
| | - Isabel Santana
- Neurology Department, Centro Hospitalar E Universitário de Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology, Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | | | - Fermín Moreno
- Cognitive Disorders Unit, Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa, Spain
- Neuroscience Area, Biodonostia Health Research Insitute, San Sebastian, Gipuzkoa, Spain
| | - Myriam Barandiaran
- Cognitive Disorders Unit, Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa, Spain
- Neuroscience Area, Biodonostia Health Research Insitute, San Sebastian, Gipuzkoa, Spain
| | - Alazne Gabilondo
- Cognitive Disorders Unit, Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa, Spain
- Neuroscience Area, Biodonostia Health Research Insitute, San Sebastian, Gipuzkoa, Spain
| | - Johannes Stubert
- Department of Obstetrics and Gynecology, Rostock University Medical Center, Rostock, Germany
| | | | - Pablo Agüero
- Department of Neurology, Fundación Jiménez Díaz, Madrid, Spain
| | - M José Sainz
- Department of Neurology, Fundación Jiménez Díaz, Madrid, Spain
| | - Tomohito Gohda
- Department of Nephrology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Maki Murakoshi
- Department of Nephrology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Nozomu Kamei
- Department of Endocrinology and Metabolism, Hiroshima Red Cross Hospital & Atomicbomb Survivors Hospital, Hiroshima, Japan
- Institute for Clinical Research, National Hospital Organization, Kure Medical Center and Chugoku Cancer Center, Hiroshima, Japan
| | - Sarah Kittel-Schneider
- Department of Psychiatry, University College Cork, Cork, Ireland
- Department of Psychiatry, Psychotherapy and Psychosomatic Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Andreas Reif
- Department of Psychiatry, Psychotherapy and Psychosomatic Medicine, University Hospital Frankfurt, Frankfurt, Germany
| | - Johannes Weigl
- Department of Psychiatry, Psychotherapy and Psychosomatic Medicine, University Hospital Würzburg, Würzburg, Germany
- Department of Psychiatry, Hospital in Tauberbischofsheim, Tauberbischofsheim, Germany
| | - Jinlong Jian
- University of Pennsylvania, Gene Therapy Program, Philadelphia, USA
| | - Chuanju Liu
- Department of Orthopaedics & Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Ginette Serrero
- A&G Pharmaceutical Inc, Columbia, MD, USA
- Program in Oncology, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Thomas Greither
- Center for Reproductive Medicine and Andrology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Gerit Theil
- Department of Urology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Ebba Lohmann
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- DZNE, German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Stefano Gazzina
- Department of Neurological and Vision Sciences, Neurophysiology Unit, ASST SpedaliCivili, Brescia, Italy
| | - Silvia Bagnoli
- Department of Neurological and Psychiatric Sciences, University of Florence, Viale Morgagni, 85, 50134, Florence, Italy
| | - Giovanni Coppola
- Department of Neurology, University of California, Los Angeles, California, USA
- Department of Psychiatry Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, California, USA
| | - Amalia Bruni
- Regional Neurogenetic Centre, ASPCZ, Lamezia Terme, Italy
| | - Mirja Quante
- Department of Neonatology, Tuebingen University Hospital, Tuebingen, Germany
| | - Wieland Kiess
- Leipzig Research Center for Civilization Diseases - LIFE, University of Leipzig, Leipzig, Germany
- Hospital for Children and Adolescents, University of Leipzig, Leipzig, Germany
- Center of Pediatric Research (CPL), University of Leipzig, Leipzig, Germany
| | - Andreas Hiemisch
- Leipzig Research Center for Civilization Diseases - LIFE, University of Leipzig, Leipzig, Germany
- Center of Pediatric Research (CPL), University of Leipzig, Leipzig, Germany
| | - Anne Jurkutat
- Leipzig Research Center for Civilization Diseases - LIFE, University of Leipzig, Leipzig, Germany
| | | | - Aaron M Carlson
- Department of Neurology, University of Colorado Anschutz Medical Campus, Denver, CO, USA
| | - Geir Bråthen
- Department of Neurology and Clinical Neurophysiology, Trondheim University Hospital, Trondheim, Norway
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, NTNU. , Trondheim, Norway
| | - Sigrid Botne Sando
- Department of Neurology and Clinical Neurophysiology, Trondheim University Hospital, Trondheim, Norway
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, NTNU. , Trondheim, Norway
| | - Gøril Rolfseng Grøntvedt
- Department of Neurology and Clinical Neurophysiology, Trondheim University Hospital, Trondheim, Norway
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, NTNU. , Trondheim, Norway
| | - Camilla Lauridsen
- Department of Research, Trondheim University Hospital, Trondheim, Norway
| | - Amanda Heslegrave
- Department of Neurodegenerative Disease, Dementia Research Institute, UCL Institute of Neurology, Queen Square, London, UK
| | - Carolin Heller
- Department of Neurodegenerative Disease, Dementia Research Institute, UCL Institute of Neurology, Queen Square, London, UK
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Emily Abel
- Department of Neurodegenerative Disease, Dementia Research Institute, UCL Institute of Neurology, Queen Square, London, UK
| | - Alba Gómez-Núñez
- Alzheimer's Disease and Other Cognitive Disorders Unit, FRCB-IDIBAPS, Institut de Neurociències, Neurology Service, Hospital Clínic de Barcelona, Universitat de Barcelona (UB), 08036, Barcelona, Spain
| | - Roger Puey
- Alzheimer's Disease and Other Cognitive Disorders Unit, FRCB-IDIBAPS, Institut de Neurociències, Neurology Service, Hospital Clínic de Barcelona, Universitat de Barcelona (UB), 08036, Barcelona, Spain
| | - Andrea Arighi
- Neurodegerative Diseases Center, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Enmanuela Rotondo
- Neurodegerative Diseases Center, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Lize C Jiskoot
- Department of Neurology and Alzheimer Center Erasmus MC, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Lieke H H Meeter
- Department of Neurology and Alzheimer Center Erasmus MC, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - João Durães
- Neurology Department, Centro Hospitalar E Universitário de Coimbra, Coimbra, Portugal
| | - Marisa Lima
- Neurology Department, Centro Hospitalar E Universitário de Coimbra, Coimbra, Portugal
| | - Miguel Tábuas-Pereira
- Neurology Department, Centro Hospitalar E Universitário de Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - João Lemos
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Bradley Boeve
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Isabelle LeBer
- Sorbonne UniversitéInserm U1127, CNRS UMR 7225, Institut du Cerveau Et La Moelle Épinière (ICM), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France
| | - Leila Sellami
- Sorbonne UniversitéInserm U1127, CNRS UMR 7225, Institut du Cerveau Et La Moelle Épinière (ICM), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France
- Centre de Référence Des Démences Rares Ou Précoces, IM2A, Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France
| | - Foudil Lamari
- UF de Biochimie Des Maladies Neurométaboliques Et Neurodégénératives, Service de Biochimie Métabolique, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France
| | - Fabienne Clot
- UF de Neurogénétique Moléculaire Et Cellulaire, Département de Génétique, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière-Charles Foix, Paris, France
| | - Barbara Borroni
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Valentina Cantoni
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Jasmine Rivolta
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Alberto Lleó
- Neurology Department. Hospital Sant Pau, Memory Unit, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
- Autonomous University of Barcelona, 08913, Barcelona, Spain
| | - Juan Fortea
- Neurology Department. Hospital Sant Pau, Memory Unit, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
- Autonomous University of Barcelona, 08913, Barcelona, Spain
| | - Daniel Alcolea
- Neurology Department. Hospital Sant Pau, Memory Unit, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
- Autonomous University of Barcelona, 08913, Barcelona, Spain
| | - Ignacio Illán-Gala
- Neurology Department. Hospital Sant Pau, Memory Unit, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
- Autonomous University of Barcelona, 08913, Barcelona, Spain
| | - Lucie Andres-Cerezo
- Institute of Rheumatology, Na Slupi 4, 12850, Prague 2, Prague, Czech Republic
| | - Philip Van Damme
- Laboratory of Neurobiology, Flanders Interuniversity Institute for Biotechnology, Katholieke Universiteit Leuven, Campus Gasthuisberg, 3000, Louvain, Belgium
| | - Jordi Clarimon
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Petra Steinacker
- Department of Neurology, Martin-Luther University Halle-Wittenberg, University Clinic Halle, Halle (Saale), Germany
| | - Emily Feneberg
- Department of Neurology, Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
| | - Markus Otto
- Department of Neurology, Martin-Luther University Halle-Wittenberg, University Clinic Halle, Halle (Saale), Germany
| | - Emma L van der Ende
- Department of Neurology and Alzheimer Center Erasmus MC, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - John C van Swieten
- Department of Neurology and Alzheimer Center Erasmus MC, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Harro Seelaar
- Department of Neurology and Alzheimer Center Erasmus MC, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Henrik Zetterberg
- Department of Neurodegenerative Disease, Dementia Research Institute, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at University College London, UCL Queen Square Institute of Neurology, University College London, London, UK
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, 43180, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Aitana Sogorb-Esteve
- Department of Neurodegenerative Disease, Dementia Research Institute, UCL Institute of Neurology, Queen Square, London, UK
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Jonathan D Rohrer
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK.
| |
Collapse
|
25
|
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
|
26
|
Li M, Wang P, Huo ST, Qiu H, Li C, Lin S, Guo L, Ji Y, Zhu Y, Liu J, Guo J, Na J, Hu Y. Human Pluripotent Stem Cells Derived Endothelial Cells Repair Choroidal Ischemia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2302940. [PMID: 38115754 PMCID: PMC10916649 DOI: 10.1002/advs.202302940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 10/12/2023] [Indexed: 12/21/2023]
Abstract
Choroidal atrophy is a common fundus pathological change closely related to the development of age-related macular degeneration (AMD), retinitis pigmentosa, and pathological myopia. Studies suggest that choroidal endothelial cells (CECs) that form the choriocapillaris vessels are the first cells lost in choroidal atrophy. It is found that endothelial cells derived from human pluripotent stem cells (hPSC-ECs) through the MESP1+ mesodermal progenitor stage express CECs-specific markers and can integrate into choriocapillaris. Single-cell RNA-seq (scRNA-seq) studies show that hPSC-ECs upregulate angiogenesis and immune-modulatory and neural protective genes after interacting with ex vivo ischemic choroid. In a rat model of choroidal ischemia (CI), transplantation of hPSC-ECs into the suprachoroidal space increases choroid thickness and vasculature density. Close-up examination shows that engrafted hPSC-ECs integrate with all layers of rat choroidal vessels and last 90 days. Remarkably, EC transplantation improves the visual function of CI rats. The work demonstrates that hPSC-ECs can be used to repair choroidal ischemia in the animal model, which may lead to a new therapy to alleviate choroidal atrophy implicated in dry AMD, pathological myopia, and other ocular diseases.
Collapse
Affiliation(s)
- Mengda Li
- Eye CenterBeijing Tsinghua Changgung HospitalBeijing102218China
- Institute for Precision MedicineTsinghua UniversityBeijing100084China
- School of Clinical MedicineTsinghua UniversityBeijing100084China
| | - Peiliang Wang
- SXMU‐Tsinghua Collaborative Innovation Center for Frontier MedicineSchool of MedicineTsinghua UniversityBeijing100084China
- State Key Laboratory for Complex, Severe, and Rare DiseasesTsinghua UniversityBeijing100084China
- Center for Stem Cell Biology and Regenerative MedicineSchool of MedicineTsinghua UniversityBeijing100084China
| | - Si Tong Huo
- Eye CenterBeijing Tsinghua Changgung HospitalBeijing102218China
- Institute for Precision MedicineTsinghua UniversityBeijing100084China
- School of Clinical MedicineTsinghua UniversityBeijing100084China
| | - Hui Qiu
- SXMU‐Tsinghua Collaborative Innovation Center for Frontier MedicineSchool of MedicineTsinghua UniversityBeijing100084China
- State Key Laboratory for Complex, Severe, and Rare DiseasesTsinghua UniversityBeijing100084China
- Center for Stem Cell Biology and Regenerative MedicineSchool of MedicineTsinghua UniversityBeijing100084China
- School of Life SciencesTsinghua UniversityBeijing100084China
| | - Chendi Li
- Eye CenterBeijing Tsinghua Changgung HospitalBeijing102218China
- Institute for Precision MedicineTsinghua UniversityBeijing100084China
- School of Clinical MedicineTsinghua UniversityBeijing100084China
| | - Siyong Lin
- Eye CenterBeijing Tsinghua Changgung HospitalBeijing102218China
- Institute for Precision MedicineTsinghua UniversityBeijing100084China
- School of Clinical MedicineTsinghua UniversityBeijing100084China
| | - Libin Guo
- Eye CenterBeijing Tsinghua Changgung HospitalBeijing102218China
- Institute for Precision MedicineTsinghua UniversityBeijing100084China
- School of Clinical MedicineTsinghua UniversityBeijing100084China
| | - Yicong Ji
- Eye CenterBeijing Tsinghua Changgung HospitalBeijing102218China
- Institute for Precision MedicineTsinghua UniversityBeijing100084China
- School of Clinical MedicineTsinghua UniversityBeijing100084China
| | - Yonglin Zhu
- Center for Stem Cell Biology and Regenerative MedicineSchool of MedicineTsinghua UniversityBeijing100084China
| | - Jinyang Liu
- SXMU‐Tsinghua Collaborative Innovation Center for Frontier MedicineSchool of MedicineTsinghua UniversityBeijing100084China
- State Key Laboratory for Complex, Severe, and Rare DiseasesTsinghua UniversityBeijing100084China
- Center for Stem Cell Biology and Regenerative MedicineSchool of MedicineTsinghua UniversityBeijing100084China
| | - Jianying Guo
- Center for Reproductive MedicineDepartment of Obstetrics and GynaecologyPeking University Third HospitalBeijing100191China
| | - Jie Na
- SXMU‐Tsinghua Collaborative Innovation Center for Frontier MedicineSchool of MedicineTsinghua UniversityBeijing100084China
- State Key Laboratory for Complex, Severe, and Rare DiseasesTsinghua UniversityBeijing100084China
- Center for Stem Cell Biology and Regenerative MedicineSchool of MedicineTsinghua UniversityBeijing100084China
| | - Yuntao Hu
- Eye CenterBeijing Tsinghua Changgung HospitalBeijing102218China
- Institute for Precision MedicineTsinghua UniversityBeijing100084China
- School of Clinical MedicineTsinghua UniversityBeijing100084China
| |
Collapse
|
27
|
Takahashi H, Bhagwagar S, Nies SH, Ye H, Han X, Chiasseu MT, Wang G, Mackenzie IR, Strittmatter SM. Reduced progranulin increases tau and α-synuclein inclusions and alters mouse tauopathy phenotypes via glucocerebrosidase. Nat Commun 2024; 15:1434. [PMID: 38365772 PMCID: PMC10873339 DOI: 10.1038/s41467-024-45692-3] [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: 05/16/2022] [Accepted: 02/01/2024] [Indexed: 02/18/2024] Open
Abstract
Comorbid proteinopathies are observed in many neurodegenerative disorders including Alzheimer's disease (AD), increase with age, and influence clinical outcomes, yet the mechanisms remain ill-defined. Here, we show that reduction of progranulin (PGRN), a lysosomal protein associated with TDP-43 proteinopathy, also increases tau inclusions, causes concomitant accumulation of α-synuclein and worsens mortality and disinhibited behaviors in tauopathy mice. The increased inclusions paradoxically protect against spatial memory deficit and hippocampal neurodegeneration. PGRN reduction in male tauopathy attenuates activity of β-glucocerebrosidase (GCase), a protein previously associated with synucleinopathy, while increasing glucosylceramide (GlcCer)-positive tau inclusions. In neuronal culture, GCase inhibition enhances tau aggregation induced by AD-tau. Furthermore, purified GlcCer directly promotes tau aggregation in vitro. Neurofibrillary tangles in human tauopathies are also GlcCer-immunoreactive. Thus, in addition to TDP-43, PGRN regulates tau- and synucleinopathies via GCase and GlcCer. A lysosomal PGRN-GCase pathway may be a common therapeutic target for age-related comorbid proteinopathies.
Collapse
Affiliation(s)
- Hideyuki Takahashi
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT, 06536, USA
| | - Sanaea Bhagwagar
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT, 06536, USA
- College of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Sarah H Nies
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT, 06536, USA
- Graduate School of Cellular and Molecular Neuroscience, University of Tübingen, D-72074, Tübingen, Germany
| | - Hongping Ye
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center At San Antonio, San Antonio, TX, 78229, USA
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center At San Antonio, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center At San Antonio, San Antonio, TX, 78229, USA
| | - Marius T Chiasseu
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT, 06536, USA
| | - Guilin Wang
- Department of Molecular Biophysics and Biochemistry, School of Medicine, Yale University, New Haven, CT, 06520, USA
| | - Ian R Mackenzie
- Department of Pathology, University of British Columbia and Vancouver General Hospital, Vancouver, BC, Canada
| | - Stephen M Strittmatter
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT, 06536, USA.
| |
Collapse
|
28
|
Sung W, Noh MY, Nahm M, Kim YS, Ki CS, Kim YE, Kim HJ, Kim SH. Progranulin haploinsufficiency mediates cytoplasmic TDP-43 aggregation with lysosomal abnormalities in human microglia. J Neuroinflammation 2024; 21:47. [PMID: 38347588 PMCID: PMC10863104 DOI: 10.1186/s12974-024-03039-1] [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: 12/21/2023] [Accepted: 02/07/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND Progranulin (PGRN) haploinsufficiency due to progranulin gene (GRN) variants can cause frontotemporal dementia (FTD) with aberrant TAR DNA-binding protein 43 (TDP-43) accumulation. Despite microglial burden with TDP-43-related pathophysiology, direct microglial TDP-43 pathology has not been clarified yet, only emphasized in neuronal pathology. Thus, the objective of this study was to investigate TDP-43 pathology in microglia of patients with PGRN haploinsufficiency. METHODS To design a human microglial cell model with PGRN haploinsufficiency, monocyte-derived microglia (iMGs) were generated from FTD-GRN patients carrying pathogenic or likely pathogenic variants (p.M1? and p.W147*) and three healthy controls. RESULTS iMGs from FTD-GRN patients with PGRN deficiency exhibited severe neuroinflammation phenotype and failure to maintain their homeostatic molecular signatures, along with impaired phagocytosis. In FTD-GRN patients-derived iMGs, significant cytoplasmic TDP-43 aggregation and accumulation of lipid droplets with profound lysosomal abnormalities were observed. These pathomechanisms were mediated by complement C1q activation and upregulation of pro-inflammatory cytokines. CONCLUSIONS Our study provides considerable cellular and molecular evidence that loss-of-function variants of GRN in human microglia can cause microglial dysfunction with abnormal TDP-43 aggregation induced by inflammatory milieu as well as the impaired lysosome. Elucidating the role of microglial TDP-43 pathology in intensifying neuroinflammation in individuals with FTD due to PGRN deficiency and examining consequential effects on microglial dysfunction might yield novel insights into the mechanisms underlying FTD and neurodegenerative disorders.
Collapse
Affiliation(s)
- Wonjae Sung
- Department of Neurology, College of Medicine, Hanyang University, 222, Wangsimni-Ro, Seongdong-Gu, Seoul, 04763, Republic of Korea
| | - Min-Young Noh
- Department of Neurology, College of Medicine, Hanyang University, 222, Wangsimni-Ro, Seongdong-Gu, Seoul, 04763, Republic of Korea
| | - Minyeop Nahm
- Dementia Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Yong Sung Kim
- Department of Neurology, College of Medicine, Hanyang University, 222, Wangsimni-Ro, Seongdong-Gu, Seoul, 04763, Republic of Korea
| | | | - Young-Eun Kim
- Department of Laboratory Medicine, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Hee-Jin Kim
- Department of Neurology, College of Medicine, Hanyang University, 222, Wangsimni-Ro, Seongdong-Gu, Seoul, 04763, Republic of Korea
| | - Seung Hyun Kim
- Department of Neurology, College of Medicine, Hanyang University, 222, Wangsimni-Ro, Seongdong-Gu, Seoul, 04763, Republic of Korea.
| |
Collapse
|
29
|
Hsiao-Nakamoto J, Chiu CL, VandeVrede L, Ravi R, Vandenberg B, De Groot J, Tsogtbaatar B, Fang M, Auger P, Gould NS, Marchioni F, Powers CA, Davis SS, Suh JH, Alkabsh J, Heuer HW, Lago AL, Scearce-Levie K, Seeley WW, Boeve BF, Rosen HJ, Berger A, Tsai R, Di Paolo G, Boxer AL, Bhalla A, Huang F. Alterations in Lysosomal, Glial and Neurodegenerative Biomarkers in Patients with Sporadic and Genetic Forms of Frontotemporal Dementia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.09.579529. [PMID: 38405775 PMCID: PMC10888909 DOI: 10.1101/2024.02.09.579529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Background Frontotemporal dementia (FTD) is the most common cause of early-onset dementia with 10-20% of cases caused by mutations in one of three genes: GRN, C9orf72, or MAPT. To effectively develop therapeutics for FTD, the identification and characterization of biomarkers to understand disease pathogenesis and evaluate the impact of specific therapeutic strategies on the target biology as well as the underlying disease pathology are essential. Moreover, tracking the longitudinal changes of these biomarkers throughout disease progression is crucial to discern their correlation with clinical manifestations for potential prognostic usage. Methods We conducted a comprehensive investigation of biomarkers indicative of lysosomal biology, glial cell activation, synaptic and neuronal health in cerebrospinal fluid (CSF) and plasma from non-carrier controls, sporadic FTD (symptomatic non-carriers) and symptomatic carriers of mutations in GRN, C9orf72, or MAPT, as well as asymptomatic GRN mutation carriers. We also assessed the longitudinal changes of biomarkers in GRN mutation carriers. Furthermore, we examined biomarker levels in disease impacted brain regions including middle temporal gyrus (MTG) and superior frontal gyrus (SFG) and disease-unaffected inferior occipital gyrus (IOG) from sporadic FTD and symptomatic GRN carriers. Results We confirmed glucosylsphingosine (GlcSph), a lysosomal biomarker regulated by progranulin, was elevated in the plasma from GRN mutation carriers, both symptomatic and asymptomatic. GlcSph and other lysosomal biomarkers such as ganglioside GM2 and globoside GB3 were increased in the disease affected SFG and MTG regions from sporadic FTD and symptomatic GRN mutation carriers, but not in the IOG, compared to the same brain regions from controls. The glial biomarkers GFAP in plasma and YKL40 in CSF were elevated in asymptomatic GRN carriers, and all symptomatic groups, except the symptomatic C9orf72 mutation group. YKL40 was also increased in SFG and MTG regions from sporadic FTD and symptomatic GRN mutation carriers. Neuronal injury and degeneration biomarkers NfL in CSF and plasma, and UCHL1 in CSF were elevated in patients with all forms of FTD. Synaptic biomarkers NPTXR, NPTX1/2, and VGF were reduced in CSF from patients with all forms of FTD, with the most pronounced reductions observed in symptomatic MAPT mutation carriers. Furthermore, we demonstrated plasma NfL was significantly positively correlated with disease severity as measured by CDR+NACC FTLD SB in genetic forms of FTD and CSF NPTXR was significantly negatively correlated with CDR+NACC FTLD SB in symptomatic GRN and MAPT mutation carriers. Conclusions In conclusion, our comprehensive investigation replicated alterations in biofluid biomarkers indicative of lysosomal function, glial activation, synaptic and neuronal health across sporadic and genetic forms of FTD and unveiled novel insights into the dysregulation of these biomarkers within brain tissues from patients with GRN mutations. The observed correlations between biomarkers and disease severity open promising avenues for prognostic applications and for indicators of drug efficacy in clinical trials. Our data also implicated a complicated relationship between biofluid and tissue biomarker changes and future investigations should delve into the mechanistic underpinnings of these biomarkers, which will serve as a foundation for the development of targeted therapeutics for FTD.
Collapse
Affiliation(s)
- Jennifer Hsiao-Nakamoto
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
- These authors contributed equally
| | - Chi-Lu Chiu
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
- These authors contributed equally
| | - Lawren VandeVrede
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Ritesh Ravi
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
| | - Brittany Vandenberg
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
- Present address: Brittany Vandenberg, Washington State University, Pullman, WA 99164, USA
| | - Jack De Groot
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
- Present address: Jack DeGroot: Prime Medicine Inc., Cambridge, MA 02139, USA
| | | | - Meng Fang
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
| | - Paul Auger
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
- Present address: Paul Auger: Nurix Therapeutics, San Francisco, CA 94158, USA
| | - Neal S Gould
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
| | - Filippo Marchioni
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
| | - Casey A Powers
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
- Present address: Casey A. Powers: Stanford University, Stanford, CA 94305, USA
| | - Sonnet S Davis
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
| | - Jung H Suh
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
| | - Jamal Alkabsh
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
| | - Hilary W Heuer
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Argentina Lario Lago
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Kimberly Scearce-Levie
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
- Present address: Kimberly Scearce-Levie: Cajal Neuroscience, Seattle, WA 98109, USA
| | - William W Seeley
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Bradley F Boeve
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Howard J Rosen
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Amy Berger
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
| | - Richard Tsai
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
| | - Gilbert Di Paolo
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
| | - Adam L Boxer
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, CA, 94158, USA
- These authors contributed equally
| | - Akhil Bhalla
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
- These authors contributed equally
| | - Fen Huang
- Denali Therapeutics Inc., 161 Oyster Point, South San Francisco, CA, 94080, USA
- These authors contributed equally
| |
Collapse
|
30
|
Smith DM, Aggarwal G, Niehoff ML, Jones SA, Banerjee S, Farr SA, Nguyen AD. Biochemical, biomarker, and behavioral characterization of the GrnR493X mouse model of frontotemporal dementia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.27.542495. [PMID: 37398305 PMCID: PMC10312473 DOI: 10.1101/2023.05.27.542495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Heterozygous loss-of-function mutations in the progranulin gene (GRN) are a major cause of frontotemporal dementia due to progranulin haploinsufficiency; complete deficiency of progranulin causes neuronal ceroid lipofuscinosis. Several progranulin-deficient mouse models have been generated, including both knockout mice and knockin mice harboring a common patient mutation (R493X). However, the GrnR493X mouse model has not been characterized completely. Additionally, while homozygous GrnR493X and Grn knockout mice have been extensively studied, data from heterozygous mice is still limited. Here, we performed more in-depth characterization of heterozygous and homozygous GrnR493X knockin mice, which includes biochemical assessments, behavioral studies, and analysis of fluid biomarkers. In the brains of homozygous GrnR493X mice, we found increased phosphorylated TDP-43 along with increased expression of lysosomal genes, markers of microgliosis and astrogliosis, pro-inflammatory cytokines, and complement factors. Heterozygous GrnR493X mice did not have increased TDP-43 phosphorylation but did exhibit limited increases in lysosomal and inflammatory gene expression. Behavioral studies found social and emotional deficits in GrnR493X mice that mirror those observed in Grn knockout mouse models, as well as impairment in memory and executive function. Overall, the GrnR493X knockin mouse model closely phenocopies Grn knockout models. Lastly, in contrast to homozygous knockin mice, heterozygous GrnR493X mice do not have elevated levels of fluid biomarkers previously identified in humans, including neurofilament light chain (NfL) and glial fibrillary acidic protein (GFAP) in both plasma and CSF. These results may help to inform pre-clinical studies that use this Grn knockin mouse model and other Grn knockout models.
Collapse
Affiliation(s)
- Denise M. Smith
- Saint Louis University School of Medicine, Department of Internal Medicine, Division of Geriatric Medicine, United States of America
- Saint Louis University School of Medicine, Department of Pharmacology and Physiology, United States of America
- Saint Louis University, Institute for Translational Neuroscience, United States of America
| | - Geetika Aggarwal
- Saint Louis University School of Medicine, Department of Internal Medicine, Division of Geriatric Medicine, United States of America
- Saint Louis University School of Medicine, Department of Pharmacology and Physiology, United States of America
- Saint Louis University, Institute for Translational Neuroscience, United States of America
| | - Michael L. Niehoff
- Saint Louis University School of Medicine, Department of Internal Medicine, Division of Geriatric Medicine, United States of America
- Veterans Affairs Medical Center, United States of America
| | - Spencer A. Jones
- Saint Louis University School of Medicine, Department of Internal Medicine, Division of Geriatric Medicine, United States of America
- Saint Louis University School of Medicine, Department of Pharmacology and Physiology, United States of America
- Saint Louis University, Institute for Translational Neuroscience, United States of America
| | - Subhashis Banerjee
- Saint Louis University School of Medicine, Department of Internal Medicine, Division of Geriatric Medicine, United States of America
- Saint Louis University School of Medicine, Department of Pharmacology and Physiology, United States of America
- Saint Louis University, Institute for Translational Neuroscience, United States of America
| | - Susan A. Farr
- Saint Louis University School of Medicine, Department of Internal Medicine, Division of Geriatric Medicine, United States of America
- Saint Louis University School of Medicine, Department of Pharmacology and Physiology, United States of America
- Saint Louis University, Institute for Translational Neuroscience, United States of America
- Veterans Affairs Medical Center, United States of America
| | - Andrew D. Nguyen
- Saint Louis University School of Medicine, Department of Internal Medicine, Division of Geriatric Medicine, United States of America
- Saint Louis University School of Medicine, Department of Pharmacology and Physiology, United States of America
- Saint Louis University, Institute for Translational Neuroscience, United States of America
| |
Collapse
|
31
|
Wang AL, Mambou EA, Kao AW. The progranulin cleavage product granulin 3 exerts a dominant negative effect on animal fitness. Hum Mol Genet 2024; 33:245-253. [PMID: 37903062 PMCID: PMC10800025 DOI: 10.1093/hmg/ddad184] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/12/2023] [Accepted: 10/24/2023] [Indexed: 11/01/2023] Open
Abstract
Progranulin is an evolutionarily conserved protein that has been implicated in human neurodevelopmental and neurodegenerative diseases. Human progranulin is comprised of multiple cysteine-rich, biologically active granulin peptides. Granulin peptides accumulate with age and stress, however their functional contributions relative to full-length progranulin remain unclear. To address this, we generated C. elegans strains that produced quantifiable levels of both full-length progranulin/PGRN-1 protein and cleaved granulin peptide. Using these strains, we demonstrated that even in the presence of intact PGRN-1, granulin peptides suppressed the activity of the lysosomal aspartyl protease activity, ASP-3/CTSD. Granulin peptides were also dominant over PGRN-1 in compromising animal fitness as measured by progress through development and stress response. Finally, the degradation of human TDP-43 was impaired when the granulin to PGRN-1 ratio was increased, representing a disease-relevant downstream impact of impaired lysosomal function. In summary, these studies suggest that not only absolute progranulin levels, but also the balance between full-length progranulin and its cleavage products, is important in regulating lysosomal biology. Given its relevance in human disease, this suggests that the processing of progranulin into granulins should be considered as part of disease pathobiology and may represent a site of therapeutic intervention.
Collapse
Affiliation(s)
- Austin L Wang
- Memory and Aging Center, Weill Institute for Neuroscience, Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| | - Edwina A Mambou
- Memory and Aging Center, Weill Institute for Neuroscience, Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| | - Aimee W Kao
- Memory and Aging Center, Weill Institute for Neuroscience, Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
- Bakar Aging Research Institute, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
32
|
Bai Y, Camargo CM, Glasauer SMK, Gifford R, Tian X, Longhini AP, Kosik KS. Single-cell mapping of lipid metabolites using an infrared probe in human-derived model systems. Nat Commun 2024; 15:350. [PMID: 38191490 PMCID: PMC10774263 DOI: 10.1038/s41467-023-44675-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 12/20/2023] [Indexed: 01/10/2024] Open
Abstract
Understanding metabolic heterogeneity is the key to uncovering the underlying mechanisms of metabolic-related diseases. Current metabolic imaging studies suffer from limitations including low resolution and specificity, and the model systems utilized often lack human relevance. Here, we present a single-cell metabolic imaging platform to enable direct imaging of lipid metabolism with high specificity in various human-derived 2D and 3D culture systems. Through the incorporation of an azide-tagged infrared probe, selective detection of newly synthesized lipids in cells and tissue became possible, while simultaneous fluorescence imaging enabled cell-type identification in complex tissues. In proof-of-concept experiments, newly synthesized lipids were directly visualized in human-relevant model systems among different cell types, mutation status, differentiation stages, and over time. We identified upregulated lipid metabolism in progranulin-knockdown human induced pluripotent stem cells and in their differentiated microglia cells. Furthermore, we observed that neurons in brain organoids exhibited a significantly lower lipid metabolism compared to astrocytes.
Collapse
Affiliation(s)
- Yeran Bai
- Neuroscience Research Institute, Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA, USA.
- Photothermal Spectroscopy Corp., Santa Barbara, CA, USA.
| | - Carolina M Camargo
- Neuroscience Research Institute, Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA, USA
| | - Stella M K Glasauer
- Neuroscience Research Institute, Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA, USA
| | - Raymond Gifford
- Neuroscience Research Institute, Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA, USA
| | - Xinran Tian
- Neuroscience Research Institute, Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA, USA
| | - Andrew P Longhini
- Neuroscience Research Institute, Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA, USA
| | - Kenneth S Kosik
- Neuroscience Research Institute, Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA, USA.
| |
Collapse
|
33
|
Doyle JJ, Parker JA. Genetic Interactions of Progranulin Across the ALS-FTD Spectrum and Beyond. MICROPUBLICATION BIOLOGY 2023; 2023:10.17912/micropub.biology.000304. [PMID: 38188422 PMCID: PMC10767572 DOI: 10.17912/micropub.biology.000304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 08/08/2021] [Accepted: 12/19/2023] [Indexed: 01/09/2024]
Abstract
Progranulin (PGRN) is a growth factor in which mutations are one of the leading causes of frontotemporal dementia (FTD), and has been implicated in an assortment of neurodegenerative diseases. Conversely, higher levels of the protein have shown potential as a general neuronal protective factor. While examining its neuroprotective applications on a broader scale would be unfeasible in mammalian models, we turned to the nematode C. elegans to map the interactions of PGRN across multiple genetic models of neurodegenerative diseases. Our results indicate that while the overexpression of PGRN appears to be protective across all models tested, the loss of PGRN exacerbated the disease phenotypes of all but three of the models tested. Given the ease of genetic analysis in nematodes, we propose this model organism as an efficient tool to build a comprehensive map of PGRN's genetic interactions.
Collapse
Affiliation(s)
- James J. Doyle
- Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
- Metabolic Disorders and Complications, RI-MUHC, Montreal, McGill, Canada
| | - J Alex Parker
- CRCHUM and Department of Neuroscience, University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|
34
|
McElhanon KE, Huff TC, Hirenallur-Shanthappa D, Miller RA, Christoforou N. Increased circulating progranulin is not sufficient to induce cardiac dysfunction or supraventricular arrhythmia. Sci Rep 2023; 13:21541. [PMID: 38057339 PMCID: PMC10700350 DOI: 10.1038/s41598-023-47311-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 11/12/2023] [Indexed: 12/08/2023] Open
Abstract
Atrial fibrillation (AF) is the most prevalent cardiac arrhythmia, and the incidence of new-onset AF has been increasing over the past two decades. Several factors contribute to the risk of developing AF including age, preexisting cardiovascular disease, chronic kidney disease, and obesity. Concurrent with the rise in AF, obesity has followed the same two-decade trend. The contribution of circulating proteins to obesity-related AF is of particular interest in the field. In this study, we investigated the effects of increased circulating levels of the glycoprotein progranulin on the development of supraventricular arrhythmias and changes to cardiac function. AAV8-mediated overexpression of full-length mouse progranulin was used to increase plasma protein levels and determine susceptibility to supraventricular arrhythmias and changes in cardiac structure and function. C57Bl/6N mice were subjected to increased circulating levels of progranulin for 20 weeks. Cardiac conduction was evaluated by surface ECG with and without isoproterenol challenge, and cardiac structure and function were measured by echocardiography after 20 weeks of circulating progranulin overexpression. Increased circulating levels of progranulin were maintained throughout the 20-week study. The cardiac structure and function remained unchanged in mice with increased circulating progranulin. ECG indices (P wave duration, P amplitude, QRS interval) were unaffected by increased progranulin levels and no arrhythmogenic events were observed following the isoproterenol challenge. In our model, increased levels of circulating progranulin were not sufficient to induce changes in cardiac structure and function or elicit ECG abnormalities suggestive of susceptibility to supraventricular arrhythmias.
Collapse
Affiliation(s)
- Kevin E McElhanon
- Rare Disease Research Unit, Worldwide Research, Development, and Medical, Pfizer, Inc., Cambridge, MA, USA
| | - Tyler C Huff
- Rare Disease Research Unit, Worldwide Research, Development, and Medical, Pfizer, Inc., Cambridge, MA, USA
| | | | - Russell A Miller
- Rare Disease Research Unit, Worldwide Research, Development, and Medical, Pfizer, Inc., Cambridge, MA, USA
| | - Nicolas Christoforou
- Rare Disease Research Unit, Worldwide Research, Development, and Medical, Pfizer, Inc., Cambridge, MA, USA.
| |
Collapse
|
35
|
Aggarwal G, Banerjee S, Jones SA, Benchaar Y, Bélanger J, Sévigny M, Smith DM, Niehoff ML, Pavlack M, de Vera IMS, Petkau TL, Leavitt BR, Ling K, Jafar-Nejad P, Rigo F, Morley JE, Farr SA, Dutchak PA, Sephton CF, Nguyen AD. Antisense oligonucleotides targeting the miR-29b binding site in the GRN mRNA increase progranulin translation. J Biol Chem 2023; 299:105475. [PMID: 37981208 PMCID: PMC10755782 DOI: 10.1016/j.jbc.2023.105475] [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: 09/18/2023] [Revised: 10/13/2023] [Accepted: 11/07/2023] [Indexed: 11/21/2023] Open
Abstract
Heterozygous GRN (progranulin) mutations cause frontotemporal dementia (FTD) due to haploinsufficiency, and increasing progranulin levels is a major therapeutic goal. Several microRNAs, including miR-29b, negatively regulate progranulin protein levels. Antisense oligonucleotides (ASOs) are emerging as a promising therapeutic modality for neurological diseases, but strategies for increasing target protein levels are limited. Here, we tested the efficacy of ASOs as enhancers of progranulin expression by sterically blocking the miR-29b binding site in the 3' UTR of the human GRN mRNA. We found 16 ASOs that increase progranulin protein in a dose-dependent manner in neuroglioma cells. A subset of these ASOs also increased progranulin protein in iPSC-derived neurons and in a humanized GRN mouse model. In FRET-based assays, the ASOs effectively competed for miR-29b from binding to the GRN 3' UTR RNA. The ASOs increased levels of newly synthesized progranulin protein by increasing its translation, as revealed by polysome profiling. Together, our results demonstrate that ASOs can be used to effectively increase target protein levels by partially blocking miR binding sites. This ASO strategy may be therapeutically feasible for progranulin-deficient FTD as well as other conditions of haploinsufficiency.
Collapse
Affiliation(s)
- Geetika Aggarwal
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St Louis, Missouri, USA; Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St Louis, Missouri, USA; Institute for Translational Neuroscience, Saint Louis University, St Louis, Missouri, USA
| | - Subhashis Banerjee
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St Louis, Missouri, USA; Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St Louis, Missouri, USA; Institute for Translational Neuroscience, Saint Louis University, St Louis, Missouri, USA
| | - Spencer A Jones
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St Louis, Missouri, USA; Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St Louis, Missouri, USA; Institute for Translational Neuroscience, Saint Louis University, St Louis, Missouri, USA
| | - Yousri Benchaar
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Jasmine Bélanger
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Myriam Sévigny
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Denise M Smith
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St Louis, Missouri, USA; Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St Louis, Missouri, USA; Institute for Translational Neuroscience, Saint Louis University, St Louis, Missouri, USA
| | - Michael L Niehoff
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St Louis, Missouri, USA; Veterans Affairs Medical Center, St Louis, Missouri, USA
| | - Monica Pavlack
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St Louis, Missouri, USA; Institute for Translational Neuroscience, Saint Louis University, St Louis, Missouri, USA
| | - Ian Mitchelle S de Vera
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St Louis, Missouri, USA; Institute for Translational Neuroscience, Saint Louis University, St Louis, Missouri, USA
| | - Terri L Petkau
- Department of Medical Genetics, Centre for Molecular Medicine & Therapeutics, B.C. Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Blair R Leavitt
- Department of Medical Genetics, Centre for Molecular Medicine & Therapeutics, B.C. Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada; Division of Neurology, Department of Medicine, University of British Columbia Hospital, Vancouver, British Columbia, Canada; Center for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Karen Ling
- Ionis Pharmaceuticals, Carlsbad, California, USA
| | | | - Frank Rigo
- Ionis Pharmaceuticals, Carlsbad, California, USA
| | - John E Morley
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St Louis, Missouri, USA
| | - Susan A Farr
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St Louis, Missouri, USA; Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St Louis, Missouri, USA; Institute for Translational Neuroscience, Saint Louis University, St Louis, Missouri, USA; Veterans Affairs Medical Center, St Louis, Missouri, USA
| | - Paul A Dutchak
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Chantelle F Sephton
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Andrew D Nguyen
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St Louis, Missouri, USA; Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St Louis, Missouri, USA; Institute for Translational Neuroscience, Saint Louis University, St Louis, Missouri, USA.
| |
Collapse
|
36
|
Thomasen PB, Salasova A, Kjaer-Sorensen K, Woloszczuková L, Lavický J, Login H, Tranberg-Jensen J, Almeida S, Beel S, Kavková M, Qvist P, Kjolby M, Ovesen PL, Nolte S, Vestergaard B, Udrea AC, Nejsum LN, Chao MV, Van Damme P, Krivanek J, Dasen J, Oxvig C, Nykjaer A. SorCS2 binds progranulin to regulate motor neuron development. Cell Rep 2023; 42:113333. [PMID: 37897724 DOI: 10.1016/j.celrep.2023.113333] [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: 04/28/2022] [Revised: 07/25/2023] [Accepted: 10/09/2023] [Indexed: 10/30/2023] Open
Abstract
Motor neuron (MN) development and nerve regeneration requires orchestrated action of a vast number of molecules. Here, we identify SorCS2 as a progranulin (PGRN) receptor that is required for MN diversification and axon outgrowth in zebrafish and mice. In zebrafish, SorCS2 knockdown also affects neuromuscular junction morphology and fish motility. In mice, SorCS2 and PGRN are co-expressed by newborn MNs from embryonic day 9.5 until adulthood. Using cell-fate tracing and nerve segmentation, we find that SorCS2 deficiency perturbs cell-fate decisions of brachial MNs accompanied by innervation deficits of posterior nerves. Additionally, adult SorCS2 knockout mice display slower motor nerve regeneration. Interestingly, primitive macrophages express high levels of PGRN, and their interaction with SorCS2-positive motor axon is required during axon pathfinding. We further show that SorCS2 binds PGRN to control its secretion, signaling, and conversion into granulins. We propose that PGRN-SorCS2 signaling controls MN development and regeneration in vertebrates.
Collapse
Affiliation(s)
- Pernille Bogetofte Thomasen
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Alena Salasova
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark.
| | - Kasper Kjaer-Sorensen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Lucie Woloszczuková
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Josef Lavický
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
| | - Hande Login
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Jeppe Tranberg-Jensen
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Sergio Almeida
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Sander Beel
- Department of Neurology and Department of Neurosciences, KU Leuven and Center for Brain & Disease Research VIB, 3000 Leuven, Belgium
| | - Michaela Kavková
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
| | - Per Qvist
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Mads Kjolby
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Peter Lund Ovesen
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Stella Nolte
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Benedicte Vestergaard
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Andreea-Cornelia Udrea
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | | | - Moses V Chao
- Department of Neuroscience and Physiology, NYU Langone Health, New York, NY 10016, USA
| | - Philip Van Damme
- Department of Neurology and Department of Neurosciences, KU Leuven and Center for Brain & Disease Research VIB, 3000 Leuven, Belgium
| | - Jan Krivanek
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
| | - Jeremy Dasen
- Department of Neuroscience and Physiology, NYU Langone Health, New York, NY 10016, USA
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Anders Nykjaer
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark.
| |
Collapse
|
37
|
Hasan S, Fernandopulle MS, Humble SW, Frankenfield AM, Li H, Prestil R, Johnson KR, Ryan BJ, Wade-Martins R, Ward ME, Hao L. Multi-modal proteomic characterization of lysosomal function and proteostasis in progranulin-deficient neurons. Mol Neurodegener 2023; 18:87. [PMID: 37974165 PMCID: PMC10655356 DOI: 10.1186/s13024-023-00673-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/25/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Progranulin (PGRN) is a lysosomal glycoprotein implicated in various neurodegenerative diseases, including frontotemporal dementia and neuronal ceroid lipofuscinosis. Over 70 mutations discovered in the GRN gene all result in reduced expression of the PGRN protein. Genetic and functional studies point toward a regulatory role for PGRN in lysosome functions. However, the detailed molecular function of PGRN within lysosomes and the impact of PGRN deficiency on lysosomes remain unclear. METHODS We developed multifaceted proteomic techniques to characterize the dynamic lysosomal biology in living human neurons and fixed mouse brain tissues. Using lysosome proximity labeling and immuno-purification of intact lysosomes, we characterized lysosome compositions and interactome in both human induced pluripotent stem cell (iPSC)-derived glutamatergic neurons (i3Neurons) and mouse brains. Using dynamic stable isotope labeling by amino acids in cell culture (dSILAC) proteomics, we measured global protein half-lives in human i3Neurons for the first time. RESULTS Leveraging the multi-modal proteomics and live-cell imaging techniques, we comprehensively characterized how PGRN deficiency changes the molecular and functional landscape of neuronal lysosomes. We found that PGRN loss impairs the lysosome's degradative capacity with increased levels of v-ATPase subunits on the lysosome membrane, increased hydrolases within the lysosome, altered protein regulations related to lysosomal transport, and elevated lysosomal pH. Consistent with impairments in lysosomal function, GRN-null i3Neurons and frontotemporal dementia patient-derived i3Neurons carrying GRN mutation showed pronounced alterations in protein turnover, such as cathepsins and proteins related to supramolecular polymerization and inherited neurodegenerative diseases. CONCLUSION This study suggested PGRN as a critical regulator of lysosomal pH and degradative capacity, which influences global proteostasis in neurons. Beyond the study of progranulin deficiency, these newly developed proteomic methods in neurons and brain tissues provided useful tools and data resources for the field to study the highly dynamic neuronal lysosome biology.
Collapse
Affiliation(s)
- Saadia Hasan
- National Institute of Neurological, Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, USA
- Department of Neurodegenerative Disease, UK Dementia Research Institute, Institute of Neurology, University College London, London, UK
- Augusta University, University of Georgia Medical Partnership, Athens, GA, USA
| | - Michael S Fernandopulle
- National Institute of Neurological, Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, USA
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
- Medical Scientist Training Program, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Stewart W Humble
- National Institute of Neurological, Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, USA
- Department of Physiology, Anatomy and Genetics, Oxford Parkinson's Disease Centre, Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford, OX1 3QU, UK
| | | | - Haorong Li
- Department of Chemistry, George Washington University, Washington, DC, USA
| | - Ryan Prestil
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Kory R Johnson
- National Institute of Neurological, Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Brent J Ryan
- Department of Physiology, Anatomy and Genetics, Oxford Parkinson's Disease Centre, Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford, OX1 3QU, UK
| | - Richard Wade-Martins
- Department of Physiology, Anatomy and Genetics, Oxford Parkinson's Disease Centre, Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford, OX1 3QU, UK
| | - Michael E Ward
- National Institute of Neurological, Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, USA.
| | - Ling Hao
- Department of Chemistry, George Washington University, Washington, DC, USA.
| |
Collapse
|
38
|
Boylan MA, Pincetic A, Romano G, Tatton N, Kenkare-Mitra S, Rosenthal A. Targeting Progranulin as an Immuno-Neurology Therapeutic Approach. Int J Mol Sci 2023; 24:15946. [PMID: 37958929 PMCID: PMC10647331 DOI: 10.3390/ijms242115946] [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: 09/19/2023] [Revised: 10/25/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
Immuno-neurology is an emerging therapeutic strategy for dementia and neurodegeneration designed to address immune surveillance failure in the brain. Microglia, as central nervous system (CNS)-resident myeloid cells, routinely perform surveillance of the brain and support neuronal function. Loss-of-function (LOF) mutations causing decreased levels of progranulin (PGRN), an immune regulatory protein, lead to dysfunctional microglia and are associated with multiple neurodegenerative diseases, including frontotemporal dementia caused by the progranulin gene (GRN) mutation (FTD-GRN), Alzheimer's disease (AD), Parkinson's disease (PD), limbic-predominant age-related transactivation response deoxyribonucleic acid binding protein 43 (TDP-43) encephalopathy (LATE), and amyotrophic lateral sclerosis (ALS). Immuno-neurology targets immune checkpoint-like proteins, offering the potential to convert aging and dysfunctional microglia into disease-fighting cells that counteract multiple disease pathologies, clear misfolded proteins and debris, promote myelin and synapse repair, optimize neuronal function, support astrocytes and oligodendrocytes, and maintain brain vasculature. Several clinical trials are underway to elevate PGRN levels as one strategy to modulate the function of microglia and counteract neurodegenerative changes associated with various disease states. If successful, these and other immuno-neurology drugs have the potential to revolutionize the treatment of neurodegenerative disorders by harnessing the brain's immune system and shifting it from an inflammatory/pathological state to an enhanced physiological/homeostatic state.
Collapse
Affiliation(s)
| | | | | | | | | | - Arnon Rosenthal
- Alector, Inc., 131 Oyster Point Blvd, Suite 600, South San Francisco, CA 94080, USA
| |
Collapse
|
39
|
Niu H, Wang BY, Wei XY, Wang YN, Zhu WH, Li WJ, Zhang Y, Wang JC. Anti-inflammatory therapeutic biomarkers identified of human bone marrow mesenchymal stem cell therapy on aging mice by serum proteomics and peptidomics study. J Proteomics 2023; 288:104979. [PMID: 37524227 DOI: 10.1016/j.jprot.2023.104979] [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: 02/10/2023] [Revised: 07/13/2023] [Accepted: 07/21/2023] [Indexed: 08/02/2023]
Abstract
Aging is accompanied by deterioration in physical condition, and creates high risks of diseases. Stem cell therapy exhibited promising potential in delaying aging. However, the unelucidated therapeutic mechanism limits future clinical application. Herein, to systematically understand the response to stem cell transfusion at the molecular level, we performed quantitative serum proteomic and peptidomics analyses in the 24-month-old aging mice model with or without mesenchymal stem cell (MSC) treatment. As a result, a total of 560 proteins and 2131 endogenous peptides were identified, among which, 6 proteins and 9 endogenous peptides derived from 6 precursor proteins were finally identified as therapeutic biomarkers after MSC transfusion on aging mice both by untargeted label-free quantification and targeted parallel reaction monitoring (PRM) quantification. Amazingly, the biological function of these differential proteins was mainly related to inflammation, which is not only the important hallmark of aging, but also the main cause of inducing aging. The reduction of these inflammatory protein content after MSC treatment further suggests the anti-inflammatory effect of MSC therapy reported elsewhere. Therefore, our study provides new evidence for the anti-inflammatory effect of MSC therapy for anti-aging and offers abundant data to support deeper investigations of the therapeutic mechanism of MSC in delaying aging.
Collapse
Affiliation(s)
- Huan Niu
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China; Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, China
| | - Bo-Yan Wang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Xiao-Yue Wei
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yan-Nan Wang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Wen-Hui Zhu
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Wei-Jie Li
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Ying Zhang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China; Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, China.
| | - Jian-Cheng Wang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China; Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China.
| |
Collapse
|
40
|
Vyas M, Deschenes NM, Osmon KJL, Chen Z, Ahmad I, Kot S, Thompson P, Richmond C, Gray SJ, Walia JS. Efficacy of Adeno-Associated Virus Serotype 9-Mediated Gene Therapy for AB-Variant GM2 Gangliosidosis. Int J Mol Sci 2023; 24:14611. [PMID: 37834060 PMCID: PMC10572999 DOI: 10.3390/ijms241914611] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 09/06/2023] [Accepted: 09/16/2023] [Indexed: 10/15/2023] Open
Abstract
GM2 gangliosidoses are a group of neurodegenerative lysosomal storage disorders that are characterized by the accumulation of GM2 gangliosides (GM2), leading to rapid neurological decline and death. The hydrolysis of GM2 requires the specific synthesis, processing, and combination of products of three genes-HEXA, HEXB, and GM2A-within the cell's lysosomes. Mutations in these genes result in Tay-Sachs disease, Sandhoff disease, or AB-variant GM2 gangliosidosis (ABGM2), respectively. ABGM2, the rarest of the three types, is characterized by a mutation in the GM2A gene, which encodes the GM2 activator (GM2A) protein. Being a monogenic disease, gene therapy is a plausible and likely effective method of treatment for ABGM2. This study aimed at assessing the effects of administering a one-time intravenous treatment of single-stranded Adeno-associated virus serotype 9 (ssAAV9)-GM2A viral vector at a dose of 1 × 1014 vector genomes (vg) per kilogram per mouse in an ABGM2 mouse model (Gm2a-/-). ssAAV9-GM2A was administered at 1-day (neonatal) or 6-weeks of age (adult-stage). The results demonstrated that, in comparison to Gm2a-/- mice that received a vehicle injection, the treated mice had reduced GM2 accumulation within the central nervous system and had long-term persistence of vector genomes in the brain and liver. This proof-of-concept study is a step forward towards the development of a clinically therapeutic approach for the treatment of patients with ABGM2.
Collapse
Affiliation(s)
- Meera Vyas
- Centre for Neuroscience Studies, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Natalie M. Deschenes
- Centre for Neuroscience Studies, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Karlaina J. L. Osmon
- Centre for Neuroscience Studies, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Zhilin Chen
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (Z.C.)
| | - Imtiaz Ahmad
- Centre for Neuroscience Studies, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Shalini Kot
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (Z.C.)
| | - Patrick Thompson
- Department of Pediatrics, Queen’s University, Kingston, ON K7L 2V7, Canada;
| | - Chris Richmond
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (Z.C.)
| | - Steven J. Gray
- Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jagdeep S. Walia
- Centre for Neuroscience Studies, Queen’s University, Kingston, ON K7L 3N6, Canada
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (Z.C.)
- Department of Pediatrics, Queen’s University, Kingston, ON K7L 2V7, Canada;
| |
Collapse
|
41
|
He Y, Kaya I, Shariatgorji R, Lundkvist J, Wahlberg LU, Nilsson A, Mamula D, Kehr J, Zareba-Paslawska J, Biverstål H, Chergui K, Zhang X, Andren PE, Svenningsson P. Prosaposin maintains lipid homeostasis in dopamine neurons and counteracts experimental parkinsonism in rodents. Nat Commun 2023; 14:5804. [PMID: 37726325 PMCID: PMC10509278 DOI: 10.1038/s41467-023-41539-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 09/08/2023] [Indexed: 09/21/2023] Open
Abstract
Prosaposin (PSAP) modulates glycosphingolipid metabolism and variants have been linked to Parkinson's disease (PD). Here, we find altered PSAP levels in the plasma, CSF and post-mortem brain of PD patients. Altered plasma and CSF PSAP levels correlate with PD-related motor impairments. Dopaminergic PSAP-deficient (cPSAPDAT) mice display hypolocomotion and depression/anxiety-like symptoms with mildly impaired dopaminergic neurotransmission, while serotonergic PSAP-deficient (cPSAPSERT) mice behave normally. Spatial lipidomics revealed an accumulation of highly unsaturated and shortened lipids and reduction of sphingolipids throughout the brains of cPSAPDAT mice. The overexpression of α-synuclein via AAV lead to more severe dopaminergic degeneration and higher p-Ser129 α-synuclein levels in cPSAPDAT mice compared to WT mice. Overexpression of PSAP via AAV and encapsulated cell biodelivery protected against 6-OHDA and α-synuclein toxicity in wild-type rodents. Thus, these findings suggest PSAP may maintain dopaminergic lipid homeostasis, which is dysregulated in PD, and counteract experimental parkinsonism.
Collapse
Affiliation(s)
- Yachao He
- Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Ibrahim Kaya
- Department of Pharmaceutical Biosciences, Medical Mass Spectrometry Imaging, Uppsala University, Uppsala, Sweden
| | - Reza Shariatgorji
- Department of Pharmaceutical Biosciences, Medical Mass Spectrometry Imaging, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Spatial Mass Spectrometry, Uppsala University, Uppsala, Sweden
| | - Johan Lundkvist
- Division of Neurogeriatrics, Department of Neurobiology, Care Science and Society, Karolinska Institutet, Stockholm, Sweden
- Sinfonia Biotherapeutics AB, Huddinge, Sweden
| | - Lars U Wahlberg
- Division of Neurogeriatrics, Department of Neurobiology, Care Science and Society, Karolinska Institutet, Stockholm, Sweden
| | - Anna Nilsson
- Department of Pharmaceutical Biosciences, Medical Mass Spectrometry Imaging, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Spatial Mass Spectrometry, Uppsala University, Uppsala, Sweden
| | - Dejan Mamula
- Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Jan Kehr
- Section of Pharmacological Neurochemistry, Department of Physiology and Pharmacology, Karolinska Institute, Solna, Sweden
| | - Justyna Zareba-Paslawska
- Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Henrik Biverstål
- Sinfonia Biotherapeutics AB, Huddinge, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Karima Chergui
- Laboratory of Molecular Neurophysiology, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Xiaoqun Zhang
- Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Per E Andren
- Department of Pharmaceutical Biosciences, Medical Mass Spectrometry Imaging, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Spatial Mass Spectrometry, Uppsala University, Uppsala, Sweden
| | - Per Svenningsson
- Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| |
Collapse
|
42
|
Grossman M, Seeley WW, Boxer AL, Hillis AE, Knopman DS, Ljubenov PA, Miller B, Piguet O, Rademakers R, Whitwell JL, Zetterberg H, van Swieten JC. Frontotemporal lobar degeneration. Nat Rev Dis Primers 2023; 9:40. [PMID: 37563165 DOI: 10.1038/s41572-023-00447-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/12/2023] [Indexed: 08/12/2023]
Abstract
Frontotemporal lobar degeneration (FTLD) is one of the most common causes of early-onset dementia and presents with early social-emotional-behavioural and/or language changes that can be accompanied by a pyramidal or extrapyramidal motor disorder. About 20-25% of individuals with FTLD are estimated to carry a mutation associated with a specific FTLD pathology. The discovery of these mutations has led to important advances in potentially disease-modifying treatments that aim to slow progression or delay disease onset and has improved understanding of brain functioning. In both mutation carriers and those with sporadic disease, the most common underlying diagnoses are linked to neuronal and glial inclusions containing tau (FTLD-tau) or TDP-43 (FTLD-TDP), although 5-10% of patients may have inclusions containing proteins from the FUS-Ewing sarcoma-TAF15 family (FTLD-FET). Biomarkers definitively identifying specific pathological entities in sporadic disease have been elusive, which has impeded development of disease-modifying treatments. Nevertheless, disease-monitoring biofluid and imaging biomarkers are becoming increasingly sophisticated and are likely to serve as useful measures of treatment response during trials of disease-modifying treatments. Symptomatic trials using novel approaches such as transcranial direct current stimulation are also beginning to show promise.
Collapse
Affiliation(s)
- Murray Grossman
- Department of Neurology and Penn Frontotemporal Degeneration Center, University of Pennsylvania, Philadelphia, PA, USA
| | - William W Seeley
- Departments of Neurology and Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA.
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA.
| | - Adam L Boxer
- Departments of Neurology and Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
| | - Argye E Hillis
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | | | - Peter A Ljubenov
- Departments of Neurology and Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
| | - Bruce Miller
- Departments of Neurology and Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
| | - Olivier Piguet
- School of Psychology and Brain and Mind Center, University of Sydney, Sydney, New South Wales, Australia
| | - Rosa Rademakers
- VIB Center for Molecular Neurology, University of Antwerp, Antwerp, Belgium
| | | | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The University of Gothenburg, Mölndal, Sweden
- 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
| | | |
Collapse
|
43
|
Feng T, Minevich G, Liu P, Qin HX, Wozniak G, Pham J, Pham K, Korgaonkar A, Kurnellas M, Defranoux NA, Long H, Mitra A, Hu F. AAV- GRN partially corrects motor deficits and ALS/FTLD-related pathology in Tmem106b-/-Grn-/- mice. iScience 2023; 26:107247. [PMID: 37519899 PMCID: PMC10371829 DOI: 10.1016/j.isci.2023.107247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/18/2023] [Accepted: 06/26/2023] [Indexed: 08/01/2023] Open
Abstract
Loss of function of progranulin (PGRN), encoded by the granulin (GRN) gene, is implicated in several neurodegenerative diseases. Several therapeutics to boost PGRN levels are currently in clinical trials. However, it is difficult to test the efficacy of PGRN-enhancing drugs in mouse models due to the mild phenotypes of Grn-/- mice. Recently, mice deficient in both PGRN and TMEM106B were shown to develop severe motor deficits and pathology. Here, we show that intracerebral ventricle injection of PGRN-expressing AAV1/9 viruses partially rescues motor deficits, neuronal loss, glial activation, and lysosomal abnormalities in Tmem106b-/-Grn-/- mice. Widespread expression of PGRN is detected in both the brain and spinal cord for both AAV subtypes. However, AAV9 but not AAV1-mediated expression of PGRN results in high levels of PGRN in the serum. Together, these data support using the Tmem106b-/-Grn-/- mouse strain as a robust mouse model to determine the efficacy of PGRN-elevating therapeutics.
Collapse
Affiliation(s)
- Tuancheng Feng
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | | | - Pengan Liu
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | - Henry Xin Qin
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | | | - Jenny Pham
- Alector Inc, South San Francisco, CA 94080, USA
| | - Khanh Pham
- Alector Inc, South San Francisco, CA 94080, USA
| | | | | | | | - Hua Long
- Alector Inc, South San Francisco, CA 94080, USA
| | | | - Fenghua Hu
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
44
|
Anderson A, Tansey MG. Loss of Progranulin Results in Increased Pan-Cathepsin Activity and Reduced LAMP1 Lysosomal Protein. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.15.549151. [PMID: 37503267 PMCID: PMC10369980 DOI: 10.1101/2023.07.15.549151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Mutations in the progranulin (PGRN) encoding gene, GRN, cause familial frontotemporal dementia (FTD) and neuronal ceroid lipofuscinosis (NCL) and PGRN is also implicated in Parkinson's disease (PD). These mutations result in decreased PGRN expression. PGRN is highly expressed in peripheral immune cells and microglia and regulates cell growth, survival, repair, and inflammation. When PGRN is lost, the lysosome becomes dysfunctional, but the exact mechanism by which PGRN plays a role in lysosome function and how this contributes to inflammation and degeneration is not entirely understood. To better understand the role of PGRN in regulating lysosome function, this study examined how loss of GRN impacts total LAMP1 protein expression and cathepsin activities. Using mouse embryonic fibroblasts (MEFs), immunocytochemistry and immunoblotting assays were performed to analyze fluorescent signal from LAMP1 (lysosomal marker) and BMV109 (marker for pan-cathepsin activity). GRN-/- MEFs exhibit increased expression of pan-cathepsin activity relative to GRN+/+ MEFs, and significantly impacts expression of LAMP1. The significant increase in pan-cathepsin activity in the GRN-/- MEFs confirms that PGRN loss does alter cathepsin expression, which may be a result of compensatory mechanisms happening within the cell. Using NTAP PGRN added to GRN-/- MEFs, specific cathepsin activity is rescued. Further investigations should include assessing LAMP1 and BMV109 expression in microglia from GRN-/- mice, in the hopes of understanding the role of PGRN in lysosomal function in immune cells of the central nervous system and the diseases in which it is implicated.
Collapse
Affiliation(s)
- Abigail Anderson
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610
| | - Malú G Tansey
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610
| |
Collapse
|
45
|
Phillips B, Western D, Wang L, Timsina J, Sun Y, Gorijala P, Yang C, Do A, Nykänen NP, Alvarez I, Aguilar M, Pastor P, Morris JC, Schindler SE, Fagan AM, Puerta R, García-González P, de Rojas I, Marquié M, Boada M, Ruiz A, Perlmutter JS, Ibanez L, Perrin RJ, Sung YJ, Cruchaga C. Proteome wide association studies of LRRK2 variants identify novel causal and druggable proteins for Parkinson's disease. NPJ Parkinsons Dis 2023; 9:107. [PMID: 37422510 PMCID: PMC10329646 DOI: 10.1038/s41531-023-00555-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 06/29/2023] [Indexed: 07/10/2023] Open
Abstract
Common and rare variants in the LRRK2 locus are associated with Parkinson's disease (PD) risk, but the downstream effects of these variants on protein levels remain unknown. We performed comprehensive proteogenomic analyses using the largest aptamer-based CSF proteomics study to date (7006 aptamers (6138 unique proteins) in 3107 individuals). The dataset comprised six different and independent cohorts (five using the SomaScan7K (ADNI, DIAN, MAP, Barcelona-1 (Pau), and Fundació ACE (Ruiz)) and the PPMI cohort using the SomaScan5K panel). We identified eleven independent SNPs in the LRRK2 locus associated with the levels of 25 proteins as well as PD risk. Of these, only eleven proteins have been previously associated with PD risk (e.g., GRN or GPNMB). Proteome-wide association study (PWAS) analyses suggested that the levels of ten of those proteins were genetically correlated with PD risk, and seven were validated in the PPMI cohort. Mendelian randomization analyses identified GPNMB, LCT, and CD68 causal for PD and nominate one more (ITGB2). These 25 proteins were enriched for microglia-specific proteins and trafficking pathways (both lysosome and intracellular). This study not only demonstrates that protein phenome-wide association studies (PheWAS) and trans-protein quantitative trail loci (pQTL) analyses are powerful for identifying novel protein interactions in an unbiased manner, but also that LRRK2 is linked with the regulation of PD-associated proteins that are enriched in microglial cells and specific lysosomal pathways.
Collapse
Affiliation(s)
- Bridget Phillips
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Daniel Western
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Lihua Wang
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jigyasha Timsina
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Yichen Sun
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Priyanka Gorijala
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Chengran Yang
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Anh Do
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Division of Biostatistics, Washington University, St. Louis, MO, 63110, USA
| | - Niko-Petteri Nykänen
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Ignacio Alvarez
- Memory Disorders Unit, Department of Neurology, University Hospital Mutua Terrassa, Terrassa, Spain
| | - Miquel Aguilar
- Memory Disorders Unit, Department of Neurology, University Hospital Mutua Terrassa, Terrassa, Spain
| | - Pau Pastor
- Unit of Neurodegenerative diseases, Department of Neurology, University Hospital Germans Trias i Pujol and The Germans Trias i Pujol Research Institute (IGTP) Badalona, Barcelona, Spain
| | - John C Morris
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Suzanne E Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Anne M Fagan
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Raquel Puerta
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Pablo García-González
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Itziar de Rojas
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Marquié
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Mercè Boada
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Agustin Ruiz
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Joel S Perlmutter
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Laura Ibanez
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Richard J Perrin
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Yun Ju Sung
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Division of Biostatistics, Washington University, St. Louis, MO, 63110, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
46
|
Dedert C, Salih L, Xu F. Progranulin Protects against Hyperglycemia-Induced Neuronal Dysfunction through GSK3β Signaling. Cells 2023; 12:1803. [PMID: 37443837 PMCID: PMC10340575 DOI: 10.3390/cells12131803] [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: 06/02/2023] [Revised: 06/20/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
Type II diabetes affects over 530 million individuals worldwide and contributes to a host of neurological pathologies. Uncontrolled high blood glucose (hyperglycemia) is a major factor in diabetic pathology, and glucose regulation is a common goal for maintenance in patients. We have found that the neuronal growth factor progranulin protects against hyperglycemic stress in neurons, and although its mechanism of action is uncertain, our findings identified Glycogen Synthase Kinase 3β (GSK3β) as being potentially involved in its effects. In this study, we treated mouse primary cortical neurons exposed to high-glucose conditions with progranulin and a selective pharmacological inhibitor of GSK3β before assessing neuronal health and function. Whole-cell and mitochondrial viability were both improved by progranulin under high-glucose stress in a GSK3β-dependent manner. This extended to autophagy flux, indicated by the expressions of autophagosome marker Light Chain 3B (LC3B) and lysosome marker Lysosome-Associated Membrane Protein 2A (LAMP2A), which were affected by progranulin and showed heterogeneous changes from GSK3β inhibition. Lastly, GSK3β inhibition attenuated downstream calcium signaling and neuronal firing effects due to acute progranulin treatment. These data indicate that GSK3β plays an important role in progranulin's neuroprotective effects under hyperglycemic stress and serves as a jumping-off point to explore progranulin's protective capabilities in other neurodegenerative models.
Collapse
Affiliation(s)
- Cass Dedert
- Department of Biology, College of Arts and Sciences, Saint Louis University, Saint Louis, MO 63103, USA; (C.D.); (L.S.)
- Institute for Translational Neuroscience, Saint Louis University, Saint Louis, MO 63103, USA
| | - Lyuba Salih
- Department of Biology, College of Arts and Sciences, Saint Louis University, Saint Louis, MO 63103, USA; (C.D.); (L.S.)
- Institute for Translational Neuroscience, Saint Louis University, Saint Louis, MO 63103, USA
| | - Fenglian Xu
- Department of Biology, College of Arts and Sciences, Saint Louis University, Saint Louis, MO 63103, USA; (C.D.); (L.S.)
- Institute for Translational Neuroscience, Saint Louis University, Saint Louis, MO 63103, USA
- Department of Pharmacology and Physiology, School of Medicine, Saint Louis University, Saint Louis, MO 63103, USA
- Department of Biomedical Engineering, School of Science and Engineering, Saint Louis University, Saint Louis, MO 63103, USA
| |
Collapse
|
47
|
Wainberg M, Andrews SJ, Tripathy SJ. Shared genetic risk loci between Alzheimer's disease and related dementias, Parkinson's disease, and amyotrophic lateral sclerosis. Alzheimers Res Ther 2023; 15:113. [PMID: 37328865 PMCID: PMC10273745 DOI: 10.1186/s13195-023-01244-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 05/16/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Genome-wide association studies (GWAS) have indicated moderate genetic overlap between Alzheimer's disease (AD) and related dementias (ADRD), Parkinson's disease (PD) and amyotrophic lateral sclerosis (ALS), neurodegenerative disorders traditionally considered etiologically distinct. However, the specific genetic variants and loci underlying this overlap remain almost entirely unknown. METHODS We leveraged state-of-the-art GWAS for ADRD, PD, and ALS. For each pair of disorders, we examined each of the GWAS hits for one disorder and tested whether they were also significant for the other disorder, applying Bonferroni correction for the number of variants tested. This approach rigorously controls the family-wise error rate for both disorders, analogously to genome-wide significance. RESULTS Eleven loci with GWAS hits for one disorder were also associated with one or both of the other disorders: one with all three disorders (the MAPT/KANSL1 locus), five with ADRD and PD (near LCORL, CLU, SETD1A/KAT8, WWOX, and GRN), three with ADRD and ALS (near GPX3, HS3ST5/HDAC2/MARCKS, and TSPOAP1), and two with PD and ALS (near GAK/TMEM175 and NEK1). Two of these loci (LCORL and NEK1) were associated with an increased risk of one disorder but decreased risk of another. Colocalization analysis supported a shared causal variant between ADRD and PD at the CLU, WWOX, and LCORL loci, between ADRD and ALS at the TSPOAP1 locus, and between PD and ALS at the NEK1 and GAK/TMEM175 loci. To address the concern that ADRD is an imperfect proxy for AD and that the ADRD and PD GWAS have overlapping participants (nearly all of which are from the UK Biobank), we confirmed that all our ADRD associations had nearly identical odds ratios in an AD GWAS that excluded the UK Biobank, and all but one remained nominally significant (p < 0.05) for AD. CONCLUSIONS In one of the most comprehensive investigations to date of pleiotropy between neurodegenerative disorders, we identify eleven genetic risk loci shared among ADRD, PD, and ALS. These loci support lysosomal/autophagic dysfunction (GAK/TMEM175, GRN, KANSL1), neuroinflammation/immunity (TSPOAP1), oxidative stress (GPX3, KANSL1), and the DNA damage response (NEK1) as transdiagnostic processes underlying multiple neurodegenerative disorders.
Collapse
Affiliation(s)
- Michael Wainberg
- Centre for Addiction and Mental Health, 250 College Street, Toronto, M5T 1R8, Canada
| | - Shea J Andrews
- Department of Psychiatry & Behavioral Sciences, University of California San Francisco, San Francisco, 94143, USA
| | - Shreejoy J Tripathy
- Centre for Addiction and Mental Health, 250 College Street, Toronto, M5T 1R8, Canada.
- Institute of Medical Sciences, University of Toronto, Toronto, M5S 1A8, Canada.
- Department of Psychiatry, University of Toronto, Toronto, M5T 1R8, Canada.
- Department of Physiology, University of Toronto, Toronto, M5S 1A8, Canada.
| |
Collapse
|
48
|
Liu Y, Zhao X, Jian J, Hasan S, Liu C. Interaction with ERp57 is required for progranulin protection against Type 2 Gaucher disease. Biosci Trends 2023; 17:126-135. [PMID: 36889696 PMCID: PMC10514708 DOI: 10.5582/bst.2023.01022] [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] [Indexed: 03/10/2023]
Abstract
Gaucher disease (GD), one of the most common lysosomal storage diseases, is caused by GBA1 mutations resulting in defective glucocerebrosidase (GCase) and consequent accumulation of its substrates β-glucosylceramide (β-GlcCer). We reported progranulin (PGRN), a secretary growth factor-like molecule and an intracellular lysosomal protein was a crucial co-factor of GCase. PGRN binds to GCase and recruits Heat Shock Protein 70 (Hsp70) to GCase through its C-terminal Granulin (Grn) E domain, termed as ND7. In addition, both PGRN and ND7 are therapeutic against GD. Herein we found that both PGRN and its derived ND7 still displayed significant protective effects against GD in Hsp70 deficient cells. To delineate the molecular mechanisms underlying PGRN's Hsp70-independent regulation of GD, we performed a biochemical co-purification and mass spectrometry with His-tagged PGRN and His-tagged ND7 in Hsp70 deficient cells, which led to the identification of ERp57, also referred to as protein disulfide isomerase A3 (PDIA3), as a protein that binds to both PGRN and ND7. Within type 2 neuropathic GD patient fibroblasts L444P, bearing GBA1 L444P mutation, deletion of ERp57 largely abolished the therapeutic effects of PGRN and ND7, as manifested by loss of effects on lysosomal storage, GCase activity, and β-GlcCer accumulation. Additionally, recombinant ERp57 effectively restored the therapeutic effects of PGRN and ND7 in ERp57 knockout L444P fibroblasts. Collectively, this study reports ERp57 as a previously unrecognized binding partner of PGRN that contributes to PGRN regulation of GD.
Collapse
Affiliation(s)
- Yuzhao Liu
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, New York, USA
- Department of Endocrinology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiangli Zhao
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, New York, USA
| | - Jinlong Jian
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, New York, USA
| | - Sadaf Hasan
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, New York, USA
| | - Chuanju Liu
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, New York, USA
- Department of Cell Biology, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
49
|
Boxy P, Nykjær A, Kisiswa L. Building better brains: the pleiotropic function of neurotrophic factors in postnatal cerebellar development. Front Mol Neurosci 2023; 16:1181397. [PMID: 37251644 PMCID: PMC10213292 DOI: 10.3389/fnmol.2023.1181397] [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: 03/07/2023] [Accepted: 04/26/2023] [Indexed: 05/31/2023] Open
Abstract
The cerebellum is a multifunctional brain region that controls diverse motor and non-motor behaviors. As a result, impairments in the cerebellar architecture and circuitry lead to a vast array of neuropsychiatric and neurodevelopmental disorders. Neurotrophins and neurotrophic growth factors play essential roles in the development as well as maintenance of the central and peripheral nervous system which is crucial for normal brain function. Their timely expression throughout embryonic and postnatal stages is important for promoting growth and survival of both neurons and glial cells. During postnatal development, the cerebellum undergoes changes in its cellular organization, which is regulated by a variety of molecular factors, including neurotrophic factors. Studies have shown that these factors and their receptors promote proper formation of the cerebellar cytoarchitecture as well as maintenance of the cerebellar circuits. In this review, we will summarize what is known on the neurotrophic factors' role in cerebellar postnatal development and how their dysregulation assists in developing various neurological disorders. Understanding the expression patterns and signaling mechanisms of these factors and their receptors is crucial for elucidating their function within the cerebellum and for developing therapeutic strategies for cerebellar-related disorders.
Collapse
Affiliation(s)
- Pia Boxy
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience (DANDRITE)–Nordic EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
- The Danish National Research Foundation Center, PROMEMO, Aarhus University, Aarhus, Denmark
| | - Anders Nykjær
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience (DANDRITE)–Nordic EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
- The Danish National Research Foundation Center, PROMEMO, Aarhus University, Aarhus, Denmark
| | - Lilian Kisiswa
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience (DANDRITE)–Nordic EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
- The Danish National Research Foundation Center, PROMEMO, Aarhus University, Aarhus, Denmark
| |
Collapse
|
50
|
Lin Y, Yu N, Lin X, Deng X, Liu F, Tao H, Dong R, Wang B, Bi Y. Preoperative cerebrospinal fluid biomarkers may be associated with postoperative delirium in patients undergoing knee/hip arthroplasty: the PNDABLE study. BMC Geriatr 2023; 23:282. [PMID: 37165310 PMCID: PMC10173592 DOI: 10.1186/s12877-023-03943-w] [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: 12/07/2021] [Accepted: 03/30/2023] [Indexed: 05/12/2023] Open
Abstract
BACKGROUND In the global aging population, the incidence of postoperative delirium (POD) is increasing. Therefore, finding its effective predictive tools becomes crucial. We aimed to identify potential Cerebrospinal fluid (CSF)biomarkers for POD. METHODS A total of 825 patients undergoing knee/hip arthroplasty under combined spinal-epidural anesthesia were selected. The patients were aged 40 to 90 years with American Society of Anesthesiologists physical status I~II. The Mini-Mental State Examination was completed 1 day before the operation. CSF was extracted after successful spinal-epidural combined puncture, and α-synuclein (α-syn), amyloid beta40 (Aβ40), amyloid beta42 (Aβ42), t-Tau, phosphorylated Tau (p-Tau), progranulin (PGRN) and soluble triggering receptor expressed on myeloid cells 2 (sTREM2) in the CSF were measured by enzyme-linked immunosorbent assays (ELISA). The patient's operation time, anesthesia time, intraoperative blood loss and fluid input were also recorded. After the operation, the occurrence rate and severity of POD were determined by the Confusion Assessment Method and the Memorial Delirium Assessment Scale (MDAS), respectively. Patients were categorized into POD group and non-POD group. Logistic regression analysis was performed on the indicators with statistically significant differences, and the area under the ROC curve (AUC) was used to estimate the predictive accuracy of the biomarkers for POD. RESULTS A total of 92 patients developed POD and the incidence of POD was 11.15%. The results of the multivariable logistic regression showed that CSF t-Tau (P = 0.004, OR = 1.006, 95%CI 1.002~1.009) and α-syn (P = 0.004, OR = 1.001, 95%CI 1.000~1.001) were positively associated with the occurrence rate of POD, while Aβ42 (P < 0.001, OR = 0.989, 95%CI 0.986~0.993), CSF PGRN (P = 0.002, OR = 0.999, 95%CI 0.999~1.000), Aβ42/ t-Tau (P < 0.001, OR = 0.181, 95%CI 0.102~0.319) and Aβ42/p-Tau (P < 0.001, OR = 0.617, 95%CI 0.526~0.725) were inversely proportional to the occurrence of POD. ROC curve analysis indicated that Aβ42/t-Tau (AUC = 0.823), CSF Aβ42 (AUC = 0.813), Aβ42/p-Tau (AUC = 0.810), α-syn (AUC = 0.644) and PGRN (AUC = 0.638) could predict the occurrence rate of POD. The combination of all these biomarkers showed a greater AUC(0.896) than using any of them alone. CONCLUSIONS CSF Aβ42, PGRN, α-syn, Aβ42/t-Tau and Aβ42/p-Tau might be associated with the occurrence rate of POD in patients undergoing knee/hip arthroplasty. TRIAL REGISTRATION Clinical Registration No. ChiCTR2000033439.
Collapse
Affiliation(s)
- Yanan Lin
- Department of Anesthesiology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, Shandong province, China
| | - Nannan Yu
- Cadre Health Department, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, Shandong province, China
| | - Xu Lin
- Department of Anesthesiology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, Shandong province, China
| | - Xiyuan Deng
- Department of Anesthesiology, Dalian Municipal Central Hospital, Dalian University of Technology, Dalian, Liaoning province, China
| | - Fanghao Liu
- Department of Anesthesiology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, Shandong province, China
| | - He Tao
- Department of Anesthesiology, Dalian Municipal Central Hospital, Dalian University of Technology, Dalian, Liaoning province, China
| | - Rui Dong
- Department of Anesthesiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Bin Wang
- Department of Anesthesiology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, Shandong province, China.
| | - Yanlin Bi
- Department of Anesthesiology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, Shandong province, China.
| |
Collapse
|