1
|
Cosma C, Talli I, Pangrazzi E, Padoan A, Cerutti H, Zaninotto M, Gabelli C, Plebani M. Progranulin measurement with a new automated method: a step forward in the diagnostic approach to neurodegenerative disorders. Clin Chem Lab Med 2025; 63:1118-1123. [PMID: 39819746 DOI: 10.1515/cclm-2024-1143] [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: 10/01/2024] [Accepted: 01/06/2025] [Indexed: 01/19/2025]
Abstract
OBJECTIVES Mutations in the GRN gene encoded glycoprotein progranulin (PGRN), cause 5-10 % of all cases of frontotemporal lobar degeneration (FTLD). The aim of our study was to verify the analytical and clinical performance of an automated chemiluminescent immunoassay method for PGRN measurement recently developed (Chorus Evo, Diesse Diagnostica, Italy). METHODS Five plasma pools and residual plasma samples (K2EDTA) from 25 control subjects (11 males, 62-79 years; 14 females, 54-76 years) and 151 patients (70 males, 53-81 years; 81 females, 44-82 years) with different neurodegenerative disorders (NDs), were assayed. In 61 out of 151 patients, genetic GRN screening was carried. RESULTS Within-run imprecision (CV%) ranged from 3.8 % (11.5 pg/L) to 10.8 % (2.5 pg/L), and between-run, from 5.6 % (68.7 pg/L) to 10.7 % (2.8 pg/L). At genetic screening, 3 out of 61 patients were classified as GRN+ carriers, 18 as "other mutations" and 40 as "no-mutations" carriers. The PGRN median level in GRN+ carriers (15.9 pg/L) was significantly lower than that in control subjects (32.8 pg/L; p=0.006), in GRN- (27.50 pg/L; p=0.007), in other mutation carriers (24.80 pg/L; p=0.05) and in NDs patients (22.40 pg/L; p=0.05) ROC analysis, demonstrates the accuracy of progranulin levels in discriminating between "GRN+" and "GRN-" carriers (AUC 0.985) as well as "GRN+" and "other mutations" carriers (AUC 0.870). CONCLUSIONS The new automated progranulin method, for robust analytical performance, is suitable for use in the clinical setting, supporting clinicians in making a differential diagnosis in patients with neurodegenerative disorder.
Collapse
Affiliation(s)
- Chiara Cosma
- Department of Medicine, University of Padova, Padova, Italy
- QI.LAB.MED, Spin-off of the University, Padova, Italy
| | - Ilaria Talli
- Department of Medicine, University of Padova, Padova, Italy
- Laboratory Medicine Unit, University-Hospital, Padova, Italy
| | - Elisa Pangrazzi
- Department of Medicine, University of Padova, Padova, Italy
- QI.LAB.MED, Spin-off of the University, Padova, Italy
| | - Andrea Padoan
- Department of Medicine, University of Padova, Padova, Italy
- QI.LAB.MED, Spin-off of the University, Padova, Italy
- Laboratory Medicine Unit, University-Hospital, Padova, Italy
| | | | | | - Carlo Gabelli
- Regional Brain Aging Centre, University-Hospital, Padova, Italy
| | - Mario Plebani
- Department of Medicine, University of Padova, Padova, Italy
- Laboratory Medicine Unit, University-Hospital, Padova, Italy
| |
Collapse
|
2
|
Gaweda-Walerych K, Aragona V, Lodato S, Sitek EJ, Narożańska E, Buratti E. Progranulin deficiency in the brain: the interplay between neuronal and non-neuronal cells. Transl Neurodegener 2025; 14:18. [PMID: 40234992 PMCID: PMC12001433 DOI: 10.1186/s40035-025-00475-8] [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: 09/20/2024] [Accepted: 02/21/2025] [Indexed: 04/17/2025] Open
Abstract
Heterozygous mutations in GRN gene lead to insufficient levels of the progranulin (PGRN) protein, resulting in frontotemporal dementia (FTD) with TAR DNA-binding protein 43 (TDP-43) inclusions, classified pathologically as frontotemporal lobar degeneration (FTLD-TDP). Homozygous GRN mutations are exceedingly rare and cause neuronal ceroid lipofuscinosis 11, a lysosomal storage disease with onset in young adulthood, or an FTD syndrome with late-onset manifestations. In this review, we highlight the broad spectrum of clinical phenotypes associated with PGRN deficiency, including primary progressive aphasia and behavioral variant of frontotemporal dementia. We explore these phenotypes alongside relevant rodent and in vitro human models, ranging from the induced pluripotent stem cell-derived neural progenitors, neurons, microglia, and astrocytes to genetically engineered heterotypic organoids containing both neurons and astrocytes. We summarize advantages and limitations of these models in recapitulating the main FTLD-GRN hallmarks, highlighting the role of non-cell-autonomous mechanisms in the formation of TDP-43 pathology, neuroinflammation, and neurodegeneration. Data obtained from patients' brain tissues and biofluids, in parallel with single-cell transcriptomics, demonstrate the complexity of interactions among the highly heterogeneous cellular clusters present in the brain, including neurons, astrocytes, microglia, oligodendroglia, endothelial cells, and pericytes. Emerging evidence has revealed that PGRN deficiency is associated with cell cluster-specific, often conserved, genetic and molecular phenotypes in the central nervous system. In this review, we focus on how these distinct cellular populations and their dysfunctional crosstalk contribute to neurodegeneration and neuroinflammation in FTD-GRN. Specifically, we characterize the phenotypes of lipid droplet-accumulating microglia and alterations of myelin lipid content resulting from lysosomal dysfunction caused by PGRN deficiency. Additionally, we consider how the deregulation of glia-neuron communication affects the exchange of organelles such as mitochondria, and the removal of excess toxic products such as protein aggregates, in PGRN-related neurodegeneration.
Collapse
Affiliation(s)
- Katarzyna Gaweda-Walerych
- Department of Neurogenetics and Functional Genomics, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland.
| | - Vanessa Aragona
- Department of Biomedical Sciences, Humanitas University, Via Levi Montalicini 4, Pieve Emanuele, 20072, Milan, Italy
- Neurodevelopment Biology Lab, IRCCS Humanitas Research Hospital, via Manzoni, 56, Rozzano, 20089, Milan, Italy
| | - Simona Lodato
- Department of Biomedical Sciences, Humanitas University, Via Levi Montalicini 4, Pieve Emanuele, 20072, Milan, Italy
- Neurodevelopment Biology Lab, IRCCS Humanitas Research Hospital, via Manzoni, 56, Rozzano, 20089, Milan, Italy
| | - Emilia J Sitek
- Division of Neurological and Psychiatric Nursing, Laboratory of Clinical Neuropsychology, Neurolinguistics, and Neuropsychotherapy, Faculty of Health Sciences, Medical University of Gdansk, 80-210, Gdansk, Poland.
- Neurology Department, St. Adalbert Hospital, Copernicus PL, 80-462, Gdansk, Poland.
| | - Ewa Narożańska
- Neurology Department, St. Adalbert Hospital, Copernicus PL, 80-462, Gdansk, Poland
| | - Emanuele Buratti
- Molecular Pathology Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), AREA Science Park, 34149, Trieste, Italy
| |
Collapse
|
3
|
Wallings RL, Gillett DA, Staley HA, Mahn S, Mark J, Neighbarger N, Kordasiewicz H, Hirst WD, Tansey MG. ASO-mediated knock-down of GPNMB in mutant-GRN and in Grn-deficient peripheral myeloid cells disrupts lysosomal function and immune responses. Mol Neurodegener 2025; 20:41. [PMID: 40200337 PMCID: PMC11980231 DOI: 10.1186/s13024-025-00829-w] [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: 07/22/2024] [Accepted: 03/14/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND GPNMB has been discussed as a potential therapeutic target in GRN-mediated neurodegeneration, based on the observed reproducible upregulation in FTD-GRN cerebrospinal fluid (CSF) and post-mortem brain. However, the functional impacts of up-regulated GPNMB are currently unknown, and it is currently unclear if targeting GPNMB will be protective or deleterious. Increases in GPNMB seen in FTD-GRN are reproduced in brains of aged Grn-deficient mice. Importantly, although brains of young Grn-deficient mice do not exhibit upregulated Gpnmb expression, peripheral immune cells of these mice exhibit increased Gpnmb expression as young as 5-to-6 months, suggesting the effects of Grn-deficiency in the periphery proceed those in the brain. Grn-deficiency is known to alter peripheral immune cell function, including impaired autophagy and altered cytokine secretion. GPNMB has potential effects on these processes, but has never been studied in peripheral immune cells of patients or preclinical models. Informing the functional significance of GPNMB upregulation in Grn-deficient states in myeloid cells has potential to inform GPNMB as a therapeutic candidate. METHODS The effects of GPNMB knock-down via antisense oligonucleotide (ASO) were assessed in peripheral blood mononuclear cells (PBMCs) from 25 neurologically healthy controls (NHCs) and age- and sex-matched FTD-GRN patients, as well as peritoneal macrophages (pMacs) from progranulin-deficient (Grn -/-) and B6 mice. Lysosomal function, antigen presentation and MHC-II processing and recycling were assessed, as well as cytokine release and transcription. RESULTS ASO-mediated knock-down of GPNMB increased lysosomal burden and IL1β cytokine secretion in FTD-GRN carriers and NHCs monocytes. ASO-mediated knock-down of Gpnmb in Grn-deficient macrophages decreased lysosomal pan-cathepsin activity and protein degradation. In addition, ASO-mediated knock-down of Gpnmb increased MHC-II surface expression, which was driven by decreased MHC-II uptake and recycling, in macrophages from Grn-deficient females. Finally, ASO-mediated knock-down of Gpnmb dysregulated IFN γ -stimulated IL6 cytokine transcription and secretion by mouse macrophages due to the absence of regulatory actions of the Gpnmb extracellular fragment (ECF). CONCLUSIONS Our data herein reveal that GPNMB has a regulatory effect on multiple immune effector functions, including capping inflammation and immune responses in myeloid cells, potentially via secretion of its ECF. Therefore, in progranulin-deficient states, the marked upregulation in GPNMB transcript and protein may represent a compensatory mechanism to preserve lysosomal function in myeloid cells. These novel findings indicate that targeted depletion of GPNMB in FTD-GRN would not be a rational therapeutic strategy because it is likely to dysregulate important immune cell effector functions mediated by GPNMB. Specifically, our data indicate that therapeutic strategies inhibiting GPNMB levels and/or activity may worsen the effects of GRN deficiency.
Collapse
Affiliation(s)
- Rebecca L Wallings
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, McKnight Brain Institute, Gainesville, FL, 32610, USA.
- Current address: Department of Neurology, School of Medicine, Stark Neurosciences Research Institute, Indiana University, Indianapolis, IN, USA.
| | - Drew A Gillett
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, McKnight Brain Institute, Gainesville, FL, 32610, USA
| | - Hannah A Staley
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, McKnight Brain Institute, Gainesville, FL, 32610, USA
| | - Savanna Mahn
- Department of Neurology, Fixel Institute for Neurological Diseases, University of Florida Health, Gainesville, FL, 32608, USA
| | - Julian Mark
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, McKnight Brain Institute, Gainesville, FL, 32610, USA
| | - Noelle Neighbarger
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, McKnight Brain Institute, Gainesville, FL, 32610, USA
| | - Holly Kordasiewicz
- Neurology, Ionis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA, 92010, USA
| | - Warren D Hirst
- Neurodegenerative Diseases Research Unit, Biogen, 115 Broadway, Cambridge, MA, 02142, USA
- Current address: DaCapo Brainscience, 700 Main Street, Cambridge, MA, 02139, USA
| | - Malú Gámez Tansey
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, McKnight Brain Institute, Gainesville, FL, 32610, USA.
- Department of Neurology, Fixel Institute for Neurological Diseases, University of Florida Health, Gainesville, FL, 32608, USA.
- Current address: Department of Neurology, School of Medicine, Stark Neurosciences Research Institute, Indiana University, Indianapolis, IN, USA.
| |
Collapse
|
4
|
Erichsen PA, Henriksen EE, Nielsen JE, Ejlerskov P, Simonsen AH, Toft A. Immunological Fluid Biomarkers in Frontotemporal Dementia: A Systematic Review. Biomolecules 2025; 15:473. [PMID: 40305176 PMCID: PMC12025258 DOI: 10.3390/biom15040473] [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: 02/03/2025] [Revised: 03/15/2025] [Accepted: 03/23/2025] [Indexed: 05/02/2025] Open
Abstract
Dysregulated immune activation plays a key role in the pathogenesis of neurodegenerative diseases, including frontotemporal dementia (FTD). This study reviews immunological biomarkers associated with FTD and its subtypes. A systematic search of PubMed and Web of Science was conducted for studies published before 1 January 2025, focusing on immunological biomarkers in CSF or blood from FTD patients with comparisons to healthy or neurological controls. A total of 124 studies were included, involving 6686 FTD patients and 202 immune biomarkers. Key findings include elevated levels of GFAP and MCP1/CCL2 in both CSF and blood and consistently increased CHIT1 and YKL-40 in CSF. Complement proteins from the classical activation pathway emerged as promising targets. Distinct immune markers were found to differentiate FTD from Alzheimer's disease (AD) and amyotrophic lateral sclerosis (ALS), with GFAP, SPARC, and SPP1 varying between FTD and AD and IL-15, HERV-K, NOD2, and CHIT1 differing between FTD and ALS. A few markers, such as Galectin-3 and PGRN, distinguished FTD subtypes. Enrichment analysis highlighted IL-10 signaling and immune cell chemotaxis as potential pathways for further exploration. This study provides an overview of immunological biomarkers in FTD, emphasizing those most relevant for future research on immune dysregulation in FTD pathogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | - Anders Toft
- Neurogenetics Clinic & Research Lab, Danish Dementia Research Centre, Rigshospitalet, 2100 Copenhagen, Denmark; (P.A.E.); (E.E.H.); (J.E.N.); (P.E.); (A.H.S.)
| |
Collapse
|
5
|
Xia M, Gao C, Shang J, Li D, Yang A, Zang W, Zhang J. A novel c.1468 G > A GRN mutation causes frontotemporal dementia in a Chinese Han family. Eur J Med Res 2025; 30:157. [PMID: 40055834 PMCID: PMC11889931 DOI: 10.1186/s40001-025-02418-5] [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: 01/21/2025] [Accepted: 02/27/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND/PURPOSE GRN mutations act as causative factors in patients with FTD clinical phenotype or FTD pathology and exhibit high clinical heterogeneity. The discovery of these mutations and the analysis of their associations with resembling Alzheimer's disease should be critical to understand the pathogenesis of FTD. METHODS Clinical analysis, neuroimaging, target region capture and high-throughput sequencing were performed in a family of 3 generations. The underlying Alzheimer's pathology was evaluated by using biomarker evidence obtained from cerebrospinal fluid (CSF) amyloid testing, 18F-florbetapir (AV-45) PET imaging and FDG18-positron emission tomography imaging. RESULTS Through target region capture and high-throughput sequencing, a three-generation family was able to identify a heterozygous G to A point mutation at position 490 (c.1468)G > A, which led to a valine to methionine substitution (V490M) at exon 12. This unique missense mutation was found at codon 1468. Eight members of the proband's family-two sisters and the proband himself-had the mutation found by Sanger sequencing. Interestingly, biomarker tests for amyloid in the proband's cerebrospinal fluid (CSF) indicated pathology consistent with Alzheimer's disease (AD). The mutation was expected to have a high likelihood of being pathogenic. CONCLUSIONS We firstly reported a novel mutation in the GRN gene at codon 490 (V490M) in exon 12 in a China FTD family. The CSF biomarker alterations of the proband revealed a reduction in Aβ42 and the Aβ42/Aβ40 ratio. The analysis of mutation might support the role of GRN in patients with FTD and contribute to the discovery of a new pathological mechanism underlying the disease.
Collapse
Affiliation(s)
- Mingrong Xia
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Chenhao Gao
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450003, Henan, China
| | - Junkui Shang
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450003, Henan, China
| | - Dan Li
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Ali Yang
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Weizhou Zang
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China.
| | - Jiewen Zhang
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China.
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450003, Henan, China.
| |
Collapse
|
6
|
Sogorb-Esteve A, Weiner S, Simrén J, Swift IJ, Bocchetta M, Todd EG, Cash DM, Bouzigues A, Russell LL, Foster PH, Ferry-Bolder E, van Swieten JC, Jiskoot LC, Seelaar H, Sanchez-Valle R, 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 CR, Le Ber I, Finger E, Tartaglia MC, Masellis M, Rowe JB, Synofzik M, Moreno F, Borroni B, Genfi, Blennow K, Zetterberg H, Rohrer JD, Gobom J. Proteomic analysis reveals distinct cerebrospinal fluid signatures across genetic frontotemporal dementia subtypes. Sci Transl Med 2025; 17:eadm9654. [PMID: 39908349 DOI: 10.1126/scitranslmed.adm9654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 07/22/2024] [Accepted: 01/14/2025] [Indexed: 02/07/2025]
Abstract
We used an untargeted mass spectrometric approach, tandem mass tag proteomics, for the identification of proteomic signatures in genetic frontotemporal dementia (FTD). A total of 238 cerebrospinal fluid (CSF) samples from the Genetic FTD Initiative were analyzed, including samples from 107 presymptomatic (44 C9orf72, 38 GRN, and 25 MAPT) and 55 symptomatic (27 C9orf72, 17 GRN, and 11 MAPT) mutation carriers as well as 76 mutation-negative controls ("noncarriers"). We found shared and distinct proteomic alterations in each genetic form of FTD. Among the proteins significantly altered in symptomatic mutation carriers compared with noncarriers, we found that a set of proteins including neuronal pentraxin 2 and fatty acid binding protein 3 changed across all three genetic forms of FTD and patients with Alzheimer's disease from previously published datasets. We observed differential changes in lysosomal proteins among symptomatic mutation carriers with marked abundance decreases in MAPT carriers but not other carriers. Further, we identified mutation-associated proteomic changes already evident in presymptomatic mutation carriers. Weighted gene coexpression network analysis combined with gene ontology annotation revealed clusters of proteins enriched in neurodegeneration and glial responses as well as synapse- or lysosome-related proteins indicating that these are the central biological processes affected in genetic FTD. These clusters correlated with measures of disease severity and were associated with cognitive decline. This study revealed distinct proteomic changes in the CSF of patients with genetic FTD, providing insights into the pathological processes involved in the disease. In addition, we identified proteins that warrant further exploration as diagnostic and prognostic biomarker candidates.
Collapse
Affiliation(s)
- Aitana Sogorb-Esteve
- UK Dementia Research Institute at University College London, WC1N 3BG London, UK
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, WC1N 3BG London, UK
| | - Sophia Weiner
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, 431 39 Mölndal, Sweden
| | - Joel Simrén
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, 431 39 Mölndal, Sweden
| | - Imogen J Swift
- UK Dementia Research Institute at University College London, WC1N 3BG London, UK
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, WC1N 3BG London, UK
| | - Martina Bocchetta
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, WC1N 3BG London, UK
- Centre for Cognitive and Clinical Neuroscience, Division of Psychology, Department of Life Sciences, College of Health, Medicine, and Life Sciences, Brunel University, UB8 3PH London, UK
| | - Emily G Todd
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, WC1N 3BG London, UK
| | - David M Cash
- UK Dementia Research Institute at University College London, WC1N 3BG London, UK
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, WC1N 3BG London, UK
| | - Arabella Bouzigues
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, WC1N 3BG London, UK
| | - Lucy L Russell
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, WC1N 3BG London, UK
| | - Phoebe H Foster
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, WC1N 3BG London, UK
| | - Eve Ferry-Bolder
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, WC1N 3BG London, UK
| | - John C van Swieten
- Department of Neurology, Erasmus Medical Centre, 3015 GD Rotterdam, Netherlands
| | - Lize C Jiskoot
- Department of Neurology, Erasmus Medical Centre, 3015 GD Rotterdam, Netherlands
| | - Harro Seelaar
- Department of Neurology, Erasmus Medical Centre, 3015 GD Rotterdam, Netherlands
| | - Raquel Sanchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, 08036 Barcelona, Spain
| | - Robert Laforce
- Clinique Interdisciplinaire de Mémoire, Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Caroline Graff
- Center for Alzheimer Research, Division of Neurogeriatrics, Department of Neurobiology, Care Sciences, and Society, Bioclinicum, Karolinska Institutet, 171 64 Solna, Sweden
- Unit for Hereditary Dementias, Theme Aging, Karolinska University Hospital, 171 77 Solna, Sweden
| | - Daniela Galimberti
- Fondazione Ca' Granda, IRCCS Ospedale Policlinico, 20122 Milan, Italy
- University of Milan, Centro Dino Ferrari, 20122 Milan, Italy
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, 3000 Leuven, Belgium
- Neurology Service, University Hospitals Leuven, 3000 Leuven, Belgium
- Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium
| | | | - Pietro Tiraboschi
- Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milano, Italy
| | - Isabel Santana
- University Hospital of Coimbra (HUC), Neurology Service, Faculty of Medicine, University of Coimbra, 3004-531 Coimbra, Portugal
- Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, 3004-531 Coimbra, Portugal
| | - Alexander Gerhard
- Division of Neuroscience and Experimental Psychology, Wolfson Molecular Imaging Centre, University of Manchester, M20 3LJ Manchester, UK
- Departments of Geriatric Medicine and Nuclear Medicine, University of Duisburg-Essen, 45141 Essen, Germany
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians Universität München, 80539 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Sandro Sorbi
- Department of Neurofarba, University of Florence, 50139 Florence, Italy
- IRCCS Fondazione Don Carlo Gnocchi, 50143 Florence, Italy
| | - Markus Otto
- Department of Neurology, University of Ulm, 89081 Ulm, Germany
- Department of Neurology, Martin-Luther-University Hospital of Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Florence Pasquier
- University of Lille, 59000 Lille, France
- Inserm 1172, Lille, 59000 Lille, France
- CHU, CNR-MAJ, Labex Distalz, LiCEND Lille, 59000 Lille, France
| | - Simon Ducharme
- Department of Psychiatry, McGill University Health Centre, McGill University, Montreal, Québec H4A 3J1, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Québec H3A 0G4, Canada
| | - Chris R Butler
- Nuffield Department of Clinical Neurosciences, Medical Sciences Division, University of Oxford, OX3 9DU Oxford, UK
- Department of Brain Sciences, Imperial College London, W12 0NN London, UK
| | - Isabelle Le Ber
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau - ICM, Inserm U1127, CNRS UMR 7225, AP-HP - Hôpital Pitié-Salpêtrière, 75013 Paris, France
- Centre de référence des démences rares ou précoces, IM2A, Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, 75013 Paris, France
- Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, 75013 Paris, France
| | - Elizabeth Finger
- Department of Clinical Neurological Sciences, University of Western Ontario, London, Ontario N6A 5A5, Canada
| | - Maria Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Mario Masellis
- Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario M4N 3M5, Canada
| | - James B Rowe
- Department of Clinical Neurosciences, University of Cambridge, CB2 3EB Cambridge, UK
| | - Matthis Synofzik
- Division Translational Genomics of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, 72076 Tübingen, Germany
- Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
| | - Fermin Moreno
- Cognitive Disorders Unit, Department of Neurology, Donostia Universitary Hospital, 20014 San Sebastian, Spain
- Neuroscience Area, Biodonostia Health Research Institute, 20014 San Sebastian, Gipuzkoa, Spain
| | - Barbara Borroni
- Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy
- Department of Continuity of Care and Frialy, ASST Spedali Civili Brescia, 25123 Brescia, Italy
| | | | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, 431 39 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, SE-43180 Mölndal, Sweden
- Institut du Cerveau et de la Moelle épinière (ICM), Pitié-Salpêtrière Hospital, Sorbonne Université, 75013 Paris, France
- University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, Anhui, P.R. China
| | - Henrik Zetterberg
- UK Dementia Research Institute at University College London, WC1N 3BG London, UK
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, 431 39 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, SE-43180 Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, WC1N 3BG London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Jonathan D Rohrer
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, WC1N 3BG London, UK
| | - Johan Gobom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, 431 39 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, SE-43180 Mölndal, Sweden
| |
Collapse
|
7
|
Liampas I, Kyriakoulopoulou P, Karakoida V, Kavvoura PA, Sgantzos M, Bogdanos DP, Stamati P, Dardiotis E, Siokas V. Blood-Based Biomarkers in Frontotemporal Dementia: A Narrative Review. Int J Mol Sci 2024; 25:11838. [PMID: 39519389 PMCID: PMC11546606 DOI: 10.3390/ijms252111838] [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: 09/21/2024] [Revised: 10/20/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
This narrative review explores the current landscape of blood biomarkers in Frontotemporal dementia (FTD). Neurofilament light chain (NfL) may be useful in the differentiation of behavioral variant FTD from primary psychiatric disorders (PPDs) or dementia with Lewy bodies (DLB). In prodromal FTD and presymptomatic mutation carriers (GRN, MAPT, C9orf72), elevated NfL may herald pheno-conversion to full-blown dementia. Baseline NfL correlates with steeper neuroanatomical changes and cognitive, behavioral and functional decline, making NfL promising in monitoring disease progression. Phosphorylated neurofilament heavy chain (pNfH) levels have a potential limited role in the demarcation of the conversion stage to full-blown FTD. Combined NfL and pNfH measurements may allow a wider stage stratification. Total tau levels lack applicability in the framework of FTD. p-tau, on the other hand, is of potential value in the discrimination of FTD from Alzheimer's dementia. Progranulin concentrations could serve the identification of GRN mutation carriers. Glial fibrillary acidic protein (GFAP) may assist in the differentiation of PPDs from behavioral variant FTD and the detection of GRN mutation carriers (additional research is warranted). Finally, TAR DNA-binding protein-43 (TDP-43) appears to be a promising diagnostic biomarker for FTD. Its potential in distinguishing TDP-43 pathology from other FTD-related pathologies requires further research.
Collapse
Affiliation(s)
- Ioannis Liampas
- Department of Neurology, University Hospital of Larissa, School of Medicine, University of Thessaly, 41100 Larissa, Greece; (P.S.); (E.D.); (V.S.)
| | | | - Vasiliki Karakoida
- School of Medicine, University of Patras, 26504 Rio Patras, Greece; (P.K.); (V.K.); (P.A.K.)
| | | | - Markos Sgantzos
- Department of Anatomy, Medical School, University of Thessaly, 41100 Larissa, Greece;
| | - Dimitrios P. Bogdanos
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41100 Larissa, Greece;
| | - Polyxeni Stamati
- Department of Neurology, University Hospital of Larissa, School of Medicine, University of Thessaly, 41100 Larissa, Greece; (P.S.); (E.D.); (V.S.)
| | - Efthimios Dardiotis
- Department of Neurology, University Hospital of Larissa, School of Medicine, University of Thessaly, 41100 Larissa, Greece; (P.S.); (E.D.); (V.S.)
| | - Vasileios Siokas
- Department of Neurology, University Hospital of Larissa, School of Medicine, University of Thessaly, 41100 Larissa, Greece; (P.S.); (E.D.); (V.S.)
| |
Collapse
|
8
|
Kashyap SN, Fox SN, Wilson KI, Murchison CF, Ambaw YA, Walther TC, Farese RV, Arrant AE, Roberson ED. Carboxy-terminal blockade of sortilin binding enhances progranulin gene therapy, a potential treatment for frontotemporal dementia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.15.613118. [PMID: 39345608 PMCID: PMC11430072 DOI: 10.1101/2024.09.15.613118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Frontotemporal dementia is commonly caused by loss-of-function mutations in the progranulin gene. Potential therapies for this disorder have entered clinical trials, including progranulin gene therapy and drugs that reduce progranulin interactions with sortilin. Both approaches ameliorate functional and pathological abnormalities in mouse models of progranulin insufficiency. Here we investigated whether modifying the progranulin carboxy terminus to block sortilin interactions would improve the efficacy of progranulin gene therapy. We compared the effects of treating progranulin-deficient mice with gene therapy vectors expressing progranulin with intact sortilin interactions, progranulin with the carboxy terminus blocked to reduce sortilin interactions, or GFP control. We found that expressing carboxy-terminally blocked progranulin generated higher levels of progranulin both at the injection site and in more distant regions. Carboxy-terminally blocked progranulin was also more effective at ameliorating microgliosis, microglial lipofuscinosis, and lipid abnormalities including ganglioside accumulation and loss of bis(monoacylglycero)phosphate lipids. Finally, only carboxy-terminally blocked progranulin reduced plasma neurofilament light chain, a biomarker of neurodegeneration, in progranulin-deficient mice. These results demonstrate that modifying the progranulin cargo to block sortilin interactions may be important for increasing the effectiveness of progranulin gene therapy. One-sentence Summary The effectiveness of progranulin gene therapy in models of FTD is improved by blocking the protein's carboxy terminus, which prevents sortilin binding.
Collapse
|
9
|
Ek M, Nilvebrant J, Nygren PÅ, Ståhl S, Lindberg H, Löfblom J. An anti-sortilin affibody-peptide fusion inhibits sortilin-mediated progranulin degradation. Front Immunol 2024; 15:1437886. [PMID: 39185427 PMCID: PMC11342335 DOI: 10.3389/fimmu.2024.1437886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 07/23/2024] [Indexed: 08/27/2024] Open
Abstract
Heterozygous loss-of-function mutations in the GRN gene are a common cause of frontotemporal dementia. Such mutations lead to decreased plasma and cerebrospinal fluid levels of progranulin (PGRN), a neurotrophic factor with lysosomal functions. Sortilin is a negative regulator of extracellular PGRN levels and has shown promise as a therapeutic target for frontotemporal dementia, enabling increased extracellular PGRN levels through inhibition of sortilin-mediated PGRN degradation. Here we report the development of a high-affinity sortilin-binding affibody-peptide fusion construct capable of increasing extracellular PGRN levels in vitro. By genetic fusion of a sortilin-binding affibody generated through phage display and a peptide derived from the progranulin C-terminus, an affinity protein (A3-PGRNC15*) with 185-pM affinity for sortilin was obtained. Treating PGRN-secreting and sortilin-expressing human glioblastoma U-251 cells with the fusion protein increased extracellular PGRN levels up to 2.5-fold, with an EC50 value of 1.3 nM. Our results introduce A3-PGRNC15* as a promising new agent with therapeutic potential for the treatment of frontotemporal dementia. Furthermore, the work highlights means to increase binding affinity through synergistic contribution from two orthogonal polypeptide units.
Collapse
Affiliation(s)
| | | | | | | | | | - John Löfblom
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|
10
|
Wallings RL, Gillett DA, Staley HA, Mahn S, Mark J, Neighbarger N, Kordasiewicz H, Hirst WD, Tansey MG. ASO-mediated knockdown of GPNMB in mutant- GRN and Grn -deficient peripheral myeloid cells disrupts lysosomal function and immune responses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.22.604676. [PMID: 39211224 PMCID: PMC11361193 DOI: 10.1101/2024.07.22.604676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Background Increases in GPNMB are detectable in FTD- GRN cerebrospinal fluid (CSF) and post-mortem brain, and brains of aged Grn -deficient mice. Although no upregulation of GPNMB is observed in the brains of young Grn -deficient mice, peripheral immune cells of these mice do exhibit this increase in GPNMB. Importantly, the functional significance of GPNMB upregulation in progranulin-deficient states is currently unknown. Given that GPNMB has been discussed as a potential therapeutic target in GRN -mediated neurodegeneration, it is vital for the field to determine what the normal function of GPNMB is in the immune system, and whether targeting GPNMB will elicit beneficial or deleterious effects. Methods The effects of GPNMB knock-down via antisense oligonucleotide (ASO) were assessed in peripheral blood mononuclear cells (PBMCs) from 25 neurologically healthy controls (NHCs) and age- and sex-matched FTD- GRN patients, as well as peritoneal macrophages (pMacs) from progranulin-deficient ( Grn -/- ) and B6 mice. Lysosomal function, antigen presentation and MHC-II processing and recycling were assessed, as well as cytokine release and transcription. Results We demonstrate here that ASO-mediated knockdown of GPNMB increases lysosomal burden and cytokine secretion in FTD-GRN carrier and neurologically healthy controls (NHCs) monocytes. ASO-mediated knockdown of GPNMB in Grn -deficient macrophages decreased lysosomal pan-cathepsin activity and protein degradation. In addition, ASO-mediated knockdown of GPNMB increased MHC-II surface expression, which was driven by decreased MHC-II uptake and recycling, in macrophages from Grn -deficient females. Finally, ASO-mediated knockdown of GPNMB dysregulated IFNγ-stimulated cytokine transcription and secretion by mouse macrophages due to the absence of regulatory actions of the GPNMB extracellular fragment (ECF). Conclusions Our data herein reveals that GPNMB has a regulatory effect on multiple immune effector functions, including capping inflammation and immune responses in myeloid cells via secretion of its ECF. Therefore, in progranulin-deficient states, the drastic upregulation in GPNMB transcript and protein may represent a compensatory mechanism to preserve lysosomal function in myeloid cells. These novel findings indicate that targeted depletion in FTD- GRN would not be a rational therapeutic strategy because it is likely to dysregulate important immune cell effector functions.
Collapse
|
11
|
Cummings J. Rarer dementias, limited options, and unaddressed needs. Int Psychogeriatr 2024; 36:451-454. [PMID: 37137670 PMCID: PMC11827695 DOI: 10.1017/s1041610223000431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Affiliation(s)
- Jeffrey Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Las Vegas, NV, USA
- Department of Brain Health, School of Integrated Health Sciences, Las Vegas, NV, USA
- University of Nevada Las Vegas, Las Vegas, NV, USA
| |
Collapse
|
12
|
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
|
13
|
Tordai DZ, Hajdú N, Rácz R, Istenes I, Békeffy M, Vági OE, Kempler M, Körei AE, Tóbiás B, Illés A, Pikó H, Kósa JP, Árvai K, Papp M, Lakatos PA, Kempler P, Putz Z. Genetic Factors Associated with the Development of Neuropathy in Type 2 Diabetes. Int J Mol Sci 2024; 25:1815. [PMID: 38339094 PMCID: PMC10855482 DOI: 10.3390/ijms25031815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/25/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
Neuropathy is a serious and frequent complication of type 2 diabetes (T2DM). This study was carried out to search for genetic factors associated with the development of diabetic neuropathy by whole exome sequencing. For this study, 24 patients with long-term type 2 diabetes with neuropathy and 24 without underwent detailed neurological assessment and whole exome sequencing. Cardiovascular autonomic function was evaluated by cardiovascular reflex tests. Heart rate variability was measured by the triangle index. Sensory nerve function was estimated by Neurometer and Medoc devices. Neuropathic symptoms were characterized by the neuropathy total symptom score (NTSS). Whole exome sequencing (WES) was performed on a Thermo Ion GeneStudio S5 system determining the coding sequences of approximately 32,000 genes comprising 50 million base pairs. Variants were detected by Ion Reporter software and annotated using ANNOVAR, integrating database information from dbSNP, ClinVar, gnomAD, and OMIM. Integrative genomics viewer (IGV) was used for visualization of the mapped reads. We have identified genetic variants that were significantly associated with increased (22-49-fold) risk of neuropathy (rs2032930 and rs2032931 of recQ-mediated genome instability protein 2 (RMI2) gene), rs604349 of myosin binding protein H like (MYBPHL) gene and with reduced (0.07-0.08-fold) risk (rs917778 of multivesicular body subunit 12B (MVB12B) and rs2234753 of retinoic acid X receptor alpha (RXRA) genes). The rs2032930 showed a significant correlation with current perception thresholds measured at 5 Hz and 250 Hz for n. medianus (p = 0.042 and p = 0.003, respectively) and at 5 Hz for n. peroneus (p = 0.037), as well as the deep breath test (p = 0.022) and the NTSS (p = 0.023). The rs2032931 was associated with current perception thresholds (p = 0.003 and p = 0.037, respectively), deep breath test (p = 0.022), and NTSS (p = 0.023). The rs604349 correlated with values measured at 2000 (p = 0.049), 250 (p = 0.018), and 5 Hz (p = 0.005) for n. medianus, as well as warm perception threshold measured by Medoc device (p = 0.042). The rs2234753 showed correlations with a current perception threshold measured at 2000 Hz for n. medianus (p = 0.020), deep breath test (p = 0.040), and NTSS (p = 0.003). There was a significant relationship between rs91778 and cold perception threshold (p = 0.013). In our study, genetic variants have been identified that may have an impact on the risk of neuropathy developing in type 2 diabetic patients. These results could open up new opportunities for early preventive measures and might provide targets for new drug developments in the future.
Collapse
Affiliation(s)
- Dóra Zsuszanna Tordai
- Department of Internal Medicine and Oncology, Semmelweis University, 1083 Budapest, Hungary; (N.H.); (I.I.); (M.B.); (O.E.V.); or (A.E.K.); (B.T.); (A.I.); (H.P.); (J.P.K.); (P.A.L.); (P.K.); or (Z.P.)
| | - Noémi Hajdú
- Department of Internal Medicine and Oncology, Semmelweis University, 1083 Budapest, Hungary; (N.H.); (I.I.); (M.B.); (O.E.V.); or (A.E.K.); (B.T.); (A.I.); (H.P.); (J.P.K.); (P.A.L.); (P.K.); or (Z.P.)
| | - Ramóna Rácz
- Department of Internal Medicine and Oncology, Semmelweis University, 1083 Budapest, Hungary; (N.H.); (I.I.); (M.B.); (O.E.V.); or (A.E.K.); (B.T.); (A.I.); (H.P.); (J.P.K.); (P.A.L.); (P.K.); or (Z.P.)
| | - Ildikó Istenes
- Department of Internal Medicine and Oncology, Semmelweis University, 1083 Budapest, Hungary; (N.H.); (I.I.); (M.B.); (O.E.V.); or (A.E.K.); (B.T.); (A.I.); (H.P.); (J.P.K.); (P.A.L.); (P.K.); or (Z.P.)
| | - Magdolna Békeffy
- Department of Internal Medicine and Oncology, Semmelweis University, 1083 Budapest, Hungary; (N.H.); (I.I.); (M.B.); (O.E.V.); or (A.E.K.); (B.T.); (A.I.); (H.P.); (J.P.K.); (P.A.L.); (P.K.); or (Z.P.)
| | - Orsolya Erzsébet Vági
- Department of Internal Medicine and Oncology, Semmelweis University, 1083 Budapest, Hungary; (N.H.); (I.I.); (M.B.); (O.E.V.); or (A.E.K.); (B.T.); (A.I.); (H.P.); (J.P.K.); (P.A.L.); (P.K.); or (Z.P.)
| | - Miklós Kempler
- Department of Internal Medicine and Hematology, Semmelweis University, 1085 Budapest, Hungary;
| | - Anna Erzsébet Körei
- Department of Internal Medicine and Oncology, Semmelweis University, 1083 Budapest, Hungary; (N.H.); (I.I.); (M.B.); (O.E.V.); or (A.E.K.); (B.T.); (A.I.); (H.P.); (J.P.K.); (P.A.L.); (P.K.); or (Z.P.)
| | - Bálint Tóbiás
- Department of Internal Medicine and Oncology, Semmelweis University, 1083 Budapest, Hungary; (N.H.); (I.I.); (M.B.); (O.E.V.); or (A.E.K.); (B.T.); (A.I.); (H.P.); (J.P.K.); (P.A.L.); (P.K.); or (Z.P.)
- PentaCore Laboratory, 1134 Budapest, Hungary;
- Vascular Diagnostics Ltd., 1026 Budapest, Hungary
- Eötvös Lóránd Scientific Network ENDOMOLPAT, Semmelweis University, 1085 Budapest, Hungary
| | - Anett Illés
- Department of Internal Medicine and Oncology, Semmelweis University, 1083 Budapest, Hungary; (N.H.); (I.I.); (M.B.); (O.E.V.); or (A.E.K.); (B.T.); (A.I.); (H.P.); (J.P.K.); (P.A.L.); (P.K.); or (Z.P.)
- PentaCore Laboratory, 1134 Budapest, Hungary;
- Eötvös Lóránd Scientific Network ENDOMOLPAT, Semmelweis University, 1085 Budapest, Hungary
| | - Henriett Pikó
- Department of Internal Medicine and Oncology, Semmelweis University, 1083 Budapest, Hungary; (N.H.); (I.I.); (M.B.); (O.E.V.); or (A.E.K.); (B.T.); (A.I.); (H.P.); (J.P.K.); (P.A.L.); (P.K.); or (Z.P.)
- PentaCore Laboratory, 1134 Budapest, Hungary;
- Eötvös Lóránd Scientific Network ENDOMOLPAT, Semmelweis University, 1085 Budapest, Hungary
| | - János Pál Kósa
- Department of Internal Medicine and Oncology, Semmelweis University, 1083 Budapest, Hungary; (N.H.); (I.I.); (M.B.); (O.E.V.); or (A.E.K.); (B.T.); (A.I.); (H.P.); (J.P.K.); (P.A.L.); (P.K.); or (Z.P.)
- PentaCore Laboratory, 1134 Budapest, Hungary;
- Vascular Diagnostics Ltd., 1026 Budapest, Hungary
- Eötvös Lóránd Scientific Network ENDOMOLPAT, Semmelweis University, 1085 Budapest, Hungary
| | - Kristóf Árvai
- PentaCore Laboratory, 1134 Budapest, Hungary;
- Vascular Diagnostics Ltd., 1026 Budapest, Hungary
| | - Márton Papp
- Centre for Bioinformatics, University of Veterinary Medicine, 1078 Budapest, Hungary;
| | - Péter András Lakatos
- Department of Internal Medicine and Oncology, Semmelweis University, 1083 Budapest, Hungary; (N.H.); (I.I.); (M.B.); (O.E.V.); or (A.E.K.); (B.T.); (A.I.); (H.P.); (J.P.K.); (P.A.L.); (P.K.); or (Z.P.)
- PentaCore Laboratory, 1134 Budapest, Hungary;
- Vascular Diagnostics Ltd., 1026 Budapest, Hungary
- Eötvös Lóránd Scientific Network ENDOMOLPAT, Semmelweis University, 1085 Budapest, Hungary
| | - Péter Kempler
- Department of Internal Medicine and Oncology, Semmelweis University, 1083 Budapest, Hungary; (N.H.); (I.I.); (M.B.); (O.E.V.); or (A.E.K.); (B.T.); (A.I.); (H.P.); (J.P.K.); (P.A.L.); (P.K.); or (Z.P.)
| | - Zsuzsanna Putz
- Department of Internal Medicine and Oncology, Semmelweis University, 1083 Budapest, Hungary; (N.H.); (I.I.); (M.B.); (O.E.V.); or (A.E.K.); (B.T.); (A.I.); (H.P.); (J.P.K.); (P.A.L.); (P.K.); or (Z.P.)
- Eötvös Lóránd Scientific Network ENDOMOLPAT, Semmelweis University, 1085 Budapest, Hungary
| |
Collapse
|
14
|
Chemparathy A, Le Guen Y, Zeng Y, Gorzynski J, Jensen TD, Yang C, Kasireddy N, Talozzi L, Belloy M, Stewart I, Gitler AD, Wagner AD, Mormino E, Henderson VW, Wyss-Coray T, Ashley E, Cruchaga C, Greicius MD. A 3'UTR Insertion Is a Candidate Causal Variant at the TMEM106B Locus Associated With Increased Risk for FTLD-TDP. Neurol Genet 2024; 10:e200124. [PMID: 39911968 PMCID: PMC10848896 DOI: 10.1212/nxg.0000000000200124] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 11/27/2023] [Indexed: 02/07/2025]
Abstract
Background and Objectives Single-nucleotide variants near TMEM106B associate with the risk of frontotemporal lobar dementia with TDP-43 inclusions (FTLD-TDP) and Alzheimer disease (AD) in genome-wide association studies (GWASs), but the causal variant at this locus remains unclear. Here, we asked whether a novel structural variant on TMEM106B is the causal variant. Methods An exploratory analysis identified structural variants on neurodegeneration-related genes. Subsequent analyses focused on an Alu element insertion on the 3'UTR of TMEM106B. This study included data from longitudinal aging and neurogenerative disease cohorts at Stanford University, case-control cohorts in the Alzheimer Disease Sequencing Project (ADSP), and expression and proteomics data from Washington University in St. Louis (WUSTL). Four hundred thirty-two individuals from 2 Stanford aging cohorts were whole-genome long-read and short-read sequenced. A total of 16,906 samples from ADSP were short-read sequenced. Genotypes, transcriptomics, and proteomics data were available in 1,979 participants from an aging and dementia cohort at WUSTL. Selection criteria were specific to each cohort. In primary analyses, the linkage disequilibrium between the TMEM106B locus variants in the FTLD-TDP GWAS and the 3'UTR insertion was estimated. We then estimated linkage by ancestry in the ADSP and evaluated the effect of the TMEM106B lead variant on mRNA and protein levels. Results The primary analysis included 432 participants (52.5% female, age range 45-92 years). We identified a 316 bp Alu insertion overlapping the TMEM106B 3'UTR tightly linked with top GWAS variants rs3173615(C) and rs1990622(A). In ADSP European ancestry participants, this insertion is in equivalent linkage with rs1990622(A) (R2 = 0.962, D' = 0.998) and rs3173615(C) (R2 = 0.960, D' = 0.996). In African ancestry participants, the insertion is in stronger linkage with rs1990622(A) (R2 = 0.992, D' = 0.998) than with rs3173615(C) (R2 = 0.811, D' = 0.994). In public data sets, rs1990622 was consistently associated with TMEM106B protein levels but not with mRNA expression. In the WUSTL data set, rs1990622 is associated with TMEM106B protein levels in plasma and CSF, but not with TMEM106B mRNA expression. Discussion We identified a novel Alu element insertion in the 3'UTR of TMEM106B in tight linkage with the lead FTLD-TDP risk variant. The lead variant is associated with TMEM106B protein levels, but not expression. The 3'UTR insertion is a lead candidate for the causal variant at this complex locus, pending confirmation with functional studies.
Collapse
Affiliation(s)
- Augustine Chemparathy
- From the Department of Neurology and Neurological Sciences (A.C., Y.L.G., N.K., L.T., M.B., I.S., V.W.H., T.W.-C., M.D.G.); Quantitative Sciences Unit (Y.L.G.), Department of Medicine; Department of Genetics (Y.Z., J.G., T.D.J., A.D.G., E.A.); Division of Cardiology (J.G., E.A.), Department of Medicine, Stanford University School of Medicine, CA; Neurogenomics and Informatics Center (C.Y., C.C.), Washington University School of Medicine, St. Louis, MO; Wu Tsai Neurosciences Institute (A.D.W., E.M.); Department of Psychology (A.D.W., E.M.); and Department of Epidemiology and Population Health (V.W.H.), Stanford University, CA
| | - Yann Le Guen
- From the Department of Neurology and Neurological Sciences (A.C., Y.L.G., N.K., L.T., M.B., I.S., V.W.H., T.W.-C., M.D.G.); Quantitative Sciences Unit (Y.L.G.), Department of Medicine; Department of Genetics (Y.Z., J.G., T.D.J., A.D.G., E.A.); Division of Cardiology (J.G., E.A.), Department of Medicine, Stanford University School of Medicine, CA; Neurogenomics and Informatics Center (C.Y., C.C.), Washington University School of Medicine, St. Louis, MO; Wu Tsai Neurosciences Institute (A.D.W., E.M.); Department of Psychology (A.D.W., E.M.); and Department of Epidemiology and Population Health (V.W.H.), Stanford University, CA
| | - Yi Zeng
- From the Department of Neurology and Neurological Sciences (A.C., Y.L.G., N.K., L.T., M.B., I.S., V.W.H., T.W.-C., M.D.G.); Quantitative Sciences Unit (Y.L.G.), Department of Medicine; Department of Genetics (Y.Z., J.G., T.D.J., A.D.G., E.A.); Division of Cardiology (J.G., E.A.), Department of Medicine, Stanford University School of Medicine, CA; Neurogenomics and Informatics Center (C.Y., C.C.), Washington University School of Medicine, St. Louis, MO; Wu Tsai Neurosciences Institute (A.D.W., E.M.); Department of Psychology (A.D.W., E.M.); and Department of Epidemiology and Population Health (V.W.H.), Stanford University, CA
| | - John Gorzynski
- From the Department of Neurology and Neurological Sciences (A.C., Y.L.G., N.K., L.T., M.B., I.S., V.W.H., T.W.-C., M.D.G.); Quantitative Sciences Unit (Y.L.G.), Department of Medicine; Department of Genetics (Y.Z., J.G., T.D.J., A.D.G., E.A.); Division of Cardiology (J.G., E.A.), Department of Medicine, Stanford University School of Medicine, CA; Neurogenomics and Informatics Center (C.Y., C.C.), Washington University School of Medicine, St. Louis, MO; Wu Tsai Neurosciences Institute (A.D.W., E.M.); Department of Psychology (A.D.W., E.M.); and Department of Epidemiology and Population Health (V.W.H.), Stanford University, CA
| | - Tanner D Jensen
- From the Department of Neurology and Neurological Sciences (A.C., Y.L.G., N.K., L.T., M.B., I.S., V.W.H., T.W.-C., M.D.G.); Quantitative Sciences Unit (Y.L.G.), Department of Medicine; Department of Genetics (Y.Z., J.G., T.D.J., A.D.G., E.A.); Division of Cardiology (J.G., E.A.), Department of Medicine, Stanford University School of Medicine, CA; Neurogenomics and Informatics Center (C.Y., C.C.), Washington University School of Medicine, St. Louis, MO; Wu Tsai Neurosciences Institute (A.D.W., E.M.); Department of Psychology (A.D.W., E.M.); and Department of Epidemiology and Population Health (V.W.H.), Stanford University, CA
| | - Chengran Yang
- From the Department of Neurology and Neurological Sciences (A.C., Y.L.G., N.K., L.T., M.B., I.S., V.W.H., T.W.-C., M.D.G.); Quantitative Sciences Unit (Y.L.G.), Department of Medicine; Department of Genetics (Y.Z., J.G., T.D.J., A.D.G., E.A.); Division of Cardiology (J.G., E.A.), Department of Medicine, Stanford University School of Medicine, CA; Neurogenomics and Informatics Center (C.Y., C.C.), Washington University School of Medicine, St. Louis, MO; Wu Tsai Neurosciences Institute (A.D.W., E.M.); Department of Psychology (A.D.W., E.M.); and Department of Epidemiology and Population Health (V.W.H.), Stanford University, CA
| | - Nandita Kasireddy
- From the Department of Neurology and Neurological Sciences (A.C., Y.L.G., N.K., L.T., M.B., I.S., V.W.H., T.W.-C., M.D.G.); Quantitative Sciences Unit (Y.L.G.), Department of Medicine; Department of Genetics (Y.Z., J.G., T.D.J., A.D.G., E.A.); Division of Cardiology (J.G., E.A.), Department of Medicine, Stanford University School of Medicine, CA; Neurogenomics and Informatics Center (C.Y., C.C.), Washington University School of Medicine, St. Louis, MO; Wu Tsai Neurosciences Institute (A.D.W., E.M.); Department of Psychology (A.D.W., E.M.); and Department of Epidemiology and Population Health (V.W.H.), Stanford University, CA
| | - Lia Talozzi
- From the Department of Neurology and Neurological Sciences (A.C., Y.L.G., N.K., L.T., M.B., I.S., V.W.H., T.W.-C., M.D.G.); Quantitative Sciences Unit (Y.L.G.), Department of Medicine; Department of Genetics (Y.Z., J.G., T.D.J., A.D.G., E.A.); Division of Cardiology (J.G., E.A.), Department of Medicine, Stanford University School of Medicine, CA; Neurogenomics and Informatics Center (C.Y., C.C.), Washington University School of Medicine, St. Louis, MO; Wu Tsai Neurosciences Institute (A.D.W., E.M.); Department of Psychology (A.D.W., E.M.); and Department of Epidemiology and Population Health (V.W.H.), Stanford University, CA
| | - Michael Belloy
- From the Department of Neurology and Neurological Sciences (A.C., Y.L.G., N.K., L.T., M.B., I.S., V.W.H., T.W.-C., M.D.G.); Quantitative Sciences Unit (Y.L.G.), Department of Medicine; Department of Genetics (Y.Z., J.G., T.D.J., A.D.G., E.A.); Division of Cardiology (J.G., E.A.), Department of Medicine, Stanford University School of Medicine, CA; Neurogenomics and Informatics Center (C.Y., C.C.), Washington University School of Medicine, St. Louis, MO; Wu Tsai Neurosciences Institute (A.D.W., E.M.); Department of Psychology (A.D.W., E.M.); and Department of Epidemiology and Population Health (V.W.H.), Stanford University, CA
| | - Ilaria Stewart
- From the Department of Neurology and Neurological Sciences (A.C., Y.L.G., N.K., L.T., M.B., I.S., V.W.H., T.W.-C., M.D.G.); Quantitative Sciences Unit (Y.L.G.), Department of Medicine; Department of Genetics (Y.Z., J.G., T.D.J., A.D.G., E.A.); Division of Cardiology (J.G., E.A.), Department of Medicine, Stanford University School of Medicine, CA; Neurogenomics and Informatics Center (C.Y., C.C.), Washington University School of Medicine, St. Louis, MO; Wu Tsai Neurosciences Institute (A.D.W., E.M.); Department of Psychology (A.D.W., E.M.); and Department of Epidemiology and Population Health (V.W.H.), Stanford University, CA
| | - Aaron D Gitler
- From the Department of Neurology and Neurological Sciences (A.C., Y.L.G., N.K., L.T., M.B., I.S., V.W.H., T.W.-C., M.D.G.); Quantitative Sciences Unit (Y.L.G.), Department of Medicine; Department of Genetics (Y.Z., J.G., T.D.J., A.D.G., E.A.); Division of Cardiology (J.G., E.A.), Department of Medicine, Stanford University School of Medicine, CA; Neurogenomics and Informatics Center (C.Y., C.C.), Washington University School of Medicine, St. Louis, MO; Wu Tsai Neurosciences Institute (A.D.W., E.M.); Department of Psychology (A.D.W., E.M.); and Department of Epidemiology and Population Health (V.W.H.), Stanford University, CA
| | - Anthony D Wagner
- From the Department of Neurology and Neurological Sciences (A.C., Y.L.G., N.K., L.T., M.B., I.S., V.W.H., T.W.-C., M.D.G.); Quantitative Sciences Unit (Y.L.G.), Department of Medicine; Department of Genetics (Y.Z., J.G., T.D.J., A.D.G., E.A.); Division of Cardiology (J.G., E.A.), Department of Medicine, Stanford University School of Medicine, CA; Neurogenomics and Informatics Center (C.Y., C.C.), Washington University School of Medicine, St. Louis, MO; Wu Tsai Neurosciences Institute (A.D.W., E.M.); Department of Psychology (A.D.W., E.M.); and Department of Epidemiology and Population Health (V.W.H.), Stanford University, CA
| | - Elizabeth Mormino
- From the Department of Neurology and Neurological Sciences (A.C., Y.L.G., N.K., L.T., M.B., I.S., V.W.H., T.W.-C., M.D.G.); Quantitative Sciences Unit (Y.L.G.), Department of Medicine; Department of Genetics (Y.Z., J.G., T.D.J., A.D.G., E.A.); Division of Cardiology (J.G., E.A.), Department of Medicine, Stanford University School of Medicine, CA; Neurogenomics and Informatics Center (C.Y., C.C.), Washington University School of Medicine, St. Louis, MO; Wu Tsai Neurosciences Institute (A.D.W., E.M.); Department of Psychology (A.D.W., E.M.); and Department of Epidemiology and Population Health (V.W.H.), Stanford University, CA
| | - Victor W Henderson
- From the Department of Neurology and Neurological Sciences (A.C., Y.L.G., N.K., L.T., M.B., I.S., V.W.H., T.W.-C., M.D.G.); Quantitative Sciences Unit (Y.L.G.), Department of Medicine; Department of Genetics (Y.Z., J.G., T.D.J., A.D.G., E.A.); Division of Cardiology (J.G., E.A.), Department of Medicine, Stanford University School of Medicine, CA; Neurogenomics and Informatics Center (C.Y., C.C.), Washington University School of Medicine, St. Louis, MO; Wu Tsai Neurosciences Institute (A.D.W., E.M.); Department of Psychology (A.D.W., E.M.); and Department of Epidemiology and Population Health (V.W.H.), Stanford University, CA
| | - Tony Wyss-Coray
- From the Department of Neurology and Neurological Sciences (A.C., Y.L.G., N.K., L.T., M.B., I.S., V.W.H., T.W.-C., M.D.G.); Quantitative Sciences Unit (Y.L.G.), Department of Medicine; Department of Genetics (Y.Z., J.G., T.D.J., A.D.G., E.A.); Division of Cardiology (J.G., E.A.), Department of Medicine, Stanford University School of Medicine, CA; Neurogenomics and Informatics Center (C.Y., C.C.), Washington University School of Medicine, St. Louis, MO; Wu Tsai Neurosciences Institute (A.D.W., E.M.); Department of Psychology (A.D.W., E.M.); and Department of Epidemiology and Population Health (V.W.H.), Stanford University, CA
| | - Euan Ashley
- From the Department of Neurology and Neurological Sciences (A.C., Y.L.G., N.K., L.T., M.B., I.S., V.W.H., T.W.-C., M.D.G.); Quantitative Sciences Unit (Y.L.G.), Department of Medicine; Department of Genetics (Y.Z., J.G., T.D.J., A.D.G., E.A.); Division of Cardiology (J.G., E.A.), Department of Medicine, Stanford University School of Medicine, CA; Neurogenomics and Informatics Center (C.Y., C.C.), Washington University School of Medicine, St. Louis, MO; Wu Tsai Neurosciences Institute (A.D.W., E.M.); Department of Psychology (A.D.W., E.M.); and Department of Epidemiology and Population Health (V.W.H.), Stanford University, CA
| | - Carlos Cruchaga
- From the Department of Neurology and Neurological Sciences (A.C., Y.L.G., N.K., L.T., M.B., I.S., V.W.H., T.W.-C., M.D.G.); Quantitative Sciences Unit (Y.L.G.), Department of Medicine; Department of Genetics (Y.Z., J.G., T.D.J., A.D.G., E.A.); Division of Cardiology (J.G., E.A.), Department of Medicine, Stanford University School of Medicine, CA; Neurogenomics and Informatics Center (C.Y., C.C.), Washington University School of Medicine, St. Louis, MO; Wu Tsai Neurosciences Institute (A.D.W., E.M.); Department of Psychology (A.D.W., E.M.); and Department of Epidemiology and Population Health (V.W.H.), Stanford University, CA
| | - Michael D Greicius
- From the Department of Neurology and Neurological Sciences (A.C., Y.L.G., N.K., L.T., M.B., I.S., V.W.H., T.W.-C., M.D.G.); Quantitative Sciences Unit (Y.L.G.), Department of Medicine; Department of Genetics (Y.Z., J.G., T.D.J., A.D.G., E.A.); Division of Cardiology (J.G., E.A.), Department of Medicine, Stanford University School of Medicine, CA; Neurogenomics and Informatics Center (C.Y., C.C.), Washington University School of Medicine, St. Louis, MO; Wu Tsai Neurosciences Institute (A.D.W., E.M.); Department of Psychology (A.D.W., E.M.); and Department of Epidemiology and Population Health (V.W.H.), Stanford University, CA
| |
Collapse
|
15
|
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
|
16
|
Meda F, Simrén J, Borroni B, Cantoni V, Archetti S, Biasiotto G, Andreasson U, Blennow K, Kvartsberg H, Zetterberg H. Analytical and clinical validation of a blood progranulin ELISA in frontotemporal dementias. Clin Chem Lab Med 2023; 61:2195-2204. [PMID: 37476993 PMCID: PMC10598571 DOI: 10.1515/cclm-2023-0562] [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: 05/29/2023] [Accepted: 06/29/2023] [Indexed: 07/22/2023]
Abstract
OBJECTIVES Heterozygous mutations in the granulin (GRN) gene may result in haploinsufficiency of progranulin (PGRN), which might lead to frontotemporal dementia (FTD). In this study, we aimed to perform analytical and clinical validation of a commercial progranulin kit for clinical use. METHODS Analytical validation parameters including assay precision, selectivity, measurement range, dilution linearity, interferences and sample stability were tested according to previously described procedures. For clinical validation, PGRN levels were measured in plasma from 32 cognitively healthy individuals, 52 confirmed GRN mutation carriers, 25 C9orf72 mutation carriers and 216 patients with different neurodegenerative diseases of which 70 were confirmed as non-mutation carriers. RESULTS Among the analytical validation parameters, assay precision and repeatability were very stable (coefficients of variation <7 %). Spike recovery was 96 %, the measurement range was 6.25-400 μg/L and dilution linearity ranged from 1:50-1:200. Hemolysis did not interfere with progranulin levels, and these were resistant to freeze/thaw cycles and storage at different temperatures. For the clinical validation, the assay was capable of distinguishing GRN mutation carriers from controls and non-GRN mutation carriers with very good sensitivity and specificity at a cut-off of 57 μg/L (97 %, 100 %, respectively). CONCLUSIONS In this study, we demonstrate robust analytical and diagnostic performance of this commercial progranulin kit for implementation in clinical laboratory practice. This easy-to-use test allows identification of potential GRN mutation carriers, which may guide further evaluation of the patient. This assay might also be used to evaluate the effect of novel PGRN-targeting drugs and therapies.
Collapse
Affiliation(s)
- Francisco Meda
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Joel Simrén
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Barbara Borroni
- Centre for Neurodegenerative Disorders, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Valentina Cantoni
- Centre for Neurodegenerative Disorders, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Silvana Archetti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Giorgio Biasiotto
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Ulf Andreasson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Hlin Kvartsberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, HKCeND, 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
|
17
|
Kaplelach AK, Fox SN, Cook AK, Hall JA, Dannemiller RS, Jaunarajs KL, Arrant AE. Regulation of extracellular progranulin in medial prefrontal cortex. Neurobiol Dis 2023; 188:106326. [PMID: 37838007 PMCID: PMC10682954 DOI: 10.1016/j.nbd.2023.106326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/28/2023] [Accepted: 10/11/2023] [Indexed: 10/16/2023] Open
Abstract
Progranulin is a secreted pro-protein that has anti-inflammatory and neurotrophic effects and is necessary for maintaining lysosomal function. Mutations in progranulin (GRN) are a major cause of frontotemporal dementia. Most pathogenic GRN mutations cause progranulin haploinsufficiency, so boosting progranulin levels is a promising therapeutic strategy. Progranulin is constitutively secreted, then taken up and trafficked to lysosomes. Before being taken up from the extracellular space, progranulin interacts with receptors that may mediate anti-inflammatory and growth factor-like effects. Modifying progranulin trafficking is a viable approach to boosting progranulin, but progranulin secretion and uptake by cells in the brain is poorly understood and may involve distinct mechanisms from other parts of the body. Understanding the cell types and processes that regulate extracellular progranulin in the brain could provide insight into progranulin's mechanism of action and inform design of progranulin-boosting therapies. To address this question we used microdialysis to measure progranulin in interstitial fluid (ISF) of mouse medial prefrontal cortex (mPFC). Grn+/- mice had approximately 50% lower ISF progranulin than wild-type mice, matching the reduction of progranulin in cortical tissue. Fluorescent in situ hybridization and immunofluorescence confirmed that microglia and neurons are the major progranulin-expressing cell types in the mPFC. Studies of conditional microglial (Mg-KO) and neuronal (N-KO) Grn knockout mice revealed that loss of progranulin from either cell type results in approximately 50% reduction in ISF progranulin. LPS injection (i.p.) produced an acute increase in ISF progranulin in mPFC. Depolarizing cells with KCl increased ISF progranulin, but this response was not altered in N-KO mice, indicating progranulin secretion by non-neuronal cells. Increasing neuronal activity with picrotoxin did not increase ISF progranulin. These data indicate that microglia and neurons are the source of most ISF progranulin in mPFC, with microglia likely secreting more progranulin per cell than neurons. The acute increase in ISF progranulin after LPS treatment is consistent with a role for extracellular progranulin in regulating inflammation, and may have been driven by microglia or peripheral immune cells. Finally, these data indicate that mPFC neurons engage in constitutive progranulin secretion that is not acutely changed by neuronal activity.
Collapse
Affiliation(s)
- Azariah K Kaplelach
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, Departments of Neurology and Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Stephanie N Fox
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, Departments of Neurology and Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anna K Cook
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, Departments of Neurology and Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Justin A Hall
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, Departments of Neurology and Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ryan S Dannemiller
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, Departments of Neurology and Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Karen L Jaunarajs
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, Departments of Neurology and Neurobiology, 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, Departments of Neurology and Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
18
|
Wauters E, Gossye H, Frydas A, Sieben A, Van Broeckhoven C. Rare exonic variant affects GRN splicing and contributes to frontotemporal lobar degeneration. Neurobiol Aging 2023; 130:61-69. [PMID: 37459659 DOI: 10.1016/j.neurobiolaging.2023.06.009] [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: 09/05/2022] [Revised: 05/20/2023] [Accepted: 06/10/2023] [Indexed: 08/13/2023]
Abstract
Heterozygous loss-of-function (LOF) mutations in the progranulin gene (GRN) cause frontotemporal lobar degeneration (FTLD) by a mechanism of haploinsufficiency. For most missense mutations, the contribution to FTLD is however unclear. We studied the pathogenicity of rare GRN missense mutations using patient biomaterials. We identified a new mutation in GRN, c.1178 A>C, in a patient with a diagnosis of primary progressive aphasia. Neuropathological examination of autopsied brain showed FTLD with TAR DNA-binding protein 43 (FTLD-TDP) type A pathology with concomitant Alzheimer's disease pathology. Serum progranulin protein levels were reduced to levels comparable to known LOF mutations. The mutation is in the last codon of exon 10, in the splice donor sequence. Our data provide evidence that the mutation leads to aberrant splicing, resulting in a frameshift (p.(Glu393AlafsTer31)) and consequently nonsense-mediated mRNA decay. Our finding demonstrates that carefully examining sequencing data around splice sites is needed since this mutation was annotated as a missense mutation. Unraveling the pathogenicity of variants of unknown significance is important for clinical diagnosis and genetic counseling.
Collapse
Affiliation(s)
- Eline Wauters
- VIB Center for Molecular Neurology, Antwerp, Belgium; Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Helena Gossye
- VIB Center for Molecular Neurology, Antwerp, Belgium; Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium; Department of Neurology, Antwerp University Hospital, Edegem, Belgium
| | - Alexandros Frydas
- VIB Center for Molecular Neurology, Antwerp, Belgium; Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Anne Sieben
- Departement of Pathology, Antwerp University Hospital, Edegem, Belgium
| | - Christine Van Broeckhoven
- VIB Center for Molecular Neurology, Antwerp, Belgium; Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
19
|
Ghirelli A, Spinelli EG, Canu E, Domi T, Basaia S, Castelnovo V, Pozzi L, Magnani G, Caso F, Caroppo P, Prioni S, Villa C, Riva N, Quattrini A, Carrera P, Filippi M, Agosta F. Case report: coexistence of C9orf72 expansion and progranulin mutation in a case of genetic frontotemporal dementia-clinical features and neuroimaging correlates. J Neurol 2023; 270:5102-5109. [PMID: 37382630 PMCID: PMC10511558 DOI: 10.1007/s00415-023-11839-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 06/30/2023]
Affiliation(s)
- Alma Ghirelli
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Edoardo Gioele Spinelli
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy
- Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elisa Canu
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy
| | - Teuta Domi
- Experimental Neuropathology Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Basaia
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy
| | - Veronica Castelnovo
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy
| | - Laura Pozzi
- Experimental Neuropathology Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giuseppe Magnani
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Caso
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paola Caroppo
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Unit of Neurology 5-Neuropathology, Milan, Italy
| | - Sara Prioni
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Unit of Neurology 5-Neuropathology, Milan, Italy
| | - Cristina Villa
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Unit of Neurology 5-Neuropathology, Milan, Italy
| | - Nilo Riva
- Experimental Neuropathology Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Angelo Quattrini
- Experimental Neuropathology Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paola Carrera
- Laboratory of Clinical Molecular Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
- Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Federica Agosta
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy.
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
20
|
Kurnellas M, Mitra A, Schwabe T, Paul R, Arrant AE, Roberson ED, Ward M, Yeh F, Long H, Rosenthal A. Latozinemab, a novel progranulin-elevating therapy for frontotemporal dementia. J Transl Med 2023; 21:387. [PMID: 37322482 PMCID: PMC10268535 DOI: 10.1186/s12967-023-04251-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 06/08/2023] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND Heterozygous loss-of-function mutations in the progranulin (PGRN) gene (GRN) cause a reduction in PGRN and lead to the development of frontotemporal dementia (FTD-GRN). PGRN is a secreted lysosomal chaperone, immune regulator, and neuronal survival factor that is shuttled to the lysosome through multiple receptors, including sortilin. Here, we report the characterization of latozinemab, a human monoclonal antibody that decreases the levels of sortilin, which is expressed on myeloid and neuronal cells and shuttles PGRN to the lysosome for degradation, and blocks its interaction with PGRN. METHODS In vitro characterization studies were first performed to assess the mechanism of action of latozinemab. After the in vitro studies, a series of in vivo studies were performed to assess the efficacy of a mouse-cross reactive anti-sortilin antibody and the pharmacokinetics, pharmacodynamics, and safety of latozinemab in nonhuman primates and humans. RESULTS In a mouse model of FTD-GRN, the rodent cross-reactive anti-sortilin antibody, S15JG, decreased total sortilin levels in white blood cell (WBC) lysates, restored PGRN to normal levels in plasma, and rescued a behavioral deficit. In cynomolgus monkeys, latozinemab decreased sortilin levels in WBCs and concomitantly increased plasma and cerebrospinal fluid (CSF) PGRN by 2- to threefold. Finally, in a first-in-human phase 1 clinical trial, a single infusion of latozinemab caused a reduction in WBC sortilin, tripled plasma PGRN and doubled CSF PGRN in healthy volunteers, and restored PGRN to physiological levels in asymptomatic GRN mutation carriers. CONCLUSIONS These findings support the development of latozinemab for the treatment of FTD-GRN and other neurodegenerative diseases where elevation of PGRN may be beneficial. Trial registration ClinicalTrials.gov, NCT03636204. Registered on 17 August 2018, https://clinicaltrials.gov/ct2/show/NCT03636204 .
Collapse
Affiliation(s)
- Michael Kurnellas
- Alector, Inc., 131 Oyster Point Blvd, #600, South San Francisco, CA, 94080, USA.
- Neuron23, South San Francisco, CA, 94080, USA.
| | - Ananya Mitra
- Alector, Inc., 131 Oyster Point Blvd, #600, South San Francisco, CA, 94080, USA
| | - Tina Schwabe
- Alector, Inc., 131 Oyster Point Blvd, #600, South San Francisco, CA, 94080, USA
- Nine Square Therapeutics, Inc., South San Francisco, CA, 94080, USA
| | - Robert Paul
- Alector, Inc., 131 Oyster Point Blvd, #600, South San Francisco, CA, 94080, USA
- Nine Square Therapeutics, Inc., South San Francisco, CA, 94080, USA
| | - Andrew E Arrant
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, and Evelyn F. McKnight Brain Institute, Departments of Neurology and Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Erik D Roberson
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, and Evelyn F. McKnight Brain Institute, Departments of Neurology and Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Michael Ward
- Alector, Inc., 131 Oyster Point Blvd, #600, South San Francisco, CA, 94080, USA
| | - Felix Yeh
- Alector, Inc., 131 Oyster Point Blvd, #600, South San Francisco, CA, 94080, USA
- Genentech, South San Francisco, CA, 94080, USA
| | - Hua Long
- Alector, Inc., 131 Oyster Point Blvd, #600, South San Francisco, CA, 94080, USA
| | - Arnon Rosenthal
- Alector, Inc., 131 Oyster Point Blvd, #600, South San Francisco, CA, 94080, USA
| |
Collapse
|
21
|
Davis SE, Cook AK, Hall JA, Voskobiynyk Y, Carullo NV, Boyle NR, Hakim AR, Anderson KM, Hobdy KP, Pugh DA, Murchison CF, McMeekin LJ, Simmons M, Margolies KA, Cowell RM, Nana AL, Spina S, Grinberg LT, Miller BL, Seeley WW, Arrant AE. Patients with sporadic FTLD exhibit similar increases in lysosomal proteins and storage material as patients with FTD due to GRN mutations. Acta Neuropathol Commun 2023; 11:70. [PMID: 37118844 PMCID: PMC10148425 DOI: 10.1186/s40478-023-01571-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 04/30/2023] Open
Abstract
Loss of function progranulin (GRN) mutations are a major autosomal dominant cause of frontotemporal dementia (FTD). Patients with FTD due to GRN mutations (FTD-GRN) develop frontotemporal lobar degeneration with TDP-43 pathology type A (FTLD-TDP type A) and exhibit elevated levels of lysosomal proteins and storage material in frontal cortex, perhaps indicating lysosomal dysfunction as a mechanism of disease. To investigate whether patients with sporadic FTLD exhibit similar signs of lysosomal dysfunction, we compared lysosomal protein levels, transcript levels, and storage material in patients with FTD-GRN or sporadic FTLD-TDP type A. We analyzed samples from frontal cortex, a degenerated brain region, and occipital cortex, a relatively spared brain region. In frontal cortex, patients with sporadic FTLD-TDP type A exhibited similar increases in lysosomal protein levels, transcript levels, and storage material as patients with FTD-GRN. In occipital cortex of both patient groups, most lysosomal measures did not differ from controls. Frontal cortex from a transgenic mouse model of TDP-opathy had similar increases in cathepsin D and lysosomal storage material, showing that TDP-opathy and neurodegeneration can drive these changes independently of progranulin. To investigate these changes in additional FTLD subtypes, we analyzed frontal cortical samples from patients with sporadic FTLD-TDP type C or Pick's disease, an FTLD-tau subtype. All sporadic FTLD groups had similar increases in cathepsin D activity, lysosomal membrane proteins, and storage material as FTD-GRN patients. However, patients with FTLD-TDP type C or Pick's disease did not have similar increases in lysosomal transcripts as patients with FTD-GRN or sporadic FTLD-TDP type A. Based on these data, accumulation of lysosomal proteins and storage material may be a common aspect of end-stage FTLD. However, the unique changes in gene expression in patients with FTD-GRN or sporadic FTLD-TDP type A may indicate distinct underlying lysosomal changes among FTLD subtypes.
Collapse
Affiliation(s)
- Skylar E Davis
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anna K Cook
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Justin A Hall
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yuliya Voskobiynyk
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nancy V Carullo
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nicholas R Boyle
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ahmad R Hakim
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kristian M Anderson
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kierra P Hobdy
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Derian A Pugh
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Charles F Murchison
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Laura J McMeekin
- Department of Neuroscience, Southern Research, Birmingham, AL, USA
| | - Micah Simmons
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Neuroscience, Southern Research, Birmingham, AL, USA
| | | | - Rita M Cowell
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Neuroscience, Southern Research, Birmingham, AL, USA
| | - Alissa L Nana
- Department of Neurology, Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Salvatore Spina
- Department of Neurology, Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Lea T Grinberg
- Department of Neurology, Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Bruce L Miller
- Department of Neurology, Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - William W Seeley
- Department of Neurology, Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Andrew E Arrant
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, USA.
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
22
|
Tan YJ, Yong ACW, Foo JN, Lian MM, Lim WK, Dominguez J, Fong ZH, Narasimhalu K, Chiew HJ, Ng KP, Ting SKS, Kandiah N, Ng ASL. C9orf72 expansions are the most common cause of genetic frontotemporal dementia in a Southeast Asian cohort. Ann Clin Transl Neurol 2023; 10:568-578. [PMID: 36799407 PMCID: PMC10109321 DOI: 10.1002/acn3.51744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 02/03/2023] [Accepted: 02/04/2023] [Indexed: 02/18/2023] Open
Abstract
OBJECTIVE Frontotemporal dementia (FTD) encompasses a spectrum of neurodegenerative disorders, including behavioural variant FTD (bvFTD), semantic variant primary progressive aphasia (svPPA) and non-fluent variant PPA (nfvPPA). While a strong genetic component is implicated in FTD, genetic FTD in Asia is less frequently reported. We aimed to investigate the frequency of Southeast Asian FTD patients harbouring known genetic FTD variants. METHODS A total of 60 FTD-spectrum patients (25 familial and 35 sporadic) from Singapore and the Philippines were included. All underwent next-generation sequencing and repeat-primed PCR for C9orf72 expansion testing. Neurofilament light chain (NfL) levels were measured in a subset of patients. RESULTS Overall, 26.6% (16/60 cases) carried pathogenic or likely pathogenic variants in a FTD-related gene, including: MAPT Gln351Arg (n = 1); GRN Cys92Ter (n = 1), Ser301Ter (n = 2), c.462 + 1G > C (n = 1); C9orf72 expansion (35-70 repeats; n = 8); TREM2 Arg47Cys (n = 1); and OPTN frameshift insertion (n = 2). Genetic mutations accounted for 48% (12/25) of patients with familial FTD, and 11.4% (4/35) of patients with sporadic FTD. C9orf72 repeat expansions were the most common genetic mutation (13.3%, 8/60), followed by GRN (6.7%, 4/60) variants. Within mutation carriers, plasma NfL was highest in a C9orf72 expansion carrier, and CSF NfL was highest in a GRN splice variant carrier. INTERPRETATION In our cohort, genetic mutations are present in one-quarter of FTD-spectrum cases, and up to half of those with family history. Our findings highlight the importance of wider implementation of genetic testing in FTD patients from Southeast Asia.
Collapse
Affiliation(s)
- Yi Jayne Tan
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore, Singapore
| | - Alisa C W Yong
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore, Singapore
| | - Jia Nee Foo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.,Human Genetics, Genome Institute of Singapore, A*STAR, Singapore, Singapore
| | - Michelle M Lian
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Weng Khong Lim
- Singhealth Duke-NUS Institute of Precision Medicine, Singapore, Singapore.,Cancer & Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore
| | | | - Zhi Hui Fong
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore, Singapore
| | - Kaavya Narasimhalu
- Singhealth Duke-NUS Institute of Precision Medicine, Singapore, Singapore.,Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Singapore, Singapore
| | - Hui Jin Chiew
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore, Singapore
| | - Kok Pin Ng
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore, Singapore
| | - Simon K S Ting
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Singapore, Singapore
| | - Nagaendran Kandiah
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore, Singapore
| | - Adeline S L Ng
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore, Singapore.,Neuroscience and Behavioural Disorders Programme, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
23
|
Ingannato A, Bessi V, Chiari A, Salvatori D, Bagnoli S, Bedin R, Ferrari C, Sorbi S, Nacmias B. GRN Missense Variants and Familial Alzheimer's Disease: Two Case Reports. J Alzheimers Dis 2023; 96:767-775. [PMID: 37899057 DOI: 10.3233/jad-230689] [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: 10/31/2023]
Abstract
BACKGROUND Progranulin protein (GRN) is a growth factor, encoded by the GRN (Granulin precursor) gene, involved in several functions including inflammation, wound repair, signal transduction, proliferation, and tumorigenesis. Mutations in GRN gene are usually the genetic etiology of frontotemporal dementia (FTD), but different studies reported GRN mutations in Alzheimer 's disease (AD) patients. OBJECTIVE Here, we analyzed FTD linked gene GRN in 23 patients with a clinical diagnosis of AD and a family history of AD (FAD), not carrying mutations in AD candidate genes (PSEN 1, PSEN 2, and APP). In addition, Microtubule-associated protein tau (MAPT) gene was studied too. All patients underwent an extensive neuropsychological battery. METHODS Genetic analyses were performed thought PCR assay and sequencing. Variants were annotated with ANNOVAR and allele frequency was checked on population databases. In silico prediction tools were consulted to check nonsynonymous variants and their effect on protein function and structure. The clinical data were retrospectively collected from medical records. RESULTS Genetic screening of MAPT and GRN in 23 FAD patients highlighted two rare different variants in two probands (2/23 = 8,7%) located in GRN gene: R433W (p.Arg433Trp) and C521Y (p.Cys521Tyr). The R433W and C521Y are variants with uncertain significant, that are predicted to affect GRN protein structure and function, with a possible damaging effect. CONCLUSIONS Our data provide evidence of the importance of GRN genetic analysis also in the study of familial AD.
Collapse
Affiliation(s)
- Assunta Ingannato
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Valentina Bessi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Annalisa Chiari
- U.O. Neurologia, Azienda Ospedaliero Universitaria di Modena, Modena, Italy
| | - Davide Salvatori
- Department of Biomedical, Metabolic, and Neural Science, University of Modena and Reggio Emilia, Modena, Italy
| | - Silvia Bagnoli
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Roberta Bedin
- U.O. Neurologia, Azienda Ospedaliero Universitaria di Modena, Modena, Italy
| | - Camilla Ferrari
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Sandro Sorbi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
- IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
- IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| |
Collapse
|
24
|
Farvadi F, Hashemi F, Amini A, Alsadat Vakilinezhad M, Raee MJ. Early Diagnosis of Alzheimer's Disease with Blood Test; Tempting but Challenging. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2023; 12:172-210. [PMID: 38313372 PMCID: PMC10837916 DOI: 10.22088/ijmcm.bums.12.2.172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 11/25/2023] [Accepted: 12/13/2023] [Indexed: 02/06/2024]
Abstract
The increasing prevalence of Alzheimer's disease (AD) has led to a health crisis. According to official statistics, more than 55 million people globally have AD or other types of dementia, making it the sixth leading cause of death. It is still difficult to diagnose AD and there is no definitive diagnosis yet; post-mortem autopsy is still the only definite method. Moreover, clinical manifestations occur very late in the course of disease progression; therefore, profound irreversible changes have already occurred when the disease manifests. Studies have shown that in the preclinical stage of AD, changes in some biomarkers are measurable prior to any neurological damage or other symptoms. Hence, creating a reliable, fast, and affordable method capable of detecting AD in early stage has attracted the most attention. Seeking clinically applicable, inexpensive, less invasive, and much more easily accessible biomarkers for early diagnosis of AD, blood-based biomarkers (BBBs) seem to be an ideal option. This review is an inclusive report of BBBs that have been shown to be altered in the course of AD progression. The aim of this report is to provide comprehensive insight into the research status of early detection of AD based on BBBs.
Collapse
Affiliation(s)
- Fakhrossadat Farvadi
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Hashemi
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, the University of Newcastle, Newcastle, Australia
| | - Azadeh Amini
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical sciences, Tehran, Iran
| | | | - Mohammad Javad Raee
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
25
|
BILGIN B, GULER M, CICEK H, URFALIOGLU S, KOKUSARI G, MARANGOZOGLU SAHIN B. Searching for Biomarkers in Proliferative Diabetic Retinopathy: Amphiregulin and Progranulin. Medeni Med J 2022; 37:327-331. [PMID: 36578150 PMCID: PMC9808851 DOI: 10.4274/mmj.galenos.2022.10270] [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] [Indexed: 11/06/2022] Open
Abstract
Objective Diabetic retinopathy is a common diabetic microvascular problem. Its diagnosis and classification are based on visible changes in clinical fundus examination. However, the discovery of possible vitreous biomarkers in patients with proliferative and nonproliferative diabetic retinopathy may guide both the differentiation and degree of retinopathy. Biomarkers that will be accepted can be also a treatment target. Amphiregulin (AREG) promotes proliferative and regenerative activity and repairs most cell types by binding and activating epidermal growth factor receptors. Progranulin (PGRN) has complex functions in many physiological and pathological processes. Thus, this study aimed to report vitreous AREG and PGRN levels in patients with diabetes and proliferative retinopathy and compare the results with those without diabetes. Methods Thirty-three eyes of 33 patients with proliferative diabetic retinopathy and 31 eyes of 31 patients without diabetes were included in this study. Vitreous humor samples were collected from all patients at the time of pars plana vitrectomy surgery immediately before the surgical procedure. Vitreous AREG and PGRN values were determined by the ELISA method. Results The mean AREG and PGRN values were similar in the groups (p=0.427, p=0.459, respectively). Conclusions The results demonstrated that vitreous AREG and PGRN levels have no significant relationship with proliferative diabetic retinopathy.
Collapse
Affiliation(s)
- Burak BILGIN
- Kahramanmaras Sutcu Imam University Faculty of Medicine, Department of Ophthalmology, Kahramanmaras, Turkey
| | - Mete GULER
- Kahramanmaras Sutcu Imam University Faculty of Medicine, Department of Ophthalmology, Kahramanmaras, Turkey
| | - Hulya CICEK
- Gaziantep University Faculty of Medicine, Department of Medical Biochemistry, Gaziantep, Turkey,* Address for Correspondence: Gaziantep University Faculty of Medicine, Department of Medical Biochemistry, Gaziantep, Turkey E-mail:
| | - Selma URFALIOGLU
- Kahramanmaras Sutcu Imam University Faculty of Medicine, Department of Ophthalmology, Kahramanmaras, Turkey
| | - Gokhan KOKUSARI
- Kahramanmaras Sutcu Imam University Faculty of Medicine, Department of Ophthalmology, Kahramanmaras, Turkey
| | - Bedia MARANGOZOGLU SAHIN
- Kahramanmaras Sutcu Imam University Faculty of Medicine, Department of Ophthalmology, Kahramanmaras, Turkey
| |
Collapse
|
26
|
Cerebrospinal Fluid Biomarker Profile in TDP-43-Related Genetic Frontotemporal Dementia. J Pers Med 2022; 12:jpm12101747. [PMID: 36294886 PMCID: PMC9605286 DOI: 10.3390/jpm12101747] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/06/2022] [Accepted: 10/14/2022] [Indexed: 11/16/2022] Open
Abstract
Cerebrospinal fluid (CSF) biomarkers, namely total tau, phospho-tau and amyloid beta peptides, have received much attention specifically regarding Alzheimer’s disease (AD), since they can detect the biochemical fingerprint of AD and serve as a diagnostic tool for accurate and early diagnosis during life. In the same way, biomarkers for other neurodegenerative disease pathologies are also needed. We present a case series of six patients with genetic frontotemporal dementia (FTD), with TDP-43 underlying proteinopathy, in an attempt to assess TDP-43 as a novel biomarker alone and in combination with established AD biomarkers for this specific patient group, based on the principles of personalized and precision medicine. Our results indicate that genetic TDP-43-FTD is characterized by increased CSF TPD-43 and increased TDP-43 × τΤ/τP-181 combination. Hence, TDP-43 combined with tau proteins could be a useful tool for the diagnosis of genetic FTD with TDP-43 underling histopathology, supplementing clinical, neuropsychological and imaging data.
Collapse
|
27
|
Recent Advances in Frontotemporal Dementia. Neurol Sci 2022:1-10. [DOI: 10.1017/cjn.2022.69] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
28
|
Tanaka Y, Kusumoto SY, Honma Y, Takeya K, Eto M. Overexpression of progranulin increases pathological protein accumulation by suppressing autophagic flux. Biochem Biophys Res Commun 2022; 611:78-84. [PMID: 35483222 DOI: 10.1016/j.bbrc.2022.04.064] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 04/13/2022] [Indexed: 01/01/2023]
Abstract
Progranulin (PGRN) haploinsufficiency from autosomal dominant mutations in the PGRN gene causes frontotemporal lobar degeneration, which is characterized by cytoplasmic inclusions predominantly containing TDP-43 (FTLD-TDP). PGRN supplementation for patients with a PGRN gene mutation has recently been proposed as a therapeutic strategy to suppress FTLD-TDP. However, it currently remains unclear whether excessive amounts of PGRN are beneficial or harmful. We herein report the effects of PGRN overexpression on autophagic flux in a cultured cell model. PGRN overexpression increased the level of an autophagosome marker without promoting autophagosome formation and decreased the signal intensity of an autolysosome marker, indicating the suppression of autophagic flux due to reductions in the formation of autolysosomes. Assessments of lysosome numbers and biogenesis using LysoTracker and cells stably expressing TFEB-GFP, respectively, indicated that PGRN overexpression increased the lysosome numbers without lysosomal biogenesis. These results suggest that PGRN overexpression suppressed autophagic flux by inhibiting autophagosome-lysosome fusion. Moreover, PGRN overexpression enhanced polyglutamine aggregation and aggregate-prone TDP-43 accumulation, indicating that the suppression of autophagic flux by excessive amounts of PGRN worsens the pathology of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yoshinori Tanaka
- Biochemistry Unit, Faculty of Veterinary Medicine, Okayama University of Science, Imabari-shi, Ehime, Japan.
| | - Shun-Ya Kusumoto
- Biochemistry Unit, Faculty of Veterinary Medicine, Okayama University of Science, Imabari-shi, Ehime, Japan
| | - Yuki Honma
- Biochemistry Unit, Faculty of Veterinary Medicine, Okayama University of Science, Imabari-shi, Ehime, Japan
| | - Kosuke Takeya
- Biochemistry Unit, Faculty of Veterinary Medicine, Okayama University of Science, Imabari-shi, Ehime, Japan
| | - Masumi Eto
- Biochemistry Unit, Faculty of Veterinary Medicine, Okayama University of Science, Imabari-shi, Ehime, Japan
| |
Collapse
|
29
|
Amin S, Carling G, Gan L. New insights and therapeutic opportunities for progranulin-deficient frontotemporal dementia. Curr Opin Neurobiol 2022; 72:131-139. [PMID: 34826653 DOI: 10.1016/j.conb.2021.10.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 10/19/2021] [Indexed: 01/02/2023]
Abstract
Frontotemporal dementia (FTD) is the second most common form of dementia. It affects the frontal and temporal lobes of the brain and has a highly heterogeneous clinical representation with patients presenting with a wide range of behavioral, language, and executive dysfunctions. Etiology of FTD is complex and consists of both familial and sporadic cases. Heterozygous mutations in the GRN gene, resulting in GRN haploinsufficiency, cause progranulin (PGRN)-deficient FTD characterized with cytoplasmic mislocalization of TAR DNA-binding protein 43 kDa (TDP-43) aggregates. GRN codes for PGRN, a secreted protein that is also localized in the endolysosomes and plays a critical role in regulating lysosomal homeostasis. How PGRN deficiency modulates immunity and causes TDP-43 pathology and FTD-related neurodegeneration remains an active area of intense investigation. In the current review, we discuss some of the significant progress made in the past two years that links PGRN deficiency with microglial-associated neuroinflammation, TDP-43 pathology, and lysosomal dysfunction. We also review the opportunities and challenges toward developing therapies and biomarkers to treat PGRN-deficient FTD.
Collapse
Affiliation(s)
- Sadaf Amin
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Gillian Carling
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA; Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Li Gan
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA.
| |
Collapse
|
30
|
Jung NY, Kim HS, Kim ES, Jeon S, Lee MJ, Pak K, Lee JH, Lee YM, Lee K, Shin JH, Ko JK, Lee JM, Yoon JA, Hwang C, Choi KU, Huh GY, Kim YE, Kim EJ. Serum progranulin is not associated with rs5848 polymorphism in Korean patients with neurodegenerative diseases. PLoS One 2022; 17:e0261007. [PMID: 35085262 PMCID: PMC8794169 DOI: 10.1371/journal.pone.0261007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 11/22/2021] [Indexed: 12/02/2022] Open
Abstract
Low serum progranulin (PGRN) is known to be associated with granulin (GRN) gene mutation and T alleles of GRN rs5848 polymorphism. However, there have been only a few Asian studies exploring these. We investigated the serum PGRN levels, rs5848 genotypes, and their relations with cerebrospinal fluid (CSF) Alzheimer’s disease (AD) biomarkers in the Korean population. Serum PGRN levels, GRN rs5848 polymorphism, and GRN mutations were evaluated in 239 participants (22 cognitively unimpaired participants and 217 patients with neurodegenerative diseases). CSF AD biomarkers were also evaluated in 214 participants. There was no significant difference in the serum PGRN levels among the diagnostic groups. We could not find any GRN mutation carrier in our sample. The differences in the frequencies of the rs5848 genotypes among the clinical groups or the effects of the rs5848 genotypes on serum PGRN were not observed. There was no correlation between the serum PGRN level or rs5848 genotype and CSF AD biomarkers. Neither the T allele nor the TT genotype had an effect on the development of AD. Our results showed that serum PGRN levels were not associated with rs5848 genotypes, indicating that multiple single nucleotide polymorphisms might affect PGRN concentrations in an ethnicity-specific manner.
Collapse
Affiliation(s)
- Na-Yeon Jung
- Department of Neurology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, Republic of Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Hyang-Sook Kim
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Eun Soo Kim
- Department of Anesthesia and Pain Medicine, School of Medicine, Pusan National University, Busan, Republic of Korea
| | - Sumin Jeon
- Department of Neurology, Pusan National University Hospital, Pusan National University School of Medicine and Medical Research Institute, Busan, Republic of Korea
| | - Myung Jun Lee
- Department of Neurology, Pusan National University Hospital, Pusan National University School of Medicine and Medical Research Institute, Busan, Republic of Korea
| | - Kyoungjune Pak
- Department of Nuclear Medicine, Pusan National University Hospital, Pusan National University School of Medicine, Busan, Republic of Korea
| | - Jae-Hyeok Lee
- Department of Neurology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, Republic of Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Young Min Lee
- Department of Psychiatry, Pusan National University Hospital, Pusan National University School of Medicine, Busan, Republic of Korea
| | - Kangyoon Lee
- Department of Psychiatry, Pusan National University Hospital, Pusan National University School of Medicine, Busan, Republic of Korea
| | - Jin-Hong Shin
- Department of Neurology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, Republic of Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Jun Kyeung Ko
- Department of Neurosurgery, Medical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Jae Meen Lee
- Department of Neurosurgery, Medical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Jin A. Yoon
- Department of Rehabilitation Medicine, Pusan National University School of Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Chungsu Hwang
- Department of Pathology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Kyung-Un Choi
- Department of Pathology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Gi Yeong Huh
- Department of Forensic Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Young-Eun Kim
- Department of Laboratory Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Eun-Joo Kim
- Department of Neurology, Pusan National University Hospital, Pusan National University School of Medicine and Medical Research Institute, Busan, Republic of Korea
- * E-mail:
| |
Collapse
|
31
|
Zanardini R, Saraceno C, Benussi L, Squitti R, Ghidoni R. Exploring Neurofilament Light Chain and Exosomes in the Genetic Forms of Frontotemporal Dementia. Front Neurosci 2022; 16:758182. [PMID: 35145377 PMCID: PMC8821515 DOI: 10.3389/fnins.2022.758182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 01/03/2022] [Indexed: 11/13/2022] Open
Abstract
Differential diagnosis of neurological disorders and their subtype classification are challenging without specific biomarkers. Genetic forms of these disorders, typified by an autosomal dominant family history, could offer a window to identify potential biomarkers by exploring the presymptomatic stages of the disease. Frontotemporal dementia (FTD) is the second cause of dementia with an age of onset < 65, and its most common mutations are in GRN, C9orf72, and MAPT genes. Several studies have demonstrated that the main proteins involved in FTD pathogenesis can be secreted in exosomes, a specific subtype of extracellular vesicles able to transfer biomolecules between cells avoiding cell-to-cell contact. Neurofilament light chain (NfL) levels in central nervous system have been advocated as biomarkers of axonal injury. NfL concentrations have been found increased in FTD and have been related to disease severity and prognosis. Little information on the relationship between NfL and exosomes in FTD has been collected, deriving mainly from traumatic brain injury. Current review deals with this matter in the attempt to provide an updated discussion of the role of NfL and exosomes as biomarkers of genetic forms of FTD.
Collapse
|
32
|
Giudici KV, Guyonnet S, Morley JE, Nguyen AD, Aggarwal G, Parini A, Li Y, Bateman RJ, Vellas B, de Souto Barreto P, MAPT/DSA Group. Interactions Between Weight Loss and Plasma Neurodegenerative Markers for Determining Cognitive Decline Among Community-Dwelling Older Adults. J Gerontol A Biol Sci Med Sci 2022; 77:1159-1168. [PMID: 35034116 PMCID: PMC9159663 DOI: 10.1093/gerona/glac015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Indexed: 01/18/2023] Open
Abstract
This study aimed to investigate the interaction between weight loss (WL) and plasma amyloid-β 42/40 (Aβ 42/40), neurofilament light chain (NfL), progranulin, and their association with cognitive decline over time among older adults. This 5-year observational approach included 470 participants from the Multidomain Alzheimer Preventive Trial, mean age 76.8 years (SD = 4.5), 59.4% women. WL was defined as ≥5% decrease over the first year. Biomarkers were measured at 12 months. Cognitive function was assessed yearly from 12 months onward by Mini-Mental State Examination (MMSE); Clinical Dementia Rating sum of boxes (CDR-SB); a composite score based on Category Naming Test; Digit Symbol Substitution Test; 10 MMSE orientation items (MMSEO) and free and total recall of the Free and Cued Selective Reminding test; and these tests individually. Twenty-seven participants (5.7%) presented WL. In adjusted analyses, combined WL + lower Aβ 42/40 (≤0.103, lowest quartile) was related with more pronounced 4-year cognitive decline according to CDR-SB (p < .0001) and MMSEO (p = .021), compared with non-WL + higher Aβ 42/40. WL + higher NfL (>94.55 pg/mL, highest quartile) or progranulin (>38.4 ng/mL, 3 higher quartiles) were related with higher cognitive decline according to CDR-SB, MMSE, MMSEO, and composite score (all p < .03), compared with non-WL + lower NfL or higher progranulin. Regrouping progranulin quartiles (Q1-Q3 vs Q4) revealed higher cognitive decline among the WL + lower progranulin group compared with non-WL + lower progranulin. In conclusion, 1-year WL was associated with subsequent higher 4-year cognitive decline among older adults presenting low Aβ 42/40 or high NfL. Future studies combining plasma biomarker assessments and body weight surveillance may be useful for identifying people at risk of cognitive impairment. Clinical trial number: NCT00672685.
Collapse
Affiliation(s)
- Kelly Virecoulon Giudici
- Address correspondence to: Kelly Virecoulon Giudici, PhD, Gérontopôle of Toulouse, Institute of Aging, Toulouse University Hospital, Université Toulouse III Paul Sabatier, 37 Allée Jules Guesde, 31000 Toulouse, France. E-mail:
| | - Sophie Guyonnet
- Gerontopole of Toulouse, Institute of Ageing, Toulouse University Hospital (CHU Toulouse), Toulouse, France,CERPOP UMR1295, University of Toulouse III, INSERM, UPS, Toulouse, France
| | - John E Morley
- Division of Geriatric Medicine, School of Medicine, Saint Louis University, St. Louis, Missouri, USA
| | - Andrew D Nguyen
- Division of Geriatric Medicine, School of Medicine, Saint Louis University, St. Louis, Missouri, USA
| | - Geetika Aggarwal
- Division of Geriatric Medicine, School of Medicine, Saint Louis University, St. Louis, Missouri, USA
| | - Angelo Parini
- Institute of Metabolic and Cardiovascular Diseases (I2MC), INSERM UMR 1048, University of Toulouse III Paul Sabatier, Toulouse, France
| | - Yan Li
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA,Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Bruno Vellas
- Gerontopole of Toulouse, Institute of Ageing, Toulouse University Hospital (CHU Toulouse), Toulouse, France,CERPOP UMR1295, University of Toulouse III, INSERM, UPS, Toulouse, France
| | - Philipe de Souto Barreto
- Gerontopole of Toulouse, Institute of Ageing, Toulouse University Hospital (CHU Toulouse), Toulouse, France,CERPOP UMR1295, University of Toulouse III, INSERM, UPS, Toulouse, France
| | | |
Collapse
|
33
|
Benussi A, Alberici A, Samra K, Russell LL, Greaves CV, Bocchetta M, Ducharme S, Finger E, Fumagalli G, Galimberti D, Jiskoot LC, Le Ber I, Masellis M, Nacmias B, Rowe JB, Sanchez-Valle R, Seelaar H, Synofzik M, Rohrer JD, Borroni B. Conceptual framework for the definition of preclinical and prodromal frontotemporal dementia. Alzheimers Dement 2021; 18:1408-1423. [PMID: 34874596 DOI: 10.1002/alz.12485] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 08/25/2021] [Accepted: 08/30/2021] [Indexed: 12/13/2022]
Abstract
The presymptomatic stages of frontotemporal dementia (FTD) are still poorly defined and encompass a long accrual of progressive biological (preclinical) and then clinical (prodromal) changes, antedating the onset of dementia. The heterogeneity of clinical presentations and the different neuropathological phenotypes have prevented a prior clear description of either preclinical or prodromal FTD. Recent advances in therapeutic approaches, at least in monogenic disease, demand a proper definition of these predementia stages. It has become clear that a consensus lexicon is needed to comprehensively describe the stages that anticipate dementia. The goal of the present work is to review existing literature on the preclinical and prodromal phases of FTD, providing recommendations to address the unmet questions, therefore laying out a strategy for operationalizing and better characterizing these presymptomatic disease stages.
Collapse
Affiliation(s)
- Alberto Benussi
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,Neurology Unit, Department of Neurological and Vision Sciences, ASST Spedali Civili, Brescia, Italy
| | - Antonella Alberici
- Neurology Unit, Department of Neurological and Vision Sciences, ASST Spedali Civili, Brescia, Italy
| | - Kiran Samra
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Lucy L Russell
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Caroline V Greaves
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Martina Bocchetta
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Simon Ducharme
- Department of Psychiatry, Douglas Mental Health University Institute and Douglas Research Centre, McGill University, Montreal, Québec, Canada.,McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Québec, Canada
| | - Elizabeth Finger
- Department of Clinical Neurological Sciences, University of Western Ontario, London, Ontario, Canada
| | - Giorgio Fumagalli
- Fondazione Ca' Granda, IRCCS Ospedale Policlinico, Milan, Italy.,University of Milan, Milan, Italy
| | - Daniela Galimberti
- Fondazione Ca' Granda, IRCCS Ospedale Policlinico, Milan, Italy.,University of Milan, Milan, Italy
| | - Lize C Jiskoot
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.,Department of Neurology, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Isabelle Le Ber
- Paris Brain Institute - Institut du Cerveau - ICM, Sorbonne Université, Inserm U1127, CNRS UMR, Paris, France.,Centre de référence des démences rares ou précoces, IM2A, Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France.,Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France.,Reference Network for Rare Neurological Diseases (ERN-RND), Paris, France
| | - Mario Masellis
- Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, and IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - James B Rowe
- Department of Clinical Neurosciences, MRC Cognition and Brain Sciences Unit and Cambridge University Hospitals NHS Trust, University of Cambridge, Cambridge, UK
| | - Raquel Sanchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Barcelona, Spain
| | - Harro Seelaar
- Department of Neurology, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Matthis Synofzik
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Tübingen, Germany.,Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | | | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Barbara Borroni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,Neurology Unit, Department of Neurological and Vision Sciences, ASST Spedali Civili, Brescia, Italy
| |
Collapse
|
34
|
Wilke C, Reich S, van Swieten JC, Borroni B, Sanchez-Valle R, Moreno F, Laforce R, Graff C, Galimberti D, Rowe JB, Masellis M, Tartaglia MC, Finger E, Vandenberghe R, de Mendonça A, Tagliavini F, Santana I, Ducharme S, Butler CR, Gerhard A, Levin J, Danek A, Otto M, Frisoni G, Ghidoni R, Sorbi S, Bocchetta M, Todd E, Kuhle J, Barro C, Rohrer JD, Synofzik M. Stratifying the Presymptomatic Phase of Genetic Frontotemporal Dementia by Serum NfL and pNfH: A Longitudinal Multicentre Study. Ann Neurol 2021; 91:33-47. [PMID: 34743360 DOI: 10.1002/ana.26265] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Although the presymptomatic stages of frontotemporal dementia (FTD) provide a unique chance to delay or even prevent neurodegeneration by early intervention, they remain poorly defined. Leveraging a large multicenter cohort of genetic FTD mutation carriers, we provide a biomarker-based stratification and biomarker cascade of the likely most treatment-relevant stage within the presymptomatic phase: the conversion stage. METHODS We longitudinally assessed serum levels of neurofilament light (NfL) and phosphorylated neurofilament heavy (pNfH) in the Genetic FTD Initiative (GENFI) cohort (n = 444), using single-molecule array technique. Subjects comprised 91 symptomatic and 179 presymptomatic subjects with mutations in the FTD genes C9orf72, GRN, or MAPT, and 174 mutation-negative within-family controls. RESULTS In a biomarker cascade, NfL increase preceded the hypothetical clinical onset by 15 years and concurred with brain atrophy onset, whereas pNfH increase started close to clinical onset. The conversion stage was marked by increased NfL, but still normal pNfH levels, while both were increased at the symptomatic stage. Intra-individual change rates were increased for NfL at the conversion stage and for pNfH at the symptomatic stage, highlighting their respective potential as stage-dependent dynamic biomarkers within the biomarker cascade. Increased NfL levels and NfL change rates allowed identification of presymptomatic subjects converting to symptomatic disease and capture of proximity-to-onset. We estimate stage-dependent sample sizes for trials aiming to decrease neurofilament levels or change rates. INTERPRETATION Blood NfL and pNfH provide dynamic stage-dependent stratification and, potentially, treatment response biomarkers in presymptomatic FTD, allowing demarcation of the conversion stage. The proposed biomarker cascade might pave the way towards a biomarker-based precision medicine approach to genetic FTD. ANN NEUROL 2021.
Collapse
Affiliation(s)
- Carlo Wilke
- Division Translational Genomics of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Tübingen, Germany.,Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Selina Reich
- Division Translational Genomics of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Tübingen, Germany.,Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | | | - Barbara Borroni
- Centre for Neurodegenerative Disorders, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Raquel Sanchez-Valle
- Alzheimer's disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Barcelona, Spain
| | - Fermin Moreno
- Cognitive Disorders Unit, Department of Neurology, Donostia University Hospital, San Sebastian, Spain.,Neuroscience Area, Biodonostia Health Research Institute, San Sebastian, Spain
| | - Robert Laforce
- Clinique Interdisciplinaire de Mémoire, Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Quebec City, Canada
| | - Caroline Graff
- Center for Alzheimer Research, Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Bioclinicum, Karolinska Institute, Solna, Sweden.,Unit for Hereditary Dementias, Theme Aging, Karolinska University Hospital, Solna, Sweden
| | - Daniela Galimberti
- Fondazione IRCCS Ospedale Policlinico, Milan, Italy.,University of Milan, Centro Dino Ferrari, Milan, Italy
| | - James B Rowe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Mario Masellis
- Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, University of Toronto, Toronto, Canada
| | - Maria C Tartaglia
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
| | - Elizabeth Finger
- Department of Clinical Neurological Sciences, University of Western Ontario, London, Canada
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium.,Neurology Service, University Hospitals Leuven, Leuven, Belgium.,Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | | | | | - Isabel Santana
- University Hospital of Coimbra (HUC), Neurology Service, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Simon Ducharme
- Department of Psychiatry, McGill University Health Centre, McGill University, Montreal, Québec, Canada.,McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Québec, Canada
| | - Chris R Butler
- Nuffield Department of Clinical Neurosciences, Medical Sciences Division, University of Oxford, Oxford, UK.,Department of Brain Sciences, Imperial College London, London, UK
| | - Alexander Gerhard
- Division of Neuroscience and Experimental Psychology, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK.,Departments of Geriatric Medicine and Nuclear Medicine, Essen University Hospital, Essen, Germany
| | - Johannes Levin
- Neurologische Klinik, Ludwig-Maximilians-Universität München, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Adrian Danek
- Neurologische Klinik, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany.,Department of Neurology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Giovanni Frisoni
- Instituto di Ricovero e Cura a Carattere Scientifico Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Roberta Ghidoni
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Sandro Sorbi
- Department of Neurofarba, University of Florence, Florence, Italy.,IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Martina Bocchetta
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, London, UK
| | - Emily Todd
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, London, UK
| | - Jens Kuhle
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland
| | - Christian Barro
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland.,Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Jonathan D Rohrer
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, London, UK
| | - Matthis Synofzik
- Division Translational Genomics of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Tübingen, Germany.,Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| |
Collapse
|
35
|
Dominguez J, Ng A, Yu J, Guevarra AC, Daroy ML, Alfon A, Catindig JA, Dizon M, Santiago J, Del Moral MC, Yu J, Jamerlan A, Ligsay A, Bagyinszky E, An SS, Kim S. Autosomal Dominant Frontotemporal Lobar Degeneration in a Filipino Family with Progranulin Mutation. Dement Geriatr Cogn Disord 2021; 49:557-564. [PMID: 33486486 DOI: 10.1159/000510106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/10/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Compared to Western populations, familial frontotemporal lobar degeneration (FTLD) is rare among Asians. Progranulin (GRN) gene mutation, which is a major cause of FTLD, is likewise rare. We present a family with FTLD from the Philippines with an autosomal dominant pattern of inheritance and GRN mutation and briefly review reports of GRN mutations in Asia. CASE PRESENTATION The proband is 66 years old with progressive nonfluent aphasia (PNFA)-corticobasal syndrome . We assessed 3 generations of her pedigree and found 11 affected relatives with heterogenous phenotypes, usually behavioral variant frontotemporal dementia (FTD) and PNFA. Neuroimaging showed atrophy and hypometabolism consistent with FTD syndromes. White matter hyperintensities were seen in affected members even in the absence of vascular risk factors. A GRN mutation R110X was found in 6 members, 3 with symptoms and 3 were asymptomatic. Plasma GRN was low (<112 ng/mL) in all mutation carriers. No mutations were found in microtubule-associated protein tau, APP, PSEN1, and PSEN2 genes, and all were APOE3. CONCLUSION This is the first Filipino family with autosomal dominant FTD documented with GRN mutation. Identifying families and cohorts would contribute to therapeutic developments in an area with FTD-GRN.
Collapse
Affiliation(s)
- Jacqueline Dominguez
- Institute for Neurosciences, St. Luke's Medical Center, Quezon City, Philippines,
| | - Arlene Ng
- Institute for Neurosciences, St. Luke's Medical Center, Quezon City, Philippines
| | - Jeryl Yu
- Institute for Neurosciences, St. Luke's Medical Center, Quezon City, Philippines
| | - Anne Cristine Guevarra
- Research and Biotechnology Division, St. Luke's Medical Center, Quezon City, Philippines
| | - Maria Luisa Daroy
- Research and Biotechnology Division, St. Luke's Medical Center, Quezon City, Philippines
| | - Alicia Alfon
- Research and Biotechnology Division, St. Luke's Medical Center, Quezon City, Philippines
| | - Joseree-Ann Catindig
- Memory Center-Institute for Neurosciences, St. Luke's Medical Center, Taguig City, Philippines
| | - Mercedes Dizon
- Institute of Radiology, St. Luke's Medical Center, Quezon City, Philippines
| | - Jonas Santiago
- PET Center, St. Luke's Medical Center, Quezon City, Philippines
| | | | - Justine Yu
- Memory Center-Institute for Neurosciences, St. Luke's Medical Center, Taguig City, Philippines
| | - Angelo Jamerlan
- Department of Bionano Technology, Gachon University, Seongnam, Republic of Korea
| | - Antonio Ligsay
- Section of Clinical Research, St. Luke's Medical Center - College of Medicine, Quezon City, Philippines
| | - Eva Bagyinszky
- Department of Industrial and Environmental Engineering, Gachon University, Seongnam, Republic of Korea
| | - Seong Soo An
- Department of Bionano Technology, Gachon University, Seongnam, Republic of Korea
| | - Sangyun Kim
- Department of Neurology, Seoul National University College of Medicine & Neurocognitive Behavior Center, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| |
Collapse
|
36
|
Deng B, Zheng Z, Zheng J, Yang W, Huang Y, Luo Y, Jin D, Shen L, Jin K, Wang Q. FTD-PSP is an Unusual Clinical Phenotype in A Frontotemporal Dementia Patient with A Novel Progranulin Mutation. Aging Dis 2021; 12:1741-1752. [PMID: 34631218 PMCID: PMC8460311 DOI: 10.14336/ad.2021.0309] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 03/09/2021] [Indexed: 12/28/2022] Open
Abstract
Progranulin (GRN) mutations are a major cause of frontotemporal dementia (FTD); the spectrum of clinical phenotypes of FTD is much more extensive than previously reported. The frequency and locations of GRN mutations in Chinese patients with FTD remain uncertain. We performed cDNA sequencing in one sporadic male patient who initially presented FTD symptoms. Brain magnetic resonance imaging (MRI) and positron emission computed tomography/computed tomography (PET/CT) were applied to further confirm the diagnosis of FTD from this patient. Cellular apoptosis and survival test were performed to identify the function of GRN. We identified one novel missense GRN mutation (c.1498G>A, p.V500I) in this patient, who initially presented typical behavioral-variant frontotemporal dementia (bvFTD) features but then presented progressive supranuclear palsy (PSP) clinical characteristics 5 years after onset. Besides, WT GRN protein showed an adequate trophic stimulus to preserve the survival of SH-SY5Y cells in the medium free of serum, while GRN mutation (c.1498G>A, p.V500I) may impair the ability of supporting cell survival. This study owns significant implications for genetic counseling and clinical heterogeneity. We illustrate the fact that FTD presenting features of bvFTD and PSP in one patient could be considered as a specific phenotype in patients with GRN mutations. GRN p.V500I led to the neuronal degeneration in vitro; this finding provides a significant evidence that this mutation may be a new causative mutation in patients with FTD.
Collapse
Affiliation(s)
- Bin Deng
- 1Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Zhe Zheng
- 1Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Jialing Zheng
- 1Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Wanlin Yang
- 1Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Yu Huang
- 1Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Yuqi Luo
- 1Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Dana Jin
- 3College of Biological Sciences, University of California, Davis, CA 95616, USA
| | - Lu Shen
- 2Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Kunlin Jin
- 4Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Qing Wang
- 1Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, China
| |
Collapse
|
37
|
Frydas A, Cacace R, van der Zee J, Van Broeckhoven C, Wauters E. Genetic variants in progranulin upstream open reading frames increase downstream protein expression. Neurobiol Aging 2021; 110:113-121. [PMID: 34620513 DOI: 10.1016/j.neurobiolaging.2021.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/29/2021] [Accepted: 09/05/2021] [Indexed: 11/25/2022]
Abstract
Premature termination codon (PTC) mutations in the granulin gene (GRN) lead to loss-of-function (LOF) of the progranulin protein (PGRN), causing frontotemporal lobar degeneration (FTLD) by haploinsufficiency. GRN expression is regulated at multiple levels, including the 5' untranslated region (UTR). The main 5' UTR of GRN and an alternative 5' UTR, contain upstream open reading frames (uORFs). These mRNA elements generally act as cis-repressors of translation. Disruption of each uORF of the alternative 5' UTR, increases protein expression with the 2 ATG-initiated uORFs being capable of initiating translation. We performed targeted sequencing of the uORF regions in a Flanders-Belgian cohort of patients with frontotemporal dementia (FTD) and identified 2 genetic variants, one in each 5' UTR. Both variants increase downstream protein levels, with the main 5' UTR variant rs76783532 causing a significant 1.5-fold increase in protein expression. We observed that the presence of functional uORFs in the alternative 5' UTR act as potential regulators of PGRN expression and demonstrate that genetic variation within GRN uORFs can alter their function.
Collapse
Affiliation(s)
- Alexandros Frydas
- VIB Center for Molecular Neurology, Antwerp, Belgium; Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Rita Cacace
- VIB Center for Molecular Neurology, Antwerp, Belgium; Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Julie van der Zee
- VIB Center for Molecular Neurology, Antwerp, Belgium; Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Christine Van Broeckhoven
- VIB Center for Molecular Neurology, Antwerp, Belgium; Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
| | - Eline Wauters
- VIB Center for Molecular Neurology, Antwerp, Belgium; Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
38
|
Davis SE, Roth JR, Aljabi Q, Hakim AR, Savell KE, Day JJ, Arrant AE. Delivering progranulin to neuronal lysosomes protects against excitotoxicity. J Biol Chem 2021; 297:100993. [PMID: 34298019 PMCID: PMC8379502 DOI: 10.1016/j.jbc.2021.100993] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/08/2021] [Accepted: 07/19/2021] [Indexed: 01/18/2023] Open
Abstract
Loss-of-function mutations in progranulin (GRN) are a major genetic cause of frontotemporal dementia (FTD), possibly due to loss of progranulin's neurotrophic and anti-inflammatory effects. Progranulin promotes neuronal growth and protects against excitotoxicity and other forms of injury. It is unclear if these neurotrophic effects are mediated through cellular signaling or through promotion of lysosomal function. Progranulin is a secreted proprotein that may activate neurotrophic signaling through cell-surface receptors. However, progranulin is efficiently trafficked to lysosomes and is necessary for maintaining lysosomal function. To determine which of these mechanisms mediates progranulin's protection against excitotoxicity, we generated lentiviral vectors expressing progranulin (PGRN) or lysosome-targeted progranulin (L-PGRN). L-PGRN was generated by fusing the LAMP-1 transmembrane and cytosolic domains to the C-terminus of progranulin. L-PGRN exhibited no detectable secretion, but was delivered to lysosomes and processed into granulins. PGRN and L-PGRN protected against NMDA excitotoxicity in rat primary cortical neurons, but L-PGRN had more consistent protective effects than PGRN. L-PGRN's protective effects were likely mediated through the autophagy-lysosomal pathway. In control neurons, an excitotoxic dose of NMDA stimulated autophagy, and inhibiting autophagy with 3-methyladenine reduced excitotoxic cell death. L-PGRN blunted the autophagic response to NMDA and occluded the protective effect of 3-methyladenine. This was not due to a general impairment of autophagy, as L-PGRN increased basal autophagy and did not alter autophagy after nutrient starvation. These data show that progranulin's protection against excitotoxicity does not require extracellular progranulin, but is mediated through lysosomes, providing a mechanistic link between progranulin's lysosomal and neurotrophic effects.
Collapse
Affiliation(s)
- Skylar E Davis
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama, USA; Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jonathan R Roth
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama, USA; Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Qays Aljabi
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama, USA; Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Ahmad R Hakim
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama, USA; Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Katherine E Savell
- Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, Alabama, USA; Department of Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jeremy J Day
- Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, Alabama, USA; Department of Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Andrew E Arrant
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama, USA; Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, Alabama, USA.
| |
Collapse
|
39
|
Ljubenkov PA, Edwards L, Iaccarino L, La Joie R, Rojas JC, Koestler M, Harris B, Boeve BF, Borroni B, van Swieten JC, Grossman M, Pasquier F, Frisoni GB, Mummery CJ, Vandenberghe R, Le Ber I, Hannequin D, McGinnis SM, Auriacombe S, Onofrj M, Goodman IJ, Riordan HJ, Wisniewski G, Hesterman J, Marek K, Haynes BA, Patzke H, Koenig G, Hilt D, Moebius H, Boxer AL. Effect of the Histone Deacetylase Inhibitor FRM-0334 on Progranulin Levels in Patients With Progranulin Gene Haploinsufficiency: A Randomized Clinical Trial. JAMA Netw Open 2021; 4:e2125584. [PMID: 34559230 PMCID: PMC8463943 DOI: 10.1001/jamanetworkopen.2021.25584] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
IMPORTANCE Histone deacetylase inhibitors have been repeatedly shown to elevate progranulin levels in preclinical models. This report describes the first randomized clinical trial of a histone deacetylase inhibitor in frontotemporal dementia (FTD) resulting from progranulin (GRN) gene variations. OBJECTIVE To characterize the safety, tolerability, plasma pharmacokinetics, and pharmacodynamic effects of oral FRM-0334 on plasma progranulin and other exploratory biomarkers, including fluorodeoxyglucose (FDG)-positron emission tomography (PET), in individuals with GRN haploinsufficiency. DESIGN, SETTING, AND PARTICIPANTS In this randomized, double-blind, placebo-controlled, dose-escalating, phase 2a safety, tolerability, and pharmacodynamic clinical study, 2 doses of a histone deacetylase inhibitor (FRM-0334) were administered to participants with prodromal to moderate FTD with granulin variations. Participants were recruited from January 13, 2015, to April 13, 2016. The study included 27 participants with prodromal (n = 8) or mild-to-moderate symptoms of FTD (n = 19) and heterozygous pathogenic variations in GRN and was conducted at multiple centers in North America, the UK, and the European Union. Data were analyzed from June 9, 2019, to May 13, 2021. INTERVENTIONS Daily oral placebo (n = 5), 300 mg of FRM-0334 (n = 11), or 500 mg of FRM-0334 (n = 11) was administered for 28 days. MAIN OUTCOMES AND MEASURES Primary outcomes were safety and tolerability of FRM-0334 and its peripheral pharmacodynamic effect on plasma progranulin. Secondary outcomes were the plasma pharmacokinetic profile of FRM-0334 and its pharmacodynamic effect on cerebrospinal fluid progranulin. Exploratory outcomes were FDG-PET, FTD clinical severity, and cerebrospinal fluid biomarkers (neurofilament light chain [NfL], amyloid β 1-42, phosphorylated tau 181, and total tau [t-tau]). RESULTS A total of 27 participants (mean [SD] age, 56.6 [10.5] years; 16 women [59.3%]; 26 White participants [96.3%]) with GRN variations were randomized and completed treatment. FRM-0334 was safe and well tolerated but did not affect plasma progranulin (4.3 pg/mL per day change after treatment; 95% CI, -10.1 to 18.8 pg/mL; P = .56), cerebrospinal fluid progranulin (0.42 pg/mL per day; 95% CI, -0.12 to 0.95 pg/mL; P = .13), or exploratory pharmacodynamic measures. Plasma FRM-0334 exposure did not increase proportionally with dose. Brain FDG-PET data were available in 26 of 27 randomized participants. In a cross-sectional analysis of 26 individuals, bifrontal cortical FDG hypometabolism was associated with worse Clinical Dementia Rating (CDR) plus National Alzheimer's Coordinating Center frontotemporal lobar degeneration sum of boxes score (b = -3.6 × 10-2 standardized uptake value ratio [SUVR] units/CDR units; 95% CI, -4.9 × 10-2 to -2.2 × 10-2; P < .001), high cerebrospinal fluid NfL (b = -9.2 × 10-5 SUVR units/pg NfL/mL; 95% CI, -1.3 × 10-4 to -5.6 × 10-5; P < .001), and high CSF t-tau (-7.2 × 10-4 SUVR units/pg t-tau/mL; 95% CI, -1.4 × 10-3 to -9.5 × 10-5; P = .03). CONCLUSIONS AND RELEVANCE In this randomized clinical trial, the current formulation of FRM-0334 did not elevate PRGN levels, which could reflect a lack of efficacy at attained exposures, low bioavailability, or some combination of the 2 factors. Bifrontal FDG-PET is a sensitive measure of symptomatic GRN haploinsufficiency. International multicenter clinical trials of FTD-GRN are feasible. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02149160.
Collapse
Affiliation(s)
- Peter A. Ljubenkov
- Memory and Aging Center, Department of Neurology, University of California at San Francisco, Weill Institute for Neurosciences, San Francisco
| | - Lauren Edwards
- Memory and Aging Center, Department of Neurology, University of California at San Francisco, Weill Institute for Neurosciences, San Francisco
| | - Leonardo Iaccarino
- Memory and Aging Center, Department of Neurology, University of California at San Francisco, Weill Institute for Neurosciences, San Francisco
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, University of California at San Francisco, Weill Institute for Neurosciences, San Francisco
| | - Julio C. Rojas
- Memory and Aging Center, Department of Neurology, University of California at San Francisco, Weill Institute for Neurosciences, San Francisco
| | - Mary Koestler
- Memory and Aging Center, Department of Neurology, University of California at San Francisco, Weill Institute for Neurosciences, San Francisco
| | - Baruch Harris
- ROME Therapeutics, Cambridge, Massachusetts
- Metera Pharmaceuticals, Cambridge, Massachusetts
| | | | - Barbara Borroni
- Neurology Unit, Centre for Neurodegenerative Disorders, ASST Spedali Civili Brescia, Italy
- Department of Clinical and Experimental Sciences, University of Brescia, Italy
| | - John C. van Swieten
- Department of Neurology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Murray Grossman
- Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Florence Pasquier
- Lille University, Inserm 1172, CHU Lille, CNR-MAJ, DISTALZ, LiCEND, Lille, France
| | - Giovanni B. Frisoni
- Lab of Alzheimer's Neuroimaging and Epidemiology, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Catherine J. Mummery
- Dementia Research Centre, Institute of Neurology, University College London, London, United Kingdom
| | - Rik Vandenberghe
- Neurology Service University Hospitals Leuven, Leuven, Belgium
- Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Isabelle Le Ber
- Paris Brain Institute, Sorbonne University, Paris, France
- Neurology Department, Reference Centre for Rare or Early Dementias, Paris, France
| | - Didier Hannequin
- CHU Rouen Normandie, Service de Neurologie, Rouen University Hospital, Rouen, France
| | - Scott M. McGinnis
- Department of Neurology, Massachusetts General Hospital, Charlestown
| | - Sophie Auriacombe
- Neurology Department, CHU Bordeaux, Bordeaux Hospital, Bordeaux, France
| | - Marco Onofrj
- Department of Neuroscience Imaging, University G d'Annunzio of Chieti-Pescara, Chieti, Italy
| | | | | | | | - Jacob Hesterman
- Institute for Neurodegenerative Disorders, New Haven, Connecticut
| | - Ken Marek
- Invicro, A Konica Minolta Company, Boston, Massachusetts
- Institute for Neurodegenerative Disorders, New Haven, Connecticut
| | - Beth Ann Haynes
- FORUM Pharmaceuticals Incorporated, Waltham, Massachusetts
- Alector, South San Francisco, California
| | | | - Gerhard Koenig
- FORUM Pharmaceuticals Incorporated, Waltham, Massachusetts
- Arkuda Therapeutics, Watertown, Massachusetts
| | - Dana Hilt
- Arkuda Therapeutics, Watertown, Massachusetts
- Lysosomal Therapeutics, Cambridge, Massachusetts
- Frequency Therapeutics, Farmington, Connecticut
| | - Hans Moebius
- FORUM Pharmaceuticals Incorporated, Waltham, Massachusetts
- Athira Pharma Inc, Seattle, Washington
| | - Adam L. Boxer
- Memory and Aging Center, Department of Neurology, University of California at San Francisco, Weill Institute for Neurosciences, San Francisco
| |
Collapse
|
40
|
Terryn J, Verfaillie CM, Van Damme P. Tweaking Progranulin Expression: Therapeutic Avenues and Opportunities. Front Mol Neurosci 2021; 14:713031. [PMID: 34366786 PMCID: PMC8343103 DOI: 10.3389/fnmol.2021.713031] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/07/2021] [Indexed: 01/01/2023] Open
Abstract
Frontotemporal dementia (FTD) is a neurodegenerative disease, leading to behavioral changes and language difficulties. Heterozygous loss-of-function mutations in progranulin (GRN) induce haploinsufficiency of the protein and are associated with up to one-third of all genetic FTD cases worldwide. While the loss of GRN is primarily associated with neurodegeneration, the biological functions of the secreted growth factor-like protein are more diverse, ranging from wound healing, inflammation, vasculogenesis, and metabolic regulation to tumor cell growth and metastasis. To date, no disease-modifying treatments exist for FTD, but different therapeutic approaches to boost GRN levels in the central nervous system are currently being developed (including AAV-mediated GRN gene delivery as well as anti-SORT1 antibody therapy). In this review, we provide an overview of the multifaceted regulation of GRN levels and the corresponding therapeutic avenues. We discuss the opportunities, advantages, and potential drawbacks of the diverse approaches. Additionally, we highlight the therapeutic potential of elevating GRN levels beyond patients with loss-of-function mutations in GRN.
Collapse
Affiliation(s)
- Joke Terryn
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium.,Laboratory of Neurobiology, Center for Brain and Disease Research, VIB, Leuven, Belgium
| | - Catherine M Verfaillie
- Department of Development and Regeneration, Interdepartmental Stem Cell Institute, KU Leuven-University of Leuven, Leuven, Belgium
| | - Philip Van Damme
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium.,Laboratory of Neurobiology, Center for Brain and Disease Research, VIB, Leuven, Belgium.,Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
41
|
Root J, Merino P, Nuckols A, Johnson M, Kukar T. Lysosome dysfunction as a cause of neurodegenerative diseases: Lessons from frontotemporal dementia and amyotrophic lateral sclerosis. Neurobiol Dis 2021; 154:105360. [PMID: 33812000 PMCID: PMC8113138 DOI: 10.1016/j.nbd.2021.105360] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 03/16/2021] [Accepted: 03/29/2021] [Indexed: 12/11/2022] Open
Abstract
Frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) are fatal neurodegenerative disorders that are thought to exist on a clinical and pathological spectrum. FTD and ALS are linked by shared genetic causes (e.g. C9orf72 hexanucleotide repeat expansions) and neuropathology, such as inclusions of ubiquitinated, misfolded proteins (e.g. TAR DNA-binding protein 43; TDP-43) in the CNS. Furthermore, some genes that cause FTD or ALS when mutated encode proteins that localize to the lysosome or modulate endosome-lysosome function, including lysosomal fusion, cargo trafficking, lysosomal acidification, autophagy, or TFEB activity. In this review, we summarize evidence that lysosomal dysfunction, caused by genetic mutations (e.g. C9orf72, GRN, MAPT, TMEM106B) or toxic-gain of function (e.g. aggregation of TDP-43 or tau), is an important pathogenic disease mechanism in FTD and ALS. Further studies into the normal function of many of these proteins are required and will help uncover the mechanisms that cause lysosomal dysfunction in FTD and ALS. Mutations or polymorphisms in genes that encode proteins important for endosome-lysosome function also occur in other age-dependent neurodegenerative diseases, including Alzheimer's (e.g. APOE, PSEN1, APP) and Parkinson's (e.g. GBA, LRRK2, ATP13A2) disease. A more complete understanding of the common and unique features of lysosome dysfunction across the spectrum of neurodegeneration will help guide the development of therapies for these devastating diseases.
Collapse
Affiliation(s)
- Jessica Root
- Department of Pharmacology and Chemical Biology, Emory University, School of Medicine, Atlanta 30322, Georgia; Center for Neurodegenerative Disease, Emory University, School of Medicine, Atlanta 30322, Georgia
| | - Paola Merino
- Department of Pharmacology and Chemical Biology, Emory University, School of Medicine, Atlanta 30322, Georgia; Center for Neurodegenerative Disease, Emory University, School of Medicine, Atlanta 30322, Georgia
| | - Austin Nuckols
- Department of Pharmacology and Chemical Biology, Emory University, School of Medicine, Atlanta 30322, Georgia; Center for Neurodegenerative Disease, Emory University, School of Medicine, Atlanta 30322, Georgia
| | - Michelle Johnson
- Department of Pharmacology and Chemical Biology, Emory University, School of Medicine, Atlanta 30322, Georgia; Center for Neurodegenerative Disease, Emory University, School of Medicine, Atlanta 30322, Georgia
| | - Thomas Kukar
- Department of Pharmacology and Chemical Biology, Emory University, School of Medicine, Atlanta 30322, Georgia; Center for Neurodegenerative Disease, Emory University, School of Medicine, Atlanta 30322, Georgia; Department of Neurology, Emory University, School of Medicine, Atlanta 30322, Georgia.
| |
Collapse
|
42
|
Duran-Aniotz C, Orellana P, Leon Rodriguez T, Henriquez F, Cabello V, Aguirre-Pinto MF, Escobedo T, Takada LT, Pina-Escudero SD, Lopez O, Yokoyama JS, Ibanez A, Parra MA, Slachevsky A. Systematic Review: Genetic, Neuroimaging, and Fluids Biomarkers for Frontotemporal Dementia Across Latin America Countries. Front Neurol 2021; 12:663407. [PMID: 34248820 PMCID: PMC8263937 DOI: 10.3389/fneur.2021.663407] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 05/27/2021] [Indexed: 11/13/2022] Open
Abstract
Frontotemporal dementia (FTD) includes a group of clinically, genetically, and pathologically heterogeneous neurodegenerative disorders, affecting the fronto-insular-temporal regions of the brain. Clinically, FTD is characterized by progressive deficits in behavior, executive function, and language and its diagnosis relies mainly on the clinical expertise of the physician/consensus group and the use of neuropsychological tests and/or structural/functional neuroimaging, depending on local availability. The modest correlation between clinical findings and FTD neuropathology makes the diagnosis difficult using clinical criteria and often leads to underdiagnosis or misdiagnosis, primarily due to lack of recognition or awareness of FTD as a disease and symptom overlap with psychiatric disorders. Despite advances in understanding the underlying neuropathology of FTD, accurate and sensitive diagnosis for this disease is still lacking. One of the major challenges is to improve diagnosis in FTD patients as early as possible. In this context, biomarkers have emerged as useful methods to provide and/or complement clinical diagnosis for this complex syndrome, although more evidence is needed to incorporate most of them into clinical practice. However, most biomarker studies have been performed using North American or European populations, with little representation of the Latin American and the Caribbean (LAC) region. In the LAC region, there are additional challenges, particularly the lack of awareness and knowledge about FTD, even in specialists. Also, LAC genetic heritage and cultures are complex, and both likely influence clinical presentations and may modify baseline biomarker levels. Even more, due to diagnostic delay, the clinical presentation might be further complicated by both neurological and psychiatric comorbidity, such as vascular brain damage, substance abuse, mood disorders, among others. This systematic review provides a brief update and an overview of the current knowledge on genetic, neuroimaging, and fluid biomarkers for FTD in LAC countries. Our review highlights the need for extensive research on biomarkers in FTD in LAC to contribute to a more comprehensive understanding of the disease and its associated biomarkers. Dementia research is certainly reduced in the LAC region, highlighting an urgent need for harmonized, innovative, and cross-regional studies with a global perspective across multiple areas of dementia knowledge.
Collapse
Affiliation(s)
- Claudia Duran-Aniotz
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibanez, Santiago, Chile
- Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibanez, Santiago, Chile
| | - Paulina Orellana
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibanez, Santiago, Chile
- Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibanez, Santiago, Chile
| | - Tomas Leon Rodriguez
- Trinity College, Global Brain Health Institute, Dublin, Ireland
- Memory and Neuropsychiatric Clinic (CMYN) Neurology Department, Hospital del Salvador and Faculty of Medicine, University of Chile, Santiago, Chile
| | - Fernando Henriquez
- Neuropsychology and Clinical Neuroscience Laboratory (LANNEC), Physiopathology Department - Institute of Biomedical Sciences (ICBM), Neuroscience and East Neuroscience Departments, Faculty of Medicine, University of Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
| | - Victoria Cabello
- Neuropsychology and Clinical Neuroscience Laboratory (LANNEC), Physiopathology Department - Institute of Biomedical Sciences (ICBM), Neuroscience and East Neuroscience Departments, Faculty of Medicine, University of Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
| | | | - Tamara Escobedo
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibanez, Santiago, Chile
- Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibanez, Santiago, Chile
| | - Leonel T. Takada
- Cognitive and Behavioral Neurology Unit - Department of Neurology, University of São Paulo, São Paulo, Brazil
| | - Stefanie D. Pina-Escudero
- Global Brain Health Institute (GBHI), University of California San Francisco (UCSF), San Francisco, CA, United States
- UCSF Department of Neurology, Memory and Aging Center, UCSF, San Francisco, CA, United States
| | - Oscar Lopez
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jennifer S. Yokoyama
- Global Brain Health Institute (GBHI), University of California San Francisco (UCSF), San Francisco, CA, United States
- UCSF Department of Neurology, Memory and Aging Center, UCSF, San Francisco, CA, United States
| | - Agustin Ibanez
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibanez, Santiago, Chile
- Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibanez, Santiago, Chile
- Trinity College, Global Brain Health Institute, Dublin, Ireland
- Global Brain Health Institute (GBHI), University of California San Francisco (UCSF), San Francisco, CA, United States
- Cognitive Neuroscience Center (CNC), Universidad de San Andrés, & National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - Mario A. Parra
- School of Psychological Sciences and Health, University of Strathclyde, Glasgow, United Kingdom
| | - Andrea Slachevsky
- Memory and Neuropsychiatric Clinic (CMYN) Neurology Department, Hospital del Salvador and Faculty of Medicine, University of Chile, Santiago, Chile
- Neuropsychology and Clinical Neuroscience Laboratory (LANNEC), Physiopathology Department - Institute of Biomedical Sciences (ICBM), Neuroscience and East Neuroscience Departments, Faculty of Medicine, University of Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
- Cognitive and Behavioral Neurology Unit - Department of Neurology, University of São Paulo, São Paulo, Brazil
- Department of Neurology and Psychiatry, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| |
Collapse
|
43
|
Lall D, Lorenzini I, Mota TA, Bell S, Mahan TE, Ulrich JD, Davtyan H, Rexach JE, Muhammad AKMG, Shelest O, Landeros J, Vazquez M, Kim J, Ghaffari L, O'Rourke JG, Geschwind DH, Blurton-Jones M, Holtzman DM, Sattler R, Baloh RH. C9orf72 deficiency promotes microglial-mediated synaptic loss in aging and amyloid accumulation. Neuron 2021; 109:2275-2291.e8. [PMID: 34133945 DOI: 10.1016/j.neuron.2021.05.020] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 02/13/2021] [Accepted: 05/17/2021] [Indexed: 12/13/2022]
Abstract
C9orf72 repeat expansions cause inherited amyotrophic lateral sclerosis (ALS)/frontotemporal dementia (FTD) and result in both loss of C9orf72 protein expression and production of potentially toxic RNA and dipeptide repeat proteins. In addition to ALS/FTD, C9orf72 repeat expansions have been reported in a broad array of neurodegenerative syndromes, including Alzheimer's disease. Here we show that C9orf72 deficiency promotes a change in the homeostatic signature in microglia and a transition to an inflammatory state characterized by an enhanced type I IFN signature. Furthermore, C9orf72-depleted microglia trigger age-dependent neuronal defects, in particular enhanced cortical synaptic pruning, leading to altered learning and memory behaviors in mice. Interestingly, C9orf72-deficient microglia promote enhanced synapse loss and neuronal deficits in a mouse model of amyloid accumulation while paradoxically improving plaque clearance. These findings suggest that altered microglial function due to decreased C9orf72 expression directly contributes to neurodegeneration in repeat expansion carriers independent of gain-of-function toxicities.
Collapse
Affiliation(s)
- Deepti Lall
- Center for Neural Science and Medicine, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Ileana Lorenzini
- Department of Neurobiology, Barrow Neurological Institute, 350 W. Thomas Road, Phoenix, AZ 85013, USA
| | - Thomas A Mota
- Center for Neural Science and Medicine, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Shaughn Bell
- Center for Neural Science and Medicine, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Thomas E Mahan
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Jason D Ulrich
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Hayk Davtyan
- Institute for Memory Impairments and Neurological Disorders, Sue & Bill Gross Stem Cell Research Center, 3200 Gross Hall, 845 Health Sciences Road, University of California, Irvine, Irvine, CA 92697, USA
| | - Jessica E Rexach
- Program in Neurogenetics, Department of Neurology, Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - A K M Ghulam Muhammad
- Center for Neural Science and Medicine, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Oksana Shelest
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Jesse Landeros
- Center for Neural Science and Medicine, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Michael Vazquez
- Center for Neural Science and Medicine, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Junwon Kim
- Department of Neurobiology, Barrow Neurological Institute, 350 W. Thomas Road, Phoenix, AZ 85013, USA
| | - Layla Ghaffari
- Department of Neurobiology, Barrow Neurological Institute, 350 W. Thomas Road, Phoenix, AZ 85013, USA
| | - Jacqueline Gire O'Rourke
- Center for Neural Science and Medicine, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Daniel H Geschwind
- Program in Neurogenetics, Department of Neurology, Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mathew Blurton-Jones
- Institute for Memory Impairments and Neurological Disorders, Sue & Bill Gross Stem Cell Research Center, 3200 Gross Hall, 845 Health Sciences Road, University of California, Irvine, Irvine, CA 92697, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Rita Sattler
- Department of Neurobiology, Barrow Neurological Institute, 350 W. Thomas Road, Phoenix, AZ 85013, USA.
| | - Robert H Baloh
- Center for Neural Science and Medicine, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA; Department of Neurology, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA.
| |
Collapse
|
44
|
Vieira SRL, Morris HR. Neurodegenerative Disease Risk in Carriers of Autosomal Recessive Disease. Front Neurol 2021; 12:679927. [PMID: 34149605 PMCID: PMC8211888 DOI: 10.3389/fneur.2021.679927] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/14/2021] [Indexed: 01/19/2023] Open
Abstract
Genetics has driven significant discoveries in the field of neurodegenerative diseases (NDDs). An emerging theme in neurodegeneration warrants an urgent and comprehensive update: that carrier status of early-onset autosomal recessive (AR) disease, typically considered benign, is associated with an increased risk of a spectrum of late-onset NDDs. Glucosylceramidase beta (GBA1) gene mutations, responsible for the AR lysosomal storage disorder Gaucher disease, are a prominent example of this principle, having been identified as an important genetic risk factor for Parkinson disease. Genetic analyses have revealed further examples, notably GRN, TREM2, EIF2AK3, and several other LSD and mitochondria function genes. In this Review, we discuss the evidence supporting the strikingly distinct allele-dependent clinical phenotypes observed in carriers of such gene mutations and its impact on the wider field of neurodegeneration.
Collapse
Affiliation(s)
| | - Huw R. Morris
- Department of Clinical and Movement Neurosciences, University College London, Queen Square Institute of Neurology, London, United Kingdom
| |
Collapse
|
45
|
Wang XM, Zeng P, Fang YY, Zhang T, Tian Q. Progranulin in neurodegenerative dementia. J Neurochem 2021; 158:119-137. [PMID: 33930186 DOI: 10.1111/jnc.15378] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/28/2021] [Accepted: 04/16/2021] [Indexed: 01/21/2023]
Abstract
Long-term or severe lack of protective factors is important in the pathogenesis of neurodegenerative dementia. Progranulin (PGRN), a neurotrophic factor expressed mainly in neurons and microglia, has various neuroprotective effects such as anti-inflammatory effects, promoting neuron survival and neurite growth, and participating in normal lysosomal function. Mutations in the PGRN gene (GRN) have been found in several neurodegenerative dementias, including frontotemporal lobar degeneration (FTLD) and Alzheimer's disease (AD). Herein, PGRN deficiency and PGRN hydrolytic products (GRNs) in the pathological changes related to dementia, including aggregation of tau and TAR DNA-binding protein 43 (TDP-43), amyloid-β (Aβ) overproduction, neuroinflammation, lysosomal dysfunction, neuronal death, and synaptic deficit have been summarized. Furthermore, as some therapeutic strategies targeting PGRN have been developed in various models, we highlighted PGRN as a potential anti-neurodegeneration target in dementia.
Collapse
Affiliation(s)
- Xiao-Ming Wang
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Zeng
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry, Huazhong University of Science and Technology, Wuhan, China
| | - Ying-Yan Fang
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Huangshi, China
| | - Teng Zhang
- Department of Neurology, Shanxian Central Hospital, The Affiliated Huxi Hospital of Jining Medical College, Heze, China
| | - Qing Tian
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
46
|
Pascual B, Funk Q, Zanotti-Fregonara P, Cykowski MD, Veronese M, Rockers E, Bradbury K, Yu M, Nakawah MO, Román GC, Schulz PE, Arumanayagam AS, Beers D, Faridar A, Fujita M, Appel SH, Masdeu JC. Neuroinflammation is highest in areas of disease progression in semantic dementia. Brain 2021; 144:1565-1575. [PMID: 33824991 DOI: 10.1093/brain/awab057] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/25/2020] [Accepted: 12/10/2020] [Indexed: 12/11/2022] Open
Abstract
Despite epidemiological and genetic data linking semantic dementia to inflammation, the topography of neuroinflammation in semantic dementia, also known as the semantic variant of primary progressive aphasia, remains unclear. The pathology starts at the tip of the left temporal lobe where, in addition to cortical atrophy, a strong signal appears with the tau PET tracer 18F-flortaucipir, even though the disease is not typically associated with tau but with TDP-43 protein aggregates. Here, we characterized the topography of inflammation in semantic variant primary progressive aphasia using high-resolution PET and the tracer 11C-PBR28 as a marker of microglial activation. We also tested the hypothesis that inflammation, by providing non-specific binding targets, could explain the 18F-flortaucipir signal in semantic variant primary progressive aphasia. Eight amyloid-PET-negative patients with semantic variant primary progressive aphasia underwent 11C-PBR28 and 18F-flortaucipir PET. Healthy controls underwent 11C-PBR28 PET (n = 12) or 18F-flortaucipir PET (n = 12). Inflammation in PET with 11C-PBR28 was analysed using Logan graphical analysis with a metabolite-corrected arterial input function. 18F-flortaucipir standardized uptake value ratios were calculated using the cerebellum as the reference region. Since monoamine oxidase B receptors are expressed by astrocytes in affected tissue, selegiline was administered to one patient with semantic variant primary progressive aphasia before repeating 18F-flortaucipir scanning to test whether monoamine oxidase B inhibition blocked flortaucipir binding, which it did not. While 11C-PBR28 uptake was mostly cortical, 18F-flortaucipir uptake was greatest in the white matter. The uptake of both tracers was increased in the left temporal lobe and in the right temporal pole, as well as in regions adjoining the left temporal pole such as insula and orbitofrontal cortex. However, peak uptake of 18F-flortaucipir localized to the left temporal pole, the epicentre of pathology, while the peak of inflammation 11C-PBR28 uptake localized to a more posterior, mid-temporal region and left insula and orbitofrontal cortex, in the periphery of the damage core. Neuroinflammation, greatest in the areas of progression of the pathological process in semantic variant primary progressive aphasia, should be further studied as a possible therapeutic target to slow disease progression.
Collapse
Affiliation(s)
- Belen Pascual
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| | - Quentin Funk
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| | - Paolo Zanotti-Fregonara
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| | - Matthew D Cykowski
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA.,Stanley H. Appel Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| | - Mattia Veronese
- Centre for Neuroimaging Sciences, King's College London, London, UK
| | - Elijah Rockers
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| | - Kathleen Bradbury
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| | - Meixiang Yu
- Cyclotron and Radiopharmaceutical Core, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| | - Mohammad O Nakawah
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| | - Gustavo C Román
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| | - Paul E Schulz
- Department of Neurology, McGovern Medical School of UT Health, Houston, TX, USA
| | - Anithachristy S Arumanayagam
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| | - David Beers
- Stanley H. Appel Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| | - Alireza Faridar
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| | - Masahiro Fujita
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| | - Stanley H Appel
- Stanley H. Appel Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| | - Joseph C Masdeu
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| |
Collapse
|
47
|
Khoury R, Liu Y, Sheheryar Q, Grossberg GT. Pharmacotherapy for Frontotemporal Dementia. CNS Drugs 2021; 35:425-438. [PMID: 33840052 DOI: 10.1007/s40263-021-00813-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/26/2021] [Indexed: 12/11/2022]
Abstract
Frontotemporal dementia is a heterogeneous spectrum of neurodegenerative disorders. The neuropathological inclusions are tau proteins, TAR DNA binding protein 43 kDa-TDP-43, or fused in sarcoma-ubiquitinated inclusions. Genetically, several autosomal mutations account for the heritability of the disorder. Phenotypically, frontotemporal dementia can present with a behavioral variant or a language variant called primary progressive aphasia. To date, there are no approved symptomatic or disease-modifying treatments for frontotemporal dementia. Currently used therapies are supported by low-level of evidence (mostly uncontrolled) studies. The off-label use of drugs is also limited by their side-effect profile including an increased risk of confusion, parkinsonian symptoms, and risk of mortality. Emerging disease-modifying treatments currently target the progranulin and the expansion on chromosome 9 open reading frame 72 genes as well as tau deposits. Advancing our understanding of the pathophysiology of the disease and improving the design of future clinical trials are much needed to optimize the chances to obtain positive outcomes.
Collapse
Affiliation(s)
- Rita Khoury
- Department of Psychiatry and Clinical Psychology, Saint Georges Hospital University Medical Center, Youssef Sursock Street, PO Box 166378, Beirut, Lebanon. .,Faculty of Medicine, University of Balamand, Beirut, Lebanon. .,Department of Psychiatry and Behavioral Neuroscience, Saint Louis University School of Medicine, Saint Louis, MO, USA.
| | - Yu Liu
- Department of Psychiatry and Behavioral Neuroscience, Saint Louis University School of Medicine, Saint Louis, MO, USA
| | - Quratulanne Sheheryar
- Department of Psychiatry and Behavioral Neuroscience, Saint Louis University School of Medicine, Saint Louis, MO, USA
| | - George T Grossberg
- Department of Psychiatry and Behavioral Neuroscience, Saint Louis University School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
48
|
FTLD Treatment: Current Practice and Future Possibilities. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1281:297-310. [PMID: 33433882 DOI: 10.1007/978-3-030-51140-1_18] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/21/2023]
Abstract
While behavioral variant frontotemporal dementia (bvFTD) and primary progressive aphasia (PPA) remain unrelenting and universally fatal conditions, there is a framework for supportive treatment in patients diagnosed with these frontotemporal dementia (FTD) syndromes and the larger spectrum of clinical syndromes associated with frontotemporal lobar degeneration (FTLD) pathology on autopsy. A managing physician has an important role in weighing therapeutic options, organizing caregiver support, and framing long-term expectations for patients and caregivers. Additionally, a dedicated neurologist may assist patients and caregivers in navigating a growing range of FTD research, including exciting opportunities in clinical therapeutic trials. This chapter will review current therapeutic options for patients with bvFTD and PPA and detail the landscape of potential new disease-modifying therapies targeting the pathophysiology or FTLD.
Collapse
|
49
|
Ntymenou S, Tsantzali I, Kalamatianos T, Voumvourakis KI, Kapaki E, Tsivgoulis G, Stranjalis G, Paraskevas GP. Blood Biomarkers in Frontotemporal Dementia: Review and Meta-Analysis. Brain Sci 2021; 11:brainsci11020244. [PMID: 33672008 PMCID: PMC7919273 DOI: 10.3390/brainsci11020244] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/07/2021] [Accepted: 02/09/2021] [Indexed: 12/12/2022] Open
Abstract
Biomarkers in cerebrospinal fluid (CSF) are useful in the differential diagnosis between frontotemporal dementia (FTD) and Alzheimer’s dementia (AD), but require lumbar puncture, which is a moderately invasive procedure that can cause anxiety to patients. Gradually, the measurement of blood biomarkers has been attracting great interest. Testing blood instead of CSF, in order to measure biomarkers, offers numerous advantages because it negates the need for lumbar puncture, it is widely available, and can be repeated, allowing the prediction of disease course. In this study, a systematic review of the existing literature was conducted, as well as meta-analysis with greater emphasis on the most studied biomarkers, p-tau and progranulin. The goal was to give prominence to evidence regarding the use of plasma biomarkers in clinical practice.
Collapse
Affiliation(s)
- Sofia Ntymenou
- Department of Neurology, Evangelismos Hospital, 10676 Athens, Greece
| | - Ioanna Tsantzali
- 2nd Department of Neurology, School of Medicine, "Attikon" University General Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Theodosis Kalamatianos
- Department of Neurosurgery, School of Medicine, Evangelismos Hospital, National and Kapodistrian University of Athens, 10676 Athens, Greece
| | - Konstantinos I Voumvourakis
- 2nd Department of Neurology, School of Medicine, "Attikon" University General Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Elisabeth Kapaki
- Ward of Cognitive and Movement Disorders, 1st Department of Neurology, School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece
- Unit of Neurochemistry and Biological Markers, Department of Neurology, School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Georgios Tsivgoulis
- 2nd Department of Neurology, School of Medicine, "Attikon" University General Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - George Stranjalis
- Department of Neurosurgery, School of Medicine, Evangelismos Hospital, National and Kapodistrian University of Athens, 10676 Athens, Greece
| | - George P Paraskevas
- 2nd Department of Neurology, School of Medicine, "Attikon" University General Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
- Unit of Neurochemistry and Biological Markers, Department of Neurology, School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece
| |
Collapse
|
50
|
Arienti F, Lazzeri G, Vizziello M, Monfrini E, Bresolin N, Saetti MC, Picillo M, Franco G, Di Fonzo A. Unravelling Genetic Factors Underlying Corticobasal Syndrome: A Systematic Review. Cells 2021; 10:171. [PMID: 33467748 PMCID: PMC7830591 DOI: 10.3390/cells10010171] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 12/26/2022] Open
Abstract
Corticobasal syndrome (CBS) is an atypical parkinsonian presentation characterized by heterogeneous clinical features and different underlying neuropathology. Most CBS cases are sporadic; nevertheless, reports of families and isolated individuals with genetically determined CBS have been reported. In this systematic review, we analyze the demographical, clinical, radiological, and anatomopathological features of genetically confirmed cases of CBS. A systematic search was performed using the PubMed, EMBASE, and Cochrane Library databases, included all publications in English from 1 January 1999 through 1 August 2020. We found forty publications with fifty-eight eligible cases. A second search for publications dealing with genetic risk factors for CBS led to the review of eight additional articles. GRN was the most common gene involved in CBS, representing 28 out of 58 cases, followed by MAPT, C9ORF72, and PRNP. A set of symptoms was shown to be significantly more common in GRN-CBS patients, including visuospatial impairment, behavioral changes, aphasia, and language alterations. In addition, specific demographical, clinical, biochemical, and radiological features may suggest mutations in other genes. We suggest a diagnostic algorithm to help in identifying potential genetic cases of CBS in order to improve the diagnostic accuracy and to better understand the still poorly defined underlying pathogenetic process.
Collapse
Affiliation(s)
- Federica Arienti
- Dino Ferrari Center, Department of Pathophysiology and Transplantation, Neuroscience Section, University of Milan, 20122 Milan, Italy; (F.A.); (G.L.); (M.V.); (E.M.); (M.C.S.)
| | - Giulia Lazzeri
- Dino Ferrari Center, Department of Pathophysiology and Transplantation, Neuroscience Section, University of Milan, 20122 Milan, Italy; (F.A.); (G.L.); (M.V.); (E.M.); (M.C.S.)
| | - Maria Vizziello
- Dino Ferrari Center, Department of Pathophysiology and Transplantation, Neuroscience Section, University of Milan, 20122 Milan, Italy; (F.A.); (G.L.); (M.V.); (E.M.); (M.C.S.)
| | - Edoardo Monfrini
- Dino Ferrari Center, Department of Pathophysiology and Transplantation, Neuroscience Section, University of Milan, 20122 Milan, Italy; (F.A.); (G.L.); (M.V.); (E.M.); (M.C.S.)
| | - Nereo Bresolin
- Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Neurology Unit, 20122 Milan, Italy; (N.B.); (G.F.)
| | - Maria Cristina Saetti
- Dino Ferrari Center, Department of Pathophysiology and Transplantation, Neuroscience Section, University of Milan, 20122 Milan, Italy; (F.A.); (G.L.); (M.V.); (E.M.); (M.C.S.)
| | - Marina Picillo
- Center for Neurodegenerative Diseases, Department of Medicine, Surgery and Dentistry, Neuroscience Section, University of Salerno, 84084 Salerno, Italy;
| | - Giulia Franco
- Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Neurology Unit, 20122 Milan, Italy; (N.B.); (G.F.)
| | - Alessio Di Fonzo
- Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Neurology Unit, 20122 Milan, Italy; (N.B.); (G.F.)
| |
Collapse
|