1
|
Dellar ER, Vendrell I, Amein B, Lester DG, Edmond EC, Yoganathan K, Dharmadasa T, Sogorb‐Esteve A, Fischer R, Talbot K, Rohrer JD, Turner MR, Thompson AG. Elevated Cerebrospinal Fluid Ubiquitin Carboxyl-Terminal Hydrolase Isozyme L1 in Asymptomatic C9orf72 Hexanucleotide Repeat Expansion Carriers. Ann Neurol 2025; 97:449-459. [PMID: 39548852 PMCID: PMC11831881 DOI: 10.1002/ana.27133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/29/2024] [Accepted: 10/25/2024] [Indexed: 11/18/2024]
Abstract
OBJECTIVE To identify biochemical changes in individuals at higher risk of developing amyotrophic lateral sclerosis (ALS) or frontotemporal dementia (FTD) via C9orf72 hexanucleotide repeat expansion (HRE) heterozygosity. METHODS Cross-sectional observational study of 48 asymptomatic C9orf72 HRE carriers, 39 asymptomatic non-carrier controls, 19 people with sporadic ALS, 10 with C9orf72 ALS, 14 with sporadic FTD, and 10 with C9orf72 FTD. Relative abundance of 30 pre-defined cerebrospinal fluid biomarkers of ALS and FTD were compared in asymptomatic C9orf72 HRE carriers and age-matched non-carrier controls. Differential abundance of these proteins was quantified using data independent acquisition mass spectrometry or electro chemiluminescent assay for neurofilament light chain. Unbiased analysis of the entire cerebrospinal fluid proteome was then carried out. RESULTS Ubiquitin carboxyl-hydrolase isozyme L1 levels were higher in asymptomatic C9orf72 HRE carriers compared with age-matched non-carriers (log2fold change 0.20, FDR-adjusted p-value = 0.034), whereas neurofilament light chain levels did not significantly differ. Ubiquitin carboxyl-hydrolase isozyme L1 levels remained elevated after matching of groups by neurofilament levels (p = 0.011), and after adjusting for age, sex, and neurofilament levels. A significant difference was also observed when restricting analysis to younger participants (<37) matched by neurofilament level (p = 0.007). INTERPRETATION Elevated cerebrospinal fluid ubiquitin carboxyl-hydrolase isozyme L1 levels in C9orf72 HRE carriers can occur in the absence of increased neurofilament levels, potentially reflecting either compensatory or pathogenic mechanisms preceding rapid neuronal loss. This brings forward the window on changes associated with the C9orf72 HRE carrier state, with potential to inform understanding of penetrance and approaches to prevention. ANN NEUROL 2025;97:449-459.
Collapse
Affiliation(s)
| | - Iolanda Vendrell
- Target Discovery InstituteCentre for Medicines Discovery, Nuffield Department of MedicineUniversity of OxfordOxfordUK
- Chinese Academy of Medical Science Oxford InstituteUniversity of OxfordOxfordUK
| | - Benazir Amein
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - David G. Lester
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Evan C. Edmond
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Katie Yoganathan
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Thanuja Dharmadasa
- The Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkville, VictoriaAustralia
| | - Aitana Sogorb‐Esteve
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
- UK Dementia Research Institute at University College LondonLondonUK
| | - Roman Fischer
- Target Discovery InstituteCentre for Medicines Discovery, Nuffield Department of MedicineUniversity of OxfordOxfordUK
- Chinese Academy of Medical Science Oxford InstituteUniversity of OxfordOxfordUK
| | - Kevin Talbot
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
- Kavli Institute for Nanoscience DiscoveryUniversity of OxfordOxfordUK
| | - Jonathan D. Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
| | - Martin R. Turner
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | | |
Collapse
|
2
|
Antonioni A, Raho EM, Granieri E, Koch G. Frontotemporal dementia. How to deal with its diagnostic complexity? Expert Rev Neurother 2025:1-35. [PMID: 39911129 DOI: 10.1080/14737175.2025.2461758] [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: 09/24/2024] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 02/07/2025]
Abstract
INTRODUCTION Frontotemporal dementia (FTD) encompasses a group of heterogeneous neurodegenerative disorders. Aside from genetic cases, its diagnosis is challenging, particularly in the early stages when symptoms are ambiguous, and structural neuroimaging does not reveal characteristic patterns. AREAS COVERED The authors performed a comprehensive literature search through MEDLINE, Scopus, and Web of Science databases to gather evidence to aid the diagnostic process for suspected FTD patients, particularly in early phases, even in sporadic cases, ranging from established to promising tools. Blood-based biomarkers might help identify very early neuropathological stages and guide further evaluations. Subsequently, neurophysiological measures reflecting functional changes in cortical excitatory/inhibitory circuits, along with functional neuroimaging assessing brain network, connectivity, metabolism, and perfusion alterations, could detect specific changes associated to FTD even decades before symptom onset. As the neuropathological process advances, cognitive-behavioral profiles and atrophy patterns emerge, distinguishing specific FTD subtypes. EXPERT OPINION Emerging disease-modifying therapies require early patient enrollment. Therefore, a diagnostic paradigm shift is needed - from relying on typical cognitive and neuroimaging profiles of advanced cases to widely applicable biomarkers, primarily fluid biomarkers, and, subsequently, neurophysiological and functional neuroimaging biomarkers where appropriate. Additionally, exploring subjective complaints and behavioral changes detected by home-based technologies might be crucial for early diagnosis.
Collapse
Affiliation(s)
- Annibale Antonioni
- Doctoral Program in Translational Neurosciences and Neurotechnologies, University of Ferrara, Ferrara, FE, Italy
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, FE, Italy
| | - Emanuela Maria Raho
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, FE, Italy
| | - Enrico Granieri
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, FE, Italy
| | - Giacomo Koch
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, FE, Italy
- Center for Translational Neurophysiology of Speech and Communication (CTNSC), Italian Institute of Technology (IIT), Ferrara, FE, Italy
- Non Invasive Brain Stimulation Unit, Istituto di Ricovero e Cura a Carattere Scientifico Santa Lucia, Roma, RM, Italy
| |
Collapse
|
3
|
Xu Z, Xu R. Current potential diagnostic biomarkers of amyotrophic lateral sclerosis. Rev Neurosci 2024; 35:917-931. [PMID: 38976599 DOI: 10.1515/revneuro-2024-0037] [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: 03/08/2024] [Accepted: 06/13/2024] [Indexed: 07/10/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) currently lacks the useful diagnostic biomarkers. The current diagnosis of ALS is mainly depended on the clinical manifestations, which contributes to the diagnostic delay and be difficult to make the accurate diagnosis at the early stage of ALS, and hinders the clinical early therapeutics. The more and more pathogenesis of ALS are found at the last 30 years, including excitotoxicity, the oxidative stress, the mitochondrial dysfunction, neuroinflammation, the altered energy metabolism, the RNA misprocessing and the most recent neuroimaging findings. The findings of these pathogenesis bring the new clues for searching the diagnostic biomarkers of ALS. At present, a large number of relevant studies about the diagnostic biomarkers are underway. The ALS pathogenesis related to the diagnostic biomarkers might lessen the diagnostic reliance on the clinical manifestations. Among them, the cortical altered signatures of ALS patients derived from both structural and functional magnetic resonance imaging and the emerging proteomic biomarkers of neuronal loss and glial activation in the cerebrospinal fluid as well as the potential biomarkers in blood, serum, urine, and saliva are leading a new phase of biomarkers. Here, we reviewed these current potential diagnostic biomarkers of ALS.
Collapse
Affiliation(s)
- Zheqi Xu
- Department of Neurology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, China
- The Clinical College of Nanchang Medical College, Nanchang 330006, China
- Medical College of Nanchang University, Nanchang 330006, China
| | - Renshi Xu
- Department of Neurology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, China
- The Clinical College of Nanchang Medical College, Nanchang 330006, China
- Medical College of Nanchang University, Nanchang 330006, China
| |
Collapse
|
4
|
Yuan D, Jiang S, Xu R. Clinical features and progress in diagnosis and treatment of amyotrophic lateral sclerosis. Ann Med 2024; 56:2399962. [PMID: 39624969 PMCID: PMC11616751 DOI: 10.1080/07853890.2024.2399962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 07/27/2024] [Accepted: 08/26/2024] [Indexed: 12/06/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease of the central nervous system. Despite a large number of studies, the current prognosis of ALS is still not ideal. This article briefly describes the clinical features including epidemiology, genetic structure and clinical manifestations, as well as the progress of new diagnostic criteria and treatment of ALS. Meanwhile, we also discussed further both developments and improvements to enhance understanding and accelerating the introduction of the effective treatments of ALS.
Collapse
Affiliation(s)
- Dongxiang Yuan
- Department of Neurology, Jiangxi Provincial People’s Hospital; The Clinical College of Nanchang Medical College; The First Affiliated Hospital of Nanchang Medical College; Xiangya Hospital of Center South University, Jiangxi Hospital; National Regional Center for Neurological Disease, Honggutan District, Nanchang, Jiangxi Province, China
| | - Shishi Jiang
- Department of Neurology, Jiangxi Provincial People’s Hospital; The Clinical College of Nanchang Medical College; The First Affiliated Hospital of Nanchang Medical College; Xiangya Hospital of Center South University, Jiangxi Hospital; National Regional Center for Neurological Disease, Honggutan District, Nanchang, Jiangxi Province, China
| | - Renshi Xu
- Department of Neurology, Jiangxi Provincial People’s Hospital; The Clinical College of Nanchang Medical College; The First Affiliated Hospital of Nanchang Medical College; Xiangya Hospital of Center South University, Jiangxi Hospital; National Regional Center for Neurological Disease, Honggutan District, Nanchang, Jiangxi Province, China
| |
Collapse
|
5
|
R K Roy A, Noohi F, Morris NA, Ljubenkov P, Heuer H, Fong J, Hall M, Lario Lago A, Rankin KP, Miller BL, Boxer AL, Rosen HJ, Seeley WW, Perry DC, Yokoyama JS, Lee SE, Sturm VE. Basal parasympathetic deficits in C9orf72 hexanucleotide repeat expansion carriers relate to smaller frontoinsula and thalamus volume and lower empathy. Neuroimage Clin 2024; 43:103649. [PMID: 39098187 PMCID: PMC11342757 DOI: 10.1016/j.nicl.2024.103649] [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: 02/26/2024] [Revised: 07/16/2024] [Accepted: 07/29/2024] [Indexed: 08/06/2024]
Abstract
Diminished basal parasympathetic nervous system activity is a feature of frontotemporal dementia that relates to left frontoinsula dysfunction and empathy impairment. Individuals with a pathogenic expansion of the hexanucleotide repeat in chromosome 9 open reading frame 72 (C9orf72), the most common genetic cause of frontotemporal dementia and amyotrophic lateral sclerosis, provide a unique opportunity to examine whether parasympathetic activity is disrupted in genetic forms of frontotemporal dementia and to investigate when parasympathetic deficits manifest in the pathophysiological cascade. We measured baseline respiratory sinus arrhythmia, a parasympathetic measure of heart rate variability, over two minutes in a sample of 102 participants that included 19 asymptomatic expansion carriers (C9+ asymp), 14 expansion carriers with mild cognitive impairment (C9+ MCI), 16 symptomatic expansion carriers with frontotemporal dementia (C9+ FTD), and 53 expansion-negative healthy controls (C9- HC) who also underwent structural magnetic resonance imaging. In follow-up analyses, we compared baseline respiratory sinus arrhythmia in the C9+ FTD group with an independent age-, sex-, and clinical severity-matched group of 26 people with sporadic behavioral variant frontotemporal dementia. The Frontotemporal Lobar Degeneration-modified Clinical Dementia Rating-Sum of Boxes score was used to quantify behavioral symptom severity, and informant ratings on the Interpersonal Reactivity Index provided measures of participants' current emotional (empathic concern) and cognitive (perspective-taking) empathy. Results indicated that the C9+ FTD group had lower baseline respiratory sinus arrhythmia than the C9+ MCI, C9+ asymp, and C9- HC groups, a deficit that was comparable to that of sporadic behavioral variant frontotemporal dementia. Linear regression analyses indicated that lower baseline respiratory sinus arrhythmia was associated with worse behavioral symptom severity and lower empathic concern and perspective-taking across the C9orf72 expansion carrier clinical spectrum. Whole-brain voxel-based morphometry analyses in participants with C9orf72 pathogenic expansions found that lower baseline respiratory sinus arrhythmia correlated with smaller gray matter volume in the left frontoinsula and bilateral thalamus, key structures that support parasympathetic function, and in the bilateral parietal lobes, occipital lobes, and cerebellum, regions that are also vulnerable in individuals with C9orf72 expansions. This study provides novel evidence that basal parasympathetic functioning is diminished in FTD due to C9orf72 expansions and suggests that baseline respiratory sinus arrhythmia may be a potential non-invasive biomarker that is sensitive to behavioral symptoms in the early stages of disease.
Collapse
Affiliation(s)
- Ashlin R K Roy
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| | - Fate Noohi
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| | - Nathaniel A Morris
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| | - Peter Ljubenkov
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| | - Hilary Heuer
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| | - Jamie Fong
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| | - Matthew Hall
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| | | | - Katherine P Rankin
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| | - Bruce L Miller
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| | - Adam L Boxer
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| | - Howard J Rosen
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| | - William W Seeley
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| | - David C Perry
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| | - Jennifer S Yokoyama
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| | - Suzee E Lee
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| | - Virginia E Sturm
- Department of Neurology, University of California, San Francisco, CA 94158, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA 94143, USA.
| |
Collapse
|
6
|
Irwin KE, Sheth U, Wong PC, Gendron TF. Fluid biomarkers for amyotrophic lateral sclerosis: a review. Mol Neurodegener 2024; 19:9. [PMID: 38267984 PMCID: PMC10809579 DOI: 10.1186/s13024-023-00685-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 11/21/2023] [Indexed: 01/26/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the loss of upper and lower motor neurons. Presently, three FDA-approved drugs are available to help slow functional decline for patients with ALS, but no cure yet exists. With an average life expectancy of only two to five years after diagnosis, there is a clear need for biomarkers to improve the care of patients with ALS and to expedite ALS treatment development. Here, we provide a review of the efforts made towards identifying diagnostic, prognostic, susceptibility/risk, and response fluid biomarkers with the intent to facilitate a more rapid and accurate ALS diagnosis, to better predict prognosis, to improve clinical trial design, and to inform interpretation of clinical trial results. Over the course of 20 + years, several promising fluid biomarker candidates for ALS have emerged. These will be discussed, as will the exciting new strategies being explored for ALS biomarker discovery and development.
Collapse
Affiliation(s)
- Katherine E Irwin
- Department of Pathology, Johns Hopkins Medicine, Baltimore, MD, 21205, USA
- Department of Neuroscience, Johns Hopkins Medicine, Baltimore, MD, 21205, USA
| | - Udit Sheth
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Philip C Wong
- Department of Pathology, Johns Hopkins Medicine, Baltimore, MD, 21205, USA.
- Department of Neuroscience, Johns Hopkins Medicine, Baltimore, MD, 21205, USA.
| | - Tania F Gendron
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA.
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, 32224, USA.
| |
Collapse
|
7
|
Sattler R, Traynor BJ, Robertson J, Van Den Bosch L, Barmada SJ, Svendsen CN, Disney MD, Gendron TF, Wong PC, Turner MR, Boxer A, Babu S, Benatar M, Kurnellas M, Rohrer JD, Donnelly CJ, Bustos LM, Van Keuren-Jensen K, Dacks PA, Sabbagh MN. Roadmap for C9ORF72 in Frontotemporal Dementia and Amyotrophic Lateral Sclerosis: Report on the C9ORF72 FTD/ALS Summit. Neurol Ther 2023; 12:1821-1843. [PMID: 37847372 PMCID: PMC10630271 DOI: 10.1007/s40120-023-00548-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 09/14/2023] [Indexed: 10/18/2023] Open
Abstract
A summit held March 2023 in Scottsdale, Arizona (USA) focused on the intronic hexanucleotide expansion in the C9ORF72 gene and its relevance in frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS; C9ORF72-FTD/ALS). The goal of this summit was to connect basic scientists, clinical researchers, drug developers, and individuals affected by C9ORF72-FTD/ALS to evaluate how collaborative efforts across the FTD-ALS disease spectrum might break down existing disease silos. Presentations and discussions covered recent discoveries in C9ORF72-FTD/ALS disease mechanisms, availability of disease biomarkers and recent advances in therapeutic development, and clinical trial design for prevention and treatment for individuals affected by C9ORF72-FTD/ALS and asymptomatic pathological expansion carriers. The C9ORF72-associated hexanucleotide repeat expansion is an important locus for both ALS and FTD. C9ORF72-FTD/ALS may be characterized by loss of function of the C9ORF72 protein and toxic gain of functions caused by both dipeptide repeat (DPR) proteins and hexanucleotide repeat RNA. C9ORF72-FTD/ALS therapeutic strategies discussed at the summit included the use of antisense oligonucleotides, adeno-associated virus (AAV)-mediated gene silencing and gene delivery, and engineered small molecules targeting RNA structures associated with the C9ORF72 expansion. Neurofilament light chain, DPR proteins, and transactive response (TAR) DNA-binding protein 43 (TDP-43)-associated molecular changes were presented as biomarker candidates. Similarly, brain imaging modalities (i.e., magnetic resonance imaging [MRI] and positron emission tomography [PET]) measuring structural, functional, and metabolic changes were discussed as important tools to monitor individuals affected with C9ORF72-FTD/ALS, at both pre-symptomatic and symptomatic disease stages. Finally, summit attendees evaluated current clinical trial designs available for FTD or ALS patients and concluded that therapeutics relevant to FTD/ALS patients, such as those specifically targeting C9ORF72, may need to be tested with composite endpoints covering clinical symptoms of both FTD and ALS. The latter will require novel clinical trial designs to be inclusive of all patient subgroups spanning the FTD/ALS spectrum.
Collapse
Affiliation(s)
- Rita Sattler
- Barrow Neurological Institute, 2910 N Third Ave, Phoenix, AZ, 85013, USA.
| | - Bryan J Traynor
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Janice Robertson
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Ludo Van Den Bosch
- VIB, Center for Brain & Disease Research, Laboratory of Neurobiology and KU Leuven, Leuven, Belgium
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), University of Leuven, Leuven, Belgium
| | - Sami J Barmada
- Department of Neurology, Neuroscience Program, University of Michigan, Ann Arbor, MI, USA
| | - Clive N Svendsen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Matthew D Disney
- Department of Chemistry, The Herbert Wertheim UF-Scripps Institute for Biomedical Research and Innovation, The Scripps Research Institute, Jupiter, FL, USA
| | - Tania F Gendron
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Philip C Wong
- Departments of Pathology and Neuroscience, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Martin R Turner
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Adam Boxer
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of San Francisco, San Francisco, CA, USA
| | - Suma Babu
- Sean M. Healey and AMG Center for ALS and the Neurological Clinical Research Institute, Massachusetts General Hospital-Harvard Medical School, Boston, MA, USA
| | - Michael Benatar
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, 33129, USA
| | | | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Christopher J Donnelly
- LiveLikeLou Center for ALS Research, Brain Institute, University of Pittsburgh, Pittsburgh, USA
- Department of Neurobiology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lynette M Bustos
- Barrow Neurological Institute, 2910 N Third Ave, Phoenix, AZ, 85013, USA
| | | | - Penny A Dacks
- The Association for Frontotemporal Degeneration and FTD Disorders Registry, King of Prussia, PA, USA
| | - Marwan N Sabbagh
- Barrow Neurological Institute, 2910 N Third Ave, Phoenix, AZ, 85013, USA.
| |
Collapse
|
8
|
Hendricks E, Quihuis AM, Hung ST, Chang J, Dorjsuren N, Der B, Staats KA, Shi Y, Sta Maria NS, Jacobs RE, Ichida JK. The C9ORF72 repeat expansion alters neurodevelopment. Cell Rep 2023; 42:112983. [PMID: 37590144 PMCID: PMC10757587 DOI: 10.1016/j.celrep.2023.112983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 07/05/2023] [Accepted: 07/31/2023] [Indexed: 08/19/2023] Open
Abstract
Genetic mutations that cause adult-onset neurodegenerative diseases are often expressed during embryonic stages, but it is unclear whether they alter neurodevelopment and how this might influence disease onset. Here, we show that the most common cause of frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS), a repeat expansion in C9ORF72, restricts neural stem cell proliferation and reduces cortical and thalamic size in utero. Surprisingly, a repeat expansion-derived dipeptide repeat protein (DPR) not known to reduce neuronal viability plays a key role in impairing neurodevelopment. Pharmacologically mimicking the effects of the repeat expansion on neurodevelopment increases susceptibility of C9ORF72 mice to motor defects. Thus, the C9ORF72 repeat expansion stunts development of the brain regions prominently affected in C9ORF72 FTD/ALS patients.
Collapse
Affiliation(s)
- Eric Hendricks
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA 90033, USA; Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Alicia M Quihuis
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Shu-Ting Hung
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA 90033, USA; Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Jonathan Chang
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA 90033, USA; Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Nomongo Dorjsuren
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA 90033, USA; Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Balint Der
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA 90033, USA
| | - Kim A Staats
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA 90033, USA
| | - Yingxiao Shi
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA 90033, USA; Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Naomi S Sta Maria
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Russell E Jacobs
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Justin K Ichida
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA 90033, USA; Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
9
|
Grossman M, Seeley WW, Boxer AL, Hillis AE, Knopman DS, Ljubenov PA, Miller B, Piguet O, Rademakers R, Whitwell JL, Zetterberg H, van Swieten JC. Frontotemporal lobar degeneration. Nat Rev Dis Primers 2023; 9:40. [PMID: 37563165 DOI: 10.1038/s41572-023-00447-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/12/2023] [Indexed: 08/12/2023]
Abstract
Frontotemporal lobar degeneration (FTLD) is one of the most common causes of early-onset dementia and presents with early social-emotional-behavioural and/or language changes that can be accompanied by a pyramidal or extrapyramidal motor disorder. About 20-25% of individuals with FTLD are estimated to carry a mutation associated with a specific FTLD pathology. The discovery of these mutations has led to important advances in potentially disease-modifying treatments that aim to slow progression or delay disease onset and has improved understanding of brain functioning. In both mutation carriers and those with sporadic disease, the most common underlying diagnoses are linked to neuronal and glial inclusions containing tau (FTLD-tau) or TDP-43 (FTLD-TDP), although 5-10% of patients may have inclusions containing proteins from the FUS-Ewing sarcoma-TAF15 family (FTLD-FET). Biomarkers definitively identifying specific pathological entities in sporadic disease have been elusive, which has impeded development of disease-modifying treatments. Nevertheless, disease-monitoring biofluid and imaging biomarkers are becoming increasingly sophisticated and are likely to serve as useful measures of treatment response during trials of disease-modifying treatments. Symptomatic trials using novel approaches such as transcranial direct current stimulation are also beginning to show promise.
Collapse
Affiliation(s)
- Murray Grossman
- Department of Neurology and Penn Frontotemporal Degeneration Center, University of Pennsylvania, Philadelphia, PA, USA
| | - William W Seeley
- Departments of Neurology and Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA.
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA.
| | - Adam L Boxer
- Departments of Neurology and Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
| | - Argye E Hillis
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | | | - Peter A Ljubenov
- Departments of Neurology and Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
| | - Bruce Miller
- Departments of Neurology and Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
| | - Olivier Piguet
- School of Psychology and Brain and Mind Center, University of Sydney, Sydney, New South Wales, Australia
| | - Rosa Rademakers
- VIB Center for Molecular Neurology, University of Antwerp, Antwerp, Belgium
| | | | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The University of Gothenburg, Mölndal, Sweden
- Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | | |
Collapse
|
10
|
Abstract
PURPOSE OF REVIEW Frontotemporal dementia (FTD) is a clinically, pathologically and genetically heterogeneous disorder. Whilst disease modifying therapy trials are mostly focused on the symptomatic phase, future studies will move earlier in the disease aiming to prevent symptom onset. This review summarizes the recent work to better understand this presymptomatic period. RECENT FINDINGS The presymptomatic phase can be split into preclinical and prodromal stages. The onset of the preclinical phase is defined by the first presence of pathological inclusions of tau, TDP-43 or fused in sarcoma in the brain. Definitive biomarkers of these pathologies do not yet exist for FTD. The prodromal phase is defined by the onset of mild symptoms. Recent work has highlighted the wide phenotypic spectrum that occurs, with the concept of mild cognitive ± behavioural ± motor impairment (MCBMI) being put forward, and additions to scales such as the CDR plus NACC FTLD now incorporating neuropsychiatric and motor symptoms. SUMMARY It will be important to better characterize the presymptomatic period moving forward and develop robust biomarkers that can be used both for stratification and outcome measures in prevention trials. The work of the FTD Prevention Initiative aims to facilitate this by bringing together data from natural history studies across the world.
Collapse
Affiliation(s)
- Lucy L Russell
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | | |
Collapse
|
11
|
Antonioni A, Raho EM, Lopriore P, Pace AP, Latino RR, Assogna M, Mancuso M, Gragnaniello D, Granieri E, Pugliatti M, Di Lorenzo F, Koch G. Frontotemporal Dementia, Where Do We Stand? A Narrative Review. Int J Mol Sci 2023; 24:11732. [PMID: 37511491 PMCID: PMC10380352 DOI: 10.3390/ijms241411732] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Frontotemporal dementia (FTD) is a neurodegenerative disease of growing interest, since it accounts for up to 10% of middle-age-onset dementias and entails a social, economic, and emotional burden for the patients and caregivers. It is characterised by a (at least initially) selective degeneration of the frontal and/or temporal lobe, generally leading to behavioural alterations, speech disorders, and psychiatric symptoms. Despite the recent advances, given its extreme heterogeneity, an overview that can bring together all the data currently available is still lacking. Here, we aim to provide a state of the art on the pathogenesis of this disease, starting with established findings and integrating them with more recent ones. In particular, advances in the genetics field will be examined, assessing them in relation to both the clinical manifestations and histopathological findings, as well as considering the link with other diseases, such as amyotrophic lateral sclerosis (ALS). Furthermore, the current diagnostic criteria will be explored, including neuroimaging methods, nuclear medicine investigations, and biomarkers on biological fluids. Of note, the promising information provided by neurophysiological investigations, i.e., electroencephalography and non-invasive brain stimulation techniques, concerning the alterations in brain networks and neurotransmitter systems will be reviewed. Finally, current and experimental therapies will be considered.
Collapse
Affiliation(s)
- Annibale Antonioni
- Unit of Clinical Neurology, Neurosciences and Rehabilitation Department, University of Ferrara, 44121 Ferrara, Italy
- Doctoral Program in Translational Neurosciences and Neurotechnologies, University of Ferrara, 44121 Ferrara, Italy
| | - Emanuela Maria Raho
- Unit of Clinical Neurology, Neurosciences and Rehabilitation Department, University of Ferrara, 44121 Ferrara, Italy
| | - Piervito Lopriore
- Neurological Institute, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Antonia Pia Pace
- Institute of Radiology, Department of Medicine, University of Udine, University Hospital S. Maria della Misericordia, Azienda Sanitaria-Universitaria Friuli Centrale, 33100 Udine, Italy
| | - Raffaela Rita Latino
- Complex Structure of Neurology, Emergency Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Martina Assogna
- Centro Demenze, Policlinico Tor Vergata, University of Rome 'Tor Vergata', 00133 Rome, Italy
- Non Invasive Brain Stimulation Unit, Istituto di Ricovero e Cura a Carattere Scientifico Santa Lucia, 00179 Rome, Italy
| | - Michelangelo Mancuso
- Neurological Institute, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Daniela Gragnaniello
- Nuerology Unit, Neurosciences and Rehabilitation Department, Ferrara University Hospital, 44124 Ferrara, Italy
| | - Enrico Granieri
- Unit of Clinical Neurology, Neurosciences and Rehabilitation Department, University of Ferrara, 44121 Ferrara, Italy
| | - Maura Pugliatti
- Unit of Clinical Neurology, Neurosciences and Rehabilitation Department, University of Ferrara, 44121 Ferrara, Italy
| | - Francesco Di Lorenzo
- Non Invasive Brain Stimulation Unit, Istituto di Ricovero e Cura a Carattere Scientifico Santa Lucia, 00179 Rome, Italy
| | - Giacomo Koch
- Non Invasive Brain Stimulation Unit, Istituto di Ricovero e Cura a Carattere Scientifico Santa Lucia, 00179 Rome, Italy
- Iit@Unife Center for Translational Neurophysiology, Istituto Italiano di Tecnologia, 44121 Ferrara, Italy
- Section of Human Physiology, Neurosciences and Rehabilitation Department, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
12
|
Van't Spijker HM, Almeida S. How villains are made: The translation of dipeptide repeat proteins in C9ORF72-ALS/FTD. Gene 2023; 858:147167. [PMID: 36621656 PMCID: PMC9928902 DOI: 10.1016/j.gene.2023.147167] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023]
Abstract
A hexanucleotide repeat expansion in the C9ORF72 gene is the most common genetic alteration associated with amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). These neurodegenerative diseases share genetic, clinical and pathological features. The mutation in C9ORF72 appears to drive pathogenesis through a combination of loss of C9ORF72 normal function and gain of toxic effects due to the repeat expansion, which result in aggregation prone expanded RNAs and dipeptide repeat (DPR) proteins. Studies in cellular and animal models indicate that the DPR proteins are the more toxic species. Thus, a large body of research has focused on identifying the cellular pathways most directly impacted by these toxic proteins, with the goal of characterizing disease pathogenesis and nominating potential targets for therapeutic development. The preventative block of the production of the toxic proteins before they can cause harm is a second strategy of intense focus. Despite the considerable amount of effort dedicated to this prophylactic approach, it is still unclear how the DPR proteins are synthesized from RNAs harboring repeat expansions. In this review, we summarize our current knowledge of the specific protein translation mechanisms shown to account for the synthesis of DPR proteins. We will then discuss how enhanced understanding of the composition of these toxic effectors could help in refining disease mechanisms, and paving the way to identify and design effective prophylactic therapies for C9ORF72 ALS-FTD.
Collapse
Affiliation(s)
- Heleen M Van't Spijker
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Sandra Almeida
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
13
|
del Campo M, Zetterberg H, Gandy S, Onyike CU, Oliveira F, Udeh‐Momoh C, Lleó A, Teunissen CE, Pijnenburg Y. New developments of biofluid-based biomarkers for routine diagnosis and disease trajectories in frontotemporal dementia. Alzheimers Dement 2022; 18:2292-2307. [PMID: 35235699 PMCID: PMC9790674 DOI: 10.1002/alz.12643] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 02/04/2022] [Accepted: 02/04/2022] [Indexed: 01/31/2023]
Abstract
Frontotemporal dementia (FTD) covers a spectrum of neurodegenerative disorders with different phenotypes, genetic backgrounds, and pathological states. Its clinicopathological diversity challenges the diagnostic process and the execution of clinical trials, calling for specific diagnostic biomarkers of pathologic FTD types. There is also a need for biomarkers that facilitate disease staging, quantification of severity, monitoring in clinics and observational studies, and for evaluation of target engagement and treatment response in clinical trials. This review discusses current FTD biofluid-based biomarker knowledge taking into account the differing applications. The limitations, knowledge gaps, and challenges for the development and implementation of such markers are also examined. Strategies to overcome these hurdles are proposed, including the technologies available, patient cohorts, and collaborative research initiatives. Access to robust and reliable biomarkers that define the exact underlying pathophysiological FTD process will meet the needs for specific diagnosis, disease quantitation, clinical monitoring, and treatment development.
Collapse
Affiliation(s)
- Marta del Campo
- Departamento de Ciencias Farmacéuticas y de la SaludFacultad de FarmaciaUniversidad San Pablo‐CEUCEU UniversitiesMadridSpain
| | - Henrik Zetterberg
- Institute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgGothenburgSweden,Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden,UK Dementia Research Institute at UCLLondonUK,Department of Neurodegenerative DiseaseUCL Institute of NeurologyLondonUK,Hong Kong Center for Neurodegenerative DiseasesHong KongChina
| | - Sam Gandy
- Department of NeurologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Chiadi U Onyike
- Division of Geriatric Psychiatry and NeuropsychiatryThe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Fabricio Oliveira
- Department of Neurology and NeurosurgeryEscola Paulista de MedicinaFederal University of São Paulo (UNIFESP)São PauloSão PauloBrazil
| | - Chi Udeh‐Momoh
- Ageing Epidemiology Research UnitSchool of Public HealthFaculty of MedicineImperial College LondonLondonUK,Translational Health SciencesFaculty of MedicineUniversity of BristolBristolUK
| | - Alberto Lleó
- Neurology DepartmentHospital de la Santa Creu I Sant PauBarcelonaSpain
| | - Charlotte E. Teunissen
- Neurochemistry LaboratoryDepartment of Clinical ChemistryAmsterdam NeuroscienceAmsterdam University Medical CentersVrije UniversiteitAmsterdamthe Netherlands
| | - Yolande Pijnenburg
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam NeuroscienceVrije Universiteit AmsterdamAmsterdam UMCAmsterdamthe Netherlands
| |
Collapse
|
14
|
Saunders TS, Gadd DA, Spires‐Jones TL, King D, Ritchie C, Muniz‐Terrera G. Associations between cerebrospinal fluid markers and cognition in ageing and dementia: A systematic review. Eur J Neurosci 2022; 56:5650-5713. [PMID: 35338546 PMCID: PMC9790745 DOI: 10.1111/ejn.15656] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/08/2022] [Accepted: 03/13/2022] [Indexed: 12/30/2022]
Abstract
A biomarker associated with cognition in neurodegenerative dementias would aid in the early detection of disease progression, complement clinical staging and act as a surrogate endpoint in clinical trials. The current systematic review evaluates the association between cerebrospinal fluid protein markers of synapse loss and neuronal injury and cognition. We performed a systematic search which revealed 67 studies reporting an association between cerebrospinal fluid markers of interest and neuropsychological performance. Despite the substantial heterogeneity between studies, we found some evidence for an association between neurofilament-light and worse cognition in Alzheimer's diseases, frontotemporal dementia and typical cognitive ageing. Moreover, there was an association between cerebrospinal fluid neurogranin and cognition in those with an Alzheimer's-like cerebrospinal fluid biomarker profile. Some evidence was found for cerebrospinal fluid neuronal pentraxin-2 as a correlate of cognition across dementia syndromes. Due to the substantial heterogeneity of the field, no firm conclusions can be drawn from this review. Future research should focus on improving standardization and reporting as well as establishing the importance of novel markers such as neuronal pentraxin-2 and whether such markers can predict longitudinal cognitive decline.
Collapse
Affiliation(s)
- Tyler S. Saunders
- UK Dementia Research InstituteThe University of EdinburghEdinburghUK
- Center for Discovery Brain SciencesThe University of EdinburghEdinburghUK
- Center for Clinical Brain SciencesThe University of EdinburghEdinburghUK
- Center for Dementia PreventionThe University of EdinburghEdinburghUK
| | - Danni A. Gadd
- Center for Genomic and Experimental Medicine, Institute of Genetics and Molecular MedicineUniversity of EdinburghEdinburghUK
| | - Tara L. Spires‐Jones
- UK Dementia Research InstituteThe University of EdinburghEdinburghUK
- Center for Discovery Brain SciencesThe University of EdinburghEdinburghUK
| | - Declan King
- UK Dementia Research InstituteThe University of EdinburghEdinburghUK
- Center for Discovery Brain SciencesThe University of EdinburghEdinburghUK
| | - Craig Ritchie
- Center for Clinical Brain SciencesThe University of EdinburghEdinburghUK
- Center for Dementia PreventionThe University of EdinburghEdinburghUK
| | - Graciela Muniz‐Terrera
- Center for Clinical Brain SciencesThe University of EdinburghEdinburghUK
- Center for Dementia PreventionThe University of EdinburghEdinburghUK
| |
Collapse
|
15
|
Ryan B, O’Mara Baker A, Ilse C, Brickell KL, Kersten HM, Williams JM, Addis DR, Tippett LJ, Curtis MA. The New Zealand Genetic Frontotemporal Dementia Study (FTDGeNZ): a longitudinal study of pre-symptomatic biomarkers. J R Soc N Z 2022; 53:511-531. [PMID: 39439966 PMCID: PMC11459842 DOI: 10.1080/03036758.2022.2101483] [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: 10/31/2021] [Accepted: 07/08/2022] [Indexed: 10/16/2022]
Abstract
The New Zealand Genetic Frontotemporal Dementia Study (FTDGeNZ) is an emerging longitudinal study of a large New Zealand pedigree with genetic frontotemporal dementia (FTD). Natural history studies of genetic FTD cohorts provide a unique opportunity to identify biomarkers of pre-symptomatic dementia, as carriers can be identified and studied decades before expected symptom onset. FTDGeNZ was established in 2016 with the aim of identifying the earliest pre-symptomatic biomarkers of FTD, in collaboration with international multi-centre cohorts. We enrolled 25 participants from a single family between April 2016 and August 2018. Participants were genotyped to determine whether they were pre-symptomatic carriers of the mutation (MAPT IVS 10 + 16 C > T), or non-carrier controls. Participants have undergone clinical assessments including neuropsychological and mood assessment; olfactory testing; assessment of social cognition; and blood collection for analyses of microRNA and protein fluid biomarkers annually. We have also performed structural and functional MRI of the brain and assessment of autobiographical memory biennially, and retinal imaging at baseline. Here, we describe the full study protocol and the baseline demographic and clinical characteristics of the FTDGeNZ cohort, and we highlight the latest findings in the field.
Collapse
Affiliation(s)
- Brigid Ryan
- Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, New Zealand
| | - Ashleigh O’Mara Baker
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- School of Psychology, University of Auckland, Auckland, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, New Zealand
| | - Christina Ilse
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, New Zealand
| | - Kiri L. Brickell
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, New Zealand
| | - Hannah M. Kersten
- School of Optometry and Vision Science, University of Auckland, Auckland, New Zealand
| | - Joanna M. Williams
- Department of Anatomy, University of Otago, Dunedin, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, New Zealand
| | - Donna Rose Addis
- School of Psychology, University of Auckland, Auckland, New Zealand
- Rotman Research Institute, Baycrest Health Sciences, Toronto, Canada
- Department of Psychology, University of Toronto, Toronto, Canada
| | - Lynette J. Tippett
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- School of Psychology, University of Auckland, Auckland, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, New Zealand
| | - Maurice A. Curtis
- Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, New Zealand
| |
Collapse
|
16
|
Wilson KM, Katona E, Glaria I, Carcolé M, Swift IJ, Sogorb-Esteve A, Heller C, Bouzigues A, Heslegrave AJ, Keshavan A, Knowles K, Patil S, Mohapatra S, Liu Y, Goyal J, Sanchez-Valle R, Laforce RJ, Synofzik M, Rowe JB, Finger E, Vandenberghe R, Butler CR, Gerhard A, Van Swieten JC, Seelaar H, Borroni B, Galimberti D, de Mendonça A, Masellis M, Tartaglia MC, Otto M, Graff C, Ducharme S, Schott JM, Malaspina A, Zetterberg H, Boyanapalli R, Rohrer JD, Isaacs AM. Development of a sensitive trial-ready poly(GP) CSF biomarker assay for C9orf72-associated frontotemporal dementia and amyotrophic lateral sclerosis. J Neurol Neurosurg Psychiatry 2022; 93:761-771. [PMID: 35379698 PMCID: PMC9279742 DOI: 10.1136/jnnp-2021-328710] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/04/2022] [Indexed: 01/15/2023]
Abstract
OBJECTIVE A GGGGCC repeat expansion in the C9orf72 gene is the most common cause of genetic frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS). As potential therapies targeting the repeat expansion are now entering clinical trials, sensitive biomarker assays of target engagement are urgently required. Our objective was to develop such an assay. METHODS We used the single molecule array (Simoa) platform to develop an immunoassay for measuring poly(GP) dipeptide repeat proteins (DPRs) generated by the C9orf72 repeat expansion in cerebrospinal fluid (CSF) of people with C9orf72-associated FTD/ALS. RESULTS AND CONCLUSIONS We show the assay to be highly sensitive and robust, passing extensive qualification criteria including low intraplate and interplate variability, a high precision and accuracy in measuring both calibrators and samples, dilutional parallelism, tolerance to sample and standard freeze-thaw and no haemoglobin interference. We used this assay to measure poly(GP) in CSF samples collected through the Genetic FTD Initiative (N=40 C9orf72 and 15 controls). We found it had 100% specificity and 100% sensitivity and a large window for detecting target engagement, as the C9orf72 CSF sample with the lowest poly(GP) signal had eightfold higher signal than controls and on average values from C9orf72 samples were 38-fold higher than controls, which all fell below the lower limit of quantification of the assay. These data indicate that a Simoa-based poly(GP) DPR assay is suitable for use in clinical trials to determine target engagement of therapeutics aimed at reducing C9orf72 repeat-containing transcripts.
Collapse
Affiliation(s)
- Katherine M Wilson
- UK Dementia Research Institute at UCL, UCL Queen Square Institute of Neurology, London, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Eszter Katona
- UK Dementia Research Institute at UCL, UCL Queen Square Institute of Neurology, London, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Idoia Glaria
- UK Dementia Research Institute at UCL, UCL Queen Square Institute of Neurology, London, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Mireia Carcolé
- UK Dementia Research Institute at UCL, UCL Queen Square Institute of Neurology, London, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Imogen J Swift
- UK Dementia Research Institute at UCL, UCL Queen Square Institute of Neurology, London, UK
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Aitana Sogorb-Esteve
- UK Dementia Research Institute at UCL, UCL Queen Square Institute of Neurology, London, UK
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Carolin Heller
- UK Dementia Research Institute at UCL, UCL Queen Square Institute of Neurology, London, UK
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Arabella Bouzigues
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Amanda J Heslegrave
- UK Dementia Research Institute at UCL, UCL Queen Square Institute of Neurology, London, UK
| | - Ashvini Keshavan
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Kathryn Knowles
- UK Dementia Research Institute at UCL, UCL Queen Square Institute of Neurology, London, UK
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | | | | | - Yuanjing Liu
- Wave Life Sciences, Cambridge, Massachusetts, USA
| | - Jaya Goyal
- Wave Life Sciences, Cambridge, Massachusetts, USA
| | - 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
| | - Robert Jr Laforce
- Clinique Interdisciplinaire de Mémoire, Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Quebec City, Quebec, Canada
| | - 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
| | - James B Rowe
- Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust and Medical Research Council Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - Elizabeth Finger
- Department of Clinical Neurological Sciences, University of Western Ontario, University of Western Ontario, London, Ontario, Canada
| | - Rik Vandenberghe
- Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium
- Neurology Service, University Hospitals, Leuven, Belgium
| | - Christopher 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, Wolfson Molecular Imaging Centre, The University of Manchester, Manchester, UK
- Departments of Geriatric Medicine and Nuclear Medicine, University of Duisburg- Essen, University of Duisburg- Essen, Essen, Germany
| | | | - Harro Seelaar
- Department of Neurology, Erasmus Medical Centre, Rotterdam, Netherlands
| | - Barbara Borroni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Daniela Galimberti
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
- Centro Dino Ferrari, University of Milan, Milan, Italy
| | | | - Mario Masellis
- Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - M Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, Ontario, Canada
- Canadian Sports Concussion Project, Toronto, Ontario, Canada
| | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Caroline Graff
- Center for Alzheimer Research, Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Bioclinicum, Karolinska Institutet, Solna, Sweden
- Unit for Hereditary Dementias, Theme Aging, Karolinska University Hospital, Solna, Sweden
| | - Simon Ducharme
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Québec, Canada
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada
| | - Jonathan M Schott
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Andrea Malaspina
- Barts and The London School of Medicine and Dentistry Blizard Institute, London, UK
- UCL Queen Square Motor Neuron Disease Centre, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, UCL, London, UK
| | - Henrik Zetterberg
- UK Dementia Research Institute at UCL, UCL Queen Square Institute of Neurology, London, UK
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | | | - Jonathan D Rohrer
- UK Dementia Research Institute at UCL, UCL Queen Square Institute of Neurology, London, UK
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Adrian M Isaacs
- UK Dementia Research Institute at UCL, UCL Queen Square Institute of Neurology, London, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UCL Queen Square Motor Neuron Disease Centre, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, UCL, London, UK
| |
Collapse
|
17
|
Krishnan G, Raitcheva D, Bartlett D, Prudencio M, McKenna-Yasek DM, Douthwright C, Oskarsson BE, Ladha S, King OD, Barmada SJ, Miller TM, Bowser R, Watts JK, Petrucelli L, Brown RH, Kankel MW, Gao FB. Poly(GR) and poly(GA) in cerebrospinal fluid as potential biomarkers for C9ORF72-ALS/FTD. Nat Commun 2022; 13:2799. [PMID: 35589711 PMCID: PMC9119980 DOI: 10.1038/s41467-022-30387-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 04/29/2022] [Indexed: 11/09/2022] Open
Abstract
GGGGCC repeat expansion in C9ORF72, which can be translated in both sense and antisense directions into five dipeptide repeat (DPR) proteins, including poly(GP), poly(GR), and poly(GA), is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Here we developed sensitive assays that can detect poly(GA) and poly(GR) in the cerebrospinal fluid (CSF) of patients with C9ORF72 mutations. CSF poly(GA) and poly(GR) levels did not correlate with age at disease onset, disease duration, or rate of decline of ALS Functional Rating Scale, and the average levels of these DPR proteins were similar in symptomatic and pre-symptomatic patients with C9ORF72 mutations. However, in a patient with C9ORF72-ALS who was treated with antisense oligonucleotide (ASO) targeting the aberrant C9ORF72 transcript, CSF poly(GA) and poly(GR) levels decreased approximately 50% within 6 weeks, indicating they may serve as sensitive fluid-based biomarkers in studies directed against the production of GGGGCC repeat RNAs or DPR proteins.
Collapse
Affiliation(s)
- Gopinath Krishnan
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | | | - Daniel Bartlett
- Biomarkers, Clinical Sciences Biogen, Cambridge, MA, 02142, USA
| | | | - Diane M McKenna-Yasek
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Catherine Douthwright
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | | | - Shafeeq Ladha
- Departments of Neurology and Translational Neuroscience, St. Joseph's Hospital and Medical Center and Barrow Neurological Institute, 350W Thomas Road, Phoenix, AZ, 85013, USA
| | - Oliver D King
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Sami J Barmada
- Department of Neurology, University of Michigan, 4005 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Timothy M Miller
- Department of Neurology, Washington University, Saint Louis, MI, 63110, USA
| | - Robert Bowser
- Departments of Neurology and Translational Neuroscience, St. Joseph's Hospital and Medical Center and Barrow Neurological Institute, 350W Thomas Road, Phoenix, AZ, 85013, USA
| | - Jonathan K Watts
- RNA Therapeutics Institute and Department of Biochemistry and Molecular Pharmacology, UMass Chan Medical School, Worcester, MA, 01605, USA
| | | | - Robert H Brown
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Mark W Kankel
- Neuromuscular & Movement Disorders, Biogen, Cambridge, MA, 02142, USA.
- Apple Tree Partners (ATP) Research Labs, Branford, CT, 06405, USA.
| | - Fen-Biao Gao
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
18
|
How can we define the presymptomatic C9orf72 disease in 2022? An overview on the current definitions of preclinical and prodromal phases. Rev Neurol (Paris) 2022; 178:426-436. [PMID: 35525633 DOI: 10.1016/j.neurol.2022.03.007] [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: 01/20/2022] [Accepted: 03/08/2022] [Indexed: 11/24/2022]
Abstract
Repeat expansions in C9orf72 gene are the main genetic cause of frontotemporal dementia, amyotrophic lateral sclerosis and related phenotypes. With the advent of disease-modifying treatments, the presymptomatic disease phase is getting increasing interest as an ideal time window in which innovant therapeutic approaches could be administered. Recommendations issued from international study groups distinguish between a preclinical disease stage, during which lesions accumulate in absence of any symptoms or signs, and a prodromal stage, marked by the appearance the first subtle cognitive, behavioral, psychiatric and motor signs, before the full-blown disease. This paper summarizes the current definitions and criteria for these stages, in particular focusing on how fluid-based, neuroimaging and cognitive biomarkers can be useful to monitor disease trajectory across the presymptomatic phase, as well as to detect the earliest signs of clinical conversion. Continuous advances in the knowledge of C9orf72 pathophysiology, and the integration of biomarkers in the clinical evaluation of mutation carriers will allow a better diagnostic definition of C9orf72 disease spectrum from the earliest stages, with relevant impact on the possibility of disease prevention.
Collapse
|
19
|
Dreger M, Steinbach R, Otto M, Turner MR, Grosskreutz J. Cerebrospinal fluid biomarkers of disease activity and progression in amyotrophic lateral sclerosis. J Neurol Neurosurg Psychiatry 2022; 93:422-435. [PMID: 35105727 PMCID: PMC8921583 DOI: 10.1136/jnnp-2021-327503] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 11/29/2021] [Indexed: 12/04/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a relentlessly progressive neurodegenerative disease, and only modest disease-modifying strategies have been established to date. Numerous clinical trials have been conducted in the past years, but have been severely hampered by the wide-ranging heterogeneity of both the biological origins and clinical characteristics of the disease. Thus, reliable biomarkers of disease activity are urgently needed to stratify patients into homogenous groups with aligned disease trajectories to allow a more effective design of clinical trial. In this review, the most promising candidate biomarkers in the cerebrospinal fluid (CSF) of patients with ALS will be summarised. Correlations between biomarker levels and clinical outcome parameters are discussed, while highlighting potential pitfalls and intercorrelations of these clinical parameters. Several CSF molecules have shown potential as biomarkers of progression and prognosis, but large, international, multicentric and longitudinal studies are crucial for validation. A more standardised choice of clinical endpoints in these studies, as well as the application of individualised models of clinical progression, would allow the quantification of disease trajectories, thereby allowing a more accurate analysis of the clinical implications of candidate biomarkers. Additionally, a comparative analysis of several biomarkers and ideally the application of a multivariate analysis including comprehensive genotypic, phenotypic and clinical characteristics collectively contributing to biomarker levels in the CSF, could promote their verification. Thus, reliable prognostic markers and markers of disease activity may improve clinical trial design and patient management in the direction of precision medicine.
Collapse
Affiliation(s)
- Marie Dreger
- Department of Neurology, Jena University Hospital, Jena, Thüringen, Germany
| | - Robert Steinbach
- Department of Neurology, Jena University Hospital, Jena, Thüringen, Germany
| | - Markus Otto
- Department of Neurology, University of Halle (Saale), Halle (Saale), Germany
| | - Martin R Turner
- Nuffield Department of Clinical Neurosciences, Oxford University, Oxford, Oxfordshire, UK
| | - Julian Grosskreutz
- Precision Neurology, Department of Neurology, University of Luebeck Human Medicine, Luebeck, Schleswig-Holstein, Germany
| |
Collapse
|
20
|
Katzeff JS, Bright F, Phan K, Kril JJ, Ittner LM, Kassiou M, Hodges JR, Piguet O, Kiernan MC, Halliday GM, Kim WS. Biomarker discovery and development for frontotemporal dementia and amyotrophic lateral sclerosis. Brain 2022; 145:1598-1609. [PMID: 35202463 PMCID: PMC9166557 DOI: 10.1093/brain/awac077] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 02/08/2022] [Accepted: 02/13/2022] [Indexed: 11/12/2022] Open
Abstract
Frontotemporal dementia refers to a group of neurodegenerative disorders characterized by behaviour and language alterations and focal brain atrophy. Amyotrophic lateral sclerosis is a rapidly progressing neurodegenerative disease characterized by loss of motor neurons resulting in muscle wasting and paralysis. Frontotemporal dementia and amyotrophic lateral sclerosis are considered to exist on a disease spectrum given substantial overlap of genetic and molecular signatures. The predominant genetic abnormality in both frontotemporal dementia and amyotrophic lateral sclerosis is an expanded hexanucleotide repeat sequence in the C9orf72 gene. In terms of brain pathology, abnormal aggregates of TAR-DNA-binding protein-43 are predominantly present in frontotemporal dementia and amyotrophic lateral sclerosis patients. Currently, sensitive and specific diagnostic and disease surveillance biomarkers are lacking for both diseases. This has impeded the capacity to monitor disease progression during life and the development of targeted drug therapies for the two diseases. The purpose of this review is to examine the status of current biofluid biomarker discovery and development in frontotemporal dementia and amyotrophic lateral sclerosis. The major pathogenic proteins implicated in different frontotemporal dementia and amyotrophic lateral sclerosis molecular subtypes and proteins associated with neurodegeneration and the immune system will be discussed. Furthermore, the use of mass spectrometry-based proteomics as an emerging tool to identify new biomarkers in frontotemporal dementia and amyotrophic lateral sclerosis will be summarized.
Collapse
Affiliation(s)
- Jared S Katzeff
- The University of Sydney, Brain and Mind Centre, Sydney, NSW, Australia.,The University of Sydney, School of Medical Sciences, Sydney, NSW, Australia
| | - Fiona Bright
- The University of Sydney, School of Medical Sciences, Sydney, NSW, Australia.,Dementia Research Centre and Department of Biomedical Sciences, Macquarie University, Sydney, NSW, Australia
| | - Katherine Phan
- The University of Sydney, Brain and Mind Centre, Sydney, NSW, Australia.,The University of Sydney, School of Medical Sciences, Sydney, NSW, Australia
| | - Jillian J Kril
- The University of Sydney, School of Medical Sciences, Sydney, NSW, Australia.,Dementia Research Centre and Department of Biomedical Sciences, Macquarie University, Sydney, NSW, Australia
| | - Lars M Ittner
- Dementia Research Centre and Department of Biomedical Sciences, Macquarie University, Sydney, NSW, Australia
| | - Michael Kassiou
- The University of Sydney, School of Chemistry, Sydney, NSW, Australia
| | - John R Hodges
- The University of Sydney, Brain and Mind Centre, Sydney, NSW, Australia
| | - Olivier Piguet
- The University of Sydney, Brain and Mind Centre, Sydney, NSW, Australia.,The University of Sydney, School of Psychology, Sydney, NSW, Australia
| | - Matthew C Kiernan
- The University of Sydney, Brain and Mind Centre, Sydney, NSW, Australia.,Institute of Clinical Neurosciences, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Glenda M Halliday
- The University of Sydney, Brain and Mind Centre, Sydney, NSW, Australia.,The University of Sydney, School of Medical Sciences, Sydney, NSW, Australia
| | - Woojin Scott Kim
- The University of Sydney, Brain and Mind Centre, Sydney, NSW, Australia.,The University of Sydney, School of Medical Sciences, Sydney, NSW, Australia
| |
Collapse
|
21
|
Kandasamy P, Liu Y, Aduda V, Akare S, Alam R, Andreucci A, Boulay D, Bowman K, Byrne M, Cannon M, Chivatakarn O, Shelke JD, Iwamoto N, Kawamoto T, Kumarasamy J, Lamore S, Lemaitre M, Lin X, Longo K, Looby R, Marappan S, Metterville J, Mohapatra S, Newman B, Paik IH, Patil S, Purcell-Estabrook E, Shimizu M, Shum P, Standley S, Taborn K, Tripathi S, Yang H, Yin Y, Zhao X, Dale E, Vargeese C. Impact of guanidine-containing backbone linkages on stereopure antisense oligonucleotides in the CNS. Nucleic Acids Res 2022; 50:5401-5423. [PMID: 35106589 PMCID: PMC9177980 DOI: 10.1093/nar/gkac037] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 12/17/2021] [Accepted: 01/13/2022] [Indexed: 12/02/2022] Open
Abstract
Attaining sufficient tissue exposure at the site of action to achieve the desired pharmacodynamic effect on a target is an important determinant for any drug discovery program, and this can be particularly challenging for oligonucleotides in deep tissues of the CNS. Herein, we report the synthesis and impact of stereopure phosphoryl guanidine-containing backbone linkages (PN linkages) to oligonucleotides acting through an RNase H-mediated mechanism, using Malat1 and C9orf72 as benchmarks. We found that the incorporation of various types of PN linkages to a stereopure oligonucleotide backbone can increase potency of silencing in cultured neurons under free-uptake conditions 10-fold compared with similarly modified stereopure phosphorothioate (PS) and phosphodiester (PO)-based molecules. One of these backbone types, called PN-1, also yielded profound silencing benefits throughout the mouse brain and spinal cord at low doses, improving both the potency and durability of response, especially in difficult to reach brain tissues. Given these benefits in preclinical models, the incorporation of PN linkages into stereopure oligonucleotides with chimeric backbone modifications has the potential to render regions of the brain beyond the spinal cord more accessible to oligonucleotides and, consequently, may also expand the scope of neurological indications amenable to oligonucleotide therapeutics.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Xuena Lin
- Wave Life Sciences, Cambridge, MA 02138, USA
| | | | | | | | | | | | | | | | | | | | | | - Pochi Shum
- Wave Life Sciences, Cambridge, MA 02138, USA
| | | | - Kris Taborn
- Wave Life Sciences, Cambridge, MA 02138, USA
| | | | - Hailin Yang
- Wave Life Sciences, Cambridge, MA 02138, USA
| | - Yuan Yin
- Wave Life Sciences, Cambridge, MA 02138, USA
| | - Xiansi Zhao
- Wave Life Sciences, Cambridge, MA 02138, USA
| | - Elena Dale
- Wave Life Sciences, Cambridge, MA 02138, USA
| | | |
Collapse
|
22
|
Reyes-Leiva D, Dols-Icardo O, Sirisi S, Cortés-Vicente E, Turon-Sans J, de Luna N, Blesa R, Belbin O, Montal V, Alcolea D, Fortea J, Lleó A, Rojas-García R, Illán-Gala I. Pathophysiological Underpinnings of Extra-Motor Neurodegeneration in Amyotrophic Lateral Sclerosis: New Insights From Biomarker Studies. Front Neurol 2022; 12:750543. [PMID: 35115992 PMCID: PMC8804092 DOI: 10.3389/fneur.2021.750543] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 12/09/2021] [Indexed: 11/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) lie at opposing ends of a clinical, genetic, and neuropathological continuum. In the last decade, it has become clear that cognitive and behavioral changes in patients with ALS are more frequent than previously recognized. Significantly, these non-motor features can impact the diagnosis, prognosis, and management of ALS. Partially overlapping neuropathological staging systems have been proposed to describe the distribution of TAR DNA-binding protein 43 (TDP-43) aggregates outside the corticospinal tract. However, the relationship between TDP-43 inclusions and neurodegeneration is not absolute and other pathophysiological processes, such as neuroinflammation (with a prominent role of microglia), cortical hyperexcitability, and synaptic dysfunction also play a central role in ALS pathophysiology. In the last decade, imaging and biofluid biomarker studies have revealed important insights into the pathophysiological underpinnings of extra-motor neurodegeneration in the ALS-FTLD continuum. In this review, we first summarize the clinical and pathophysiological correlates of extra-motor neurodegeneration in ALS. Next, we discuss the diagnostic and prognostic value of biomarkers in ALS and their potential to characterize extra-motor neurodegeneration. Finally, we debate about how biomarkers could improve the diagnosis and classification of ALS. Emerging imaging biomarkers of extra-motor neurodegeneration that enable the monitoring of disease progression are particularly promising. In addition, a growing arsenal of biofluid biomarkers linked to neurodegeneration and neuroinflammation are improving the diagnostic accuracy and identification of patients with a faster progression rate. The development and validation of biomarkers that detect the pathological aggregates of TDP-43 in vivo are notably expected to further elucidate the pathophysiological underpinnings of extra-motor neurodegeneration in ALS. Novel biomarkers tracking the different aspects of ALS pathophysiology are paving the way to precision medicine approaches in the ALS-FTLD continuum. These are essential steps to improve the diagnosis and staging of ALS and the design of clinical trials testing novel disease-modifying treatments.
Collapse
Affiliation(s)
- David Reyes-Leiva
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Oriol Dols-Icardo
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Sonia Sirisi
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Elena Cortés-Vicente
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Janina Turon-Sans
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Noemi de Luna
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Rafael Blesa
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Olivia Belbin
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Victor Montal
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Daniel Alcolea
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Alberto Lleó
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Ricard Rojas-García
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Ignacio Illán-Gala
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
- *Correspondence: Ignacio Illán-Gala
| |
Collapse
|
23
|
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: 28] [Impact Index Per Article: 7.0] [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
|
24
|
Hübener-Schmid J, Kuhlbrodt K, Peladan J, Faber J, Santana MM, Hengel H, Jacobi H, Reetz K, Garcia-Moreno H, Raposo M, van Gaalen J, Infante J, Steiner KM, de Vries J, Verbeek MM, Giunti P, Pereira de Almeida L, Lima M, van de Warrenburg B, Schöls L, Klockgether T, Synofzik M, Riess O. Polyglutamine-Expanded Ataxin-3: A Target Engagement Marker for Spinocerebellar Ataxia Type 3 in Peripheral Blood. Mov Disord 2021; 36:2675-2681. [PMID: 34397117 DOI: 10.1002/mds.28749] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/27/2021] [Accepted: 07/21/2021] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Spinocerebellar ataxia type 3 is a rare neurodegenerative disease caused by a CAG repeat expansion in the ataxin-3 gene. Although no curative therapy is yet available, preclinical gene-silencing approaches to reduce polyglutamine (polyQ) toxicity demonstrate promising results. In view of upcoming clinical trials, quantitative and easily accessible molecular markers are of critical importance as pharmacodynamic and particularly as target engagement markers. OBJECTIVE We aimed at developing an ultrasensitive immunoassay to measure specifically polyQ-expanded ataxin-3 in plasma and cerebrospinal fluid (CSF). METHODS Using the novel single molecule counting ataxin-3 immunoassay, we analyzed cross-sectional and longitudinal patient biomaterials. RESULTS Statistical analyses revealed a correlation with clinical parameters and a stability of polyQ-expanded ataxin-3 during conversion from the pre-ataxic to the ataxic phases. CONCLUSIONS The novel immunoassay is able to quantify polyQ-expanded ataxin-3 in plasma and CSF, whereas ataxin-3 levels in plasma correlate with disease severity. Longitudinal analyses demonstrated a high stability of polyQ-expanded ataxin-3 over a short period. © 2021 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Jeannette Hübener-Schmid
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany.,Centre for Rare Diseases, University of Tübingen, Tübingen, Germany
| | | | | | - Jennifer Faber
- DZNE, German Center for Neurodegenerative Diseases, Bonn, Germany.,Department of Neurology, University Hospital Bonn, Bonn, Germany
| | - Magda M Santana
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Center for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal.,IIIUC-Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Holger Hengel
- Department of Neurodegenerative Diseases and Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,German Centre for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Heike Jacobi
- Department of Neurology, University Hospital of Heidelberg, Heidelberg, Germany
| | - Kathrin Reetz
- Department of Neurology, RWTH Aachen University, Aachen, Germany.,JARA-Brain Institute Molecular Neuroscience and Neuroimaging, Jülich, Germany
| | - Hector Garcia-Moreno
- Department of Clinical and Movement Neuroscience, Ataxia Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.,Department of Neurogenetics, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - Mafalda Raposo
- Faculdade de Ciências e Tecnologia, Universidade dos Açores, Ponta Delgada, Portugal & Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Judith van Gaalen
- Department of Neurology, Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Jon Infante
- Service of Neurology, University Hospital Marqués de Valdecilla (IDIVAL), University of Cantabria (UC), Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Santander, Spain
| | - Katharina M Steiner
- Department of Neurology, Center for Translational Neuro- and Behavioral Sciences, Essen University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Jeroen de Vries
- Department of Neurology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Marcel M Verbeek
- Department of Neurology, Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands.,Department of Laboratory Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Paola Giunti
- Department of Clinical and Movement Neuroscience, Ataxia Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.,Department of Neurogenetics, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - Luis Pereira de Almeida
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Center for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal.,Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Manuela Lima
- Faculdade de Ciências e Tecnologia, Universidade dos Açores, Ponta Delgada, Portugal & Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Bart van de Warrenburg
- Department of Neurology, Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Ludger Schöls
- Department of Neurodegenerative Diseases and Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,German Centre for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Thomas Klockgether
- DZNE, German Center for Neurodegenerative Diseases, Bonn, Germany.,Department of Neurology, University Hospital Bonn, Bonn, Germany
| | - Matthis Synofzik
- Department of Neurodegenerative Diseases and Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,German Centre for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | | | - Olaf Riess
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany.,Centre for Rare Diseases, University of Tübingen, Tübingen, Germany
| |
Collapse
|
25
|
Cook CN, Wu Y, Odeh HM, Gendron TF, Jansen-West K, Del Rosso G, Yue M, Jiang P, Gomes E, Tong J, Daughrity LM, Avendano NM, Castanedes-Casey M, Shao W, Oskarsson B, Tomassy GS, McCampbell A, Rigo F, Dickson DW, Shorter J, Zhang YJ, Petrucelli L. C9orf72 poly(GR) aggregation induces TDP-43 proteinopathy. Sci Transl Med 2021; 12:12/559/eabb3774. [PMID: 32878979 DOI: 10.1126/scitranslmed.abb3774] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 06/19/2020] [Indexed: 12/14/2022]
Abstract
TAR DNA-binding protein 43 (TDP-43) inclusions are a pathological hallmark of frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS), including cases caused by G4C2 repeat expansions in the C9orf72 gene (c9FTD/ALS). Providing mechanistic insight into the link between C9orf72 mutations and TDP-43 pathology, we demonstrated that a glycine-arginine repeat protein [poly(GR)] translated from expanded G4C2 repeats was sufficient to promote aggregation of endogenous TDP-43. In particular, toxic poly(GR) proteins mediated sequestration of full-length TDP-43 in an RNA-independent manner to induce cytoplasmic TDP-43 inclusion formation. Moreover, in GFP-(GR)200 mice, poly(GR) caused the mislocalization of nucleocytoplasmic transport factors and nuclear pore complex proteins. These mislocalization events resulted in the aberrant accumulation of endogenous TDP-43 in the cytoplasm where it co-aggregated with poly(GR). Last, we demonstrated that treating G4C2 repeat-expressing mice with repeat-targeting antisense oligonucleotides lowered poly(GR) burden, which was accompanied by reduced TDP-43 pathology and neurodegeneration, including lowering of plasma neurofilament light (NFL) concentration. These results contribute to clarification of the mechanism by which poly(GR) drives TDP-43 proteinopathy, confirm that G4C2-targeted therapeutics reduce TDP-43 pathology in vivo, and demonstrate that alterations in plasma NFL provide insight into the therapeutic efficacy of disease-modifying treatments.
Collapse
Affiliation(s)
- Casey N Cook
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA.,Neurobiology of Disease Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Rochester, MN 55902, USA
| | - Yanwei Wu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Hana M Odeh
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tania F Gendron
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA.,Neurobiology of Disease Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Rochester, MN 55902, USA
| | - Karen Jansen-West
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Giulia Del Rosso
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Mei Yue
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Peizhou Jiang
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Edward Gomes
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jimei Tong
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Nicole M Avendano
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Wei Shao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Björn Oskarsson
- Department of Neurology, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | | | - Frank Rigo
- Ionis Pharmaceuticals, Carlsbad, CA 92010, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA.,Neurobiology of Disease Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Rochester, MN 55902, USA
| | - James Shorter
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Yong-Jie Zhang
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA. .,Neurobiology of Disease Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Rochester, MN 55902, USA
| | - Leonard Petrucelli
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA. .,Neurobiology of Disease Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Rochester, MN 55902, USA
| |
Collapse
|
26
|
Verde F, Otto M, Silani V. Neurofilament Light Chain as Biomarker for Amyotrophic Lateral Sclerosis and Frontotemporal Dementia. Front Neurosci 2021; 15:679199. [PMID: 34234641 PMCID: PMC8255624 DOI: 10.3389/fnins.2021.679199] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 04/29/2021] [Indexed: 11/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are two related currently incurable neurodegenerative diseases. ALS is characterized by degeneration of upper and lower motor neurons causing relentless paralysis of voluntary muscles, whereas in FTD, progressive atrophy of the frontal and temporal lobes of the brain results in deterioration of cognitive functions, language, personality, and behavior. In contrast to Alzheimer's disease (AD), ALS and FTD still lack a specific neurochemical biomarker reflecting neuropathology ex vivo. However, in the past 10 years, considerable progress has been made in the characterization of neurofilament light chain (NFL) as cerebrospinal fluid (CSF) and blood biomarker for both diseases. NFL is a structural component of the axonal cytoskeleton and is released into the CSF as a consequence of axonal damage or degeneration, thus behaving in general as a relatively non-specific marker of neuroaxonal pathology. However, in ALS, the elevation of its CSF levels exceeds that observed in most other neurological diseases, making it useful for the discrimination from mimic conditions and potentially worthy of consideration for introduction into diagnostic criteria. Moreover, NFL correlates with disease progression rate and is negatively associated with survival, thus providing prognostic information. In FTD patients, CSF NFL is elevated compared with healthy individuals and, to a lesser extent, patients with other forms of dementia, but the latter difference is not sufficient to enable a satisfying diagnostic performance at individual patient level. However, also in FTD, CSF NFL correlates with several measures of disease severity. Due to technological progress, NFL can now be quantified also in peripheral blood, where it is present at much lower concentrations compared with CSF, thus allowing less invasive sampling, scalability, and longitudinal measurements. The latter has promoted innovative studies demonstrating longitudinal kinetics of NFL in presymptomatic individuals harboring gene mutations causing ALS and FTD. Especially in ALS, NFL levels are generally stable over time, which, together with their correlation with progression rate, makes NFL an ideal pharmacodynamic biomarker for therapeutic trials. In this review, we illustrate the significance of NFL as biomarker for ALS and FTD and discuss unsolved issues and potential for future developments.
Collapse
Affiliation(s)
- Federico Verde
- Department of Neurology-Stroke Unit and Laboratory of Neuroscience, Istituto Auxologico Italiano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy.,Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy
| | - Markus Otto
- Department of Neurology, Ulm University Hospital, Ulm, Germany
| | - Vincenzo Silani
- Department of Neurology-Stroke Unit and Laboratory of Neuroscience, Istituto Auxologico Italiano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy.,Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
27
|
Figgie MP, Appleby BS. Clinical Use of Improved Diagnostic Testing for Detection of Prion Disease. Viruses 2021; 13:v13050789. [PMID: 33925126 PMCID: PMC8146465 DOI: 10.3390/v13050789] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/21/2021] [Accepted: 04/23/2021] [Indexed: 12/12/2022] Open
Abstract
Prion diseases are difficult to recognize as many symptoms are shared among other neurologic pathologies and the full spectra of symptoms usually do not appear until late in the disease course. Additionally, many commonly used laboratory markers are non-specific to prion disease. The recent introduction of second-generation real time quaking induced conversion (RT-QuIC) has revolutionized pre-mortem diagnosis of prion disease due to its extremely high sensitivity and specificity. However, RT-QuIC does not provide prognostic data and has decreased diagnostic accuracy in some rarer, atypical prion diseases. The objective of this review is to provide an overview of the current clinical utility of fluid-based biomarkers, neurodiagnostic testing, and brain imaging in the diagnosis of prion disease and to suggest guidelines for their clinical use, with a focus on rarer prion diseases with atypical features. Recent advancements in laboratory-based testing and imaging criteria have shown improved diagnostic accuracy and prognostic potential in prion disease, but because these diagnostic tests are not sensitive in some prion disease subtypes and diagnostic test sensitivities are unknown in the event that CWD transmits to humans, it is important to continue investigations into the clinical utility of various testing modalities.
Collapse
Affiliation(s)
- Mark P. Figgie
- Department of Neurology, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, OH 44106, USA;
| | - Brian S. Appleby
- Department of Neurology, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, OH 44106, USA;
- National Prion Disease Pathology Surveillance Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Correspondence:
| |
Collapse
|
28
|
De Vocht J, Blommaert J, Devrome M, Radwan A, Van Weehaeghe D, De Schaepdryver M, Ceccarini J, Rezaei A, Schramm G, van Aalst J, Chiò A, Pagani M, Stam D, Van Esch H, Lamaire N, Verhaegen M, Mertens N, Poesen K, van den Berg LH, van Es MA, Vandenberghe R, Vandenbulcke M, Van den Stock J, Koole M, Dupont P, Van Laere K, Van Damme P. Use of Multimodal Imaging and Clinical Biomarkers in Presymptomatic Carriers of C9orf72 Repeat Expansion. JAMA Neurol 2021; 77:1008-1017. [PMID: 32421156 PMCID: PMC7417970 DOI: 10.1001/jamaneurol.2020.1087] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Question Can metabolic brain changes be detected in presymptomatic individuals who are carriers of a hexanucleotide repeat expansion in the C9orf72 gene (preSxC9) using time-of-flight fluorine 18–labeled fluorodeoxyglucose positron emission tomographic imaging and magnetic resonance imaging, and what is the association between the mutation and clinical and fluid biomarkers of amyotrophic lateral sclerosis and frontotemporal dementia? Findings In a case-control study including 17 preSxC9 participants and 25 healthy controls, fluorine 18–labeled fluorodeoxyglucose positron emission tomographic imaging noted significant clusters of relative hypometabolism in frontotemporal regions, the insular cortices, basal ganglia, and thalami in the preSxC9 participants. Use of this strategy allowed detection of changes at an individual level. Meaning Glucose metabolic changes appear to occur early in the sequence of events leading to manifest amyotrophic lateral sclerosis and frontotemporal dementia. Fluorine 18–labeled fluorodeoxyglucose positron emission tomographic imaging may provide a sensitive biomarker of a presymptomatic phase of disease. Importance During a time with the potential for novel treatment strategies, early detection of disease manifestations at an individual level in presymptomatic carriers of a hexanucleotide repeat expansion in the C9orf72 gene (preSxC9) is becoming increasingly relevant. Objectives To evaluate changes in glucose metabolism before symptom onset of amyotrophic lateral sclerosis or frontotemporal dementia in preSxC9 using simultaneous fluorine 18–labeled fluorodeoxyglucose ([18F]FDG positron emission tomographic (PET) and magnetic resonance imaging as well as the mutation’s association with clinical and fluid biomarkers. Design, Setting, and Participants A prospective, case-control study enrolled 46 participants from November 30, 2015, until December 11, 2018. The study was conducted at the neuromuscular reference center of the University Hospitals Leuven, Leuven, Belgium. Main Outcomes and Measures Neuroimaging data were spatially normalized and analyzed at the voxel level at a height threshold of P < .001, cluster-level familywise error–corrected threshold of P < .05, and statistical significance was set at P < .05 for the volume-of-interest level analysis, using Benjamini-Hochberg correction for multiple correction. W-score maps were computed using the individuals serving as controls as a reference to quantify the degree of [18F]FDG PET abnormality. The threshold for abnormality on the W-score maps was designated as an absolute W-score greater than or equal to 1.96. Neurofilament levels and performance on cognitive and neurologic examinations were determined. All hypothesis tests were 1-sided. Results Of the 42 included participants, there were 17 with the preSxC9 mutation (12 women [71%]; mean [SD] age, 51 [9] years) and 25 healthy controls (12 women [48%]; mean [SD] age, 47 [10] years). Compared with control participants, significant clusters of relative hypometabolism were found in frontotemporal regions, basal ganglia, and thalami of preSxC9 participants and relative hypermetabolism in the peri-Rolandic region, superior frontal gyrus, and precuneus cortex. W-score frequency maps revealed reduced glucose metabolism with local maxima in the insular cortices, central opercular cortex, and thalami in up to 82% of preSxC9 participants and increased glucose metabolism in the precentral gyrus and precuneus cortex in up to 71% of preSxC9 participants. Other findings in the preSxC9 group were upper motor neuron involvement in 10 participants (59%), cognitive abnormalities in 5 participants (29%), and elevated neurofilament levels in 3 of 16 individuals (19%) who underwent lumbar puncture. Conclusions and Relevance The results suggest that [18F]FDG PET can identify glucose metabolic changes in preSxC9 at an individual level, preceding significantly elevated neurofilament levels and onset of symptoms.
Collapse
Affiliation(s)
- Joke De Vocht
- KU Leuven, Department of Neurosciences, Experimental Neurology, B-3000 Leuven, Belgium.,KU Leuven, University Hospitals Leuven, University Psychiatric Center, Adult Psychiatry, B-3000 Leuven, Belgium.,University Hospitals Leuven, Department of Neurology, B-3000 Leuven, Belgium.,VIB - Center of Brain & Disease Research, Laboratory of Neurobiology, B-3000 Leuven, Belgium
| | | | - Martijn Devrome
- KU Leuven, University Hospitals Leuven, Department of Imaging and Pathology, Division of Nuclear Medicine, B-3000 Leuven, Belgium
| | - Ahmed Radwan
- KU Leuven, Department of Imaging and Pathology, Translational MRI, B-3000 Leuven, Belgium
| | - Donatienne Van Weehaeghe
- KU Leuven, University Hospitals Leuven, Department of Imaging and Pathology, Division of Nuclear Medicine, B-3000 Leuven, Belgium
| | - Maxim De Schaepdryver
- KU Leuven, Department of Neurosciences, Laboratory for Molecular Neurobiomarker Research, B-3000 Leuven, Belgium
| | - Jenny Ceccarini
- KU Leuven, University Hospitals Leuven, Department of Imaging and Pathology, Division of Nuclear Medicine, B-3000 Leuven, Belgium
| | - Ahmadreza Rezaei
- KU Leuven, University Hospitals Leuven, Department of Imaging and Pathology, Division of Nuclear Medicine, B-3000 Leuven, Belgium
| | - Georg Schramm
- KU Leuven, University Hospitals Leuven, Department of Imaging and Pathology, Division of Nuclear Medicine, B-3000 Leuven, Belgium
| | - June van Aalst
- KU Leuven, University Hospitals Leuven, Department of Imaging and Pathology, Division of Nuclear Medicine, B-3000 Leuven, Belgium
| | - Adriano Chiò
- ALS Center, Rita Levi Montalcini Department of Neuroscience, University of Turin, Turin, Italy
| | - Marco Pagani
- Institute of Cognitive Sciences and Technologies, CNR, Rome, Italy.,Medical Radiation Physics and Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Daphne Stam
- KU Leuven, Leuven Brain Institute, Laboratory for Translational Neuropsychiatry, B-3000 Leuven, Belgium
| | - Hilde Van Esch
- University Hospitals Leuven, Center for Human Genetics, B-3000 Leuven, Belgium
| | - Nikita Lamaire
- University Hospitals Leuven, Department of Neurology, B-3000 Leuven, Belgium
| | - Marianne Verhaegen
- KU Leuven, University Hospitals Leuven, University Psychiatric Center, Adult Psychiatry, B-3000 Leuven, Belgium
| | - Nathalie Mertens
- KU Leuven, University Hospitals Leuven, Department of Imaging and Pathology, Division of Nuclear Medicine, B-3000 Leuven, Belgium
| | - Koen Poesen
- KU Leuven, Department of Neurosciences, Laboratory for Molecular Neurobiomarker Research, B-3000 Leuven, Belgium
| | - Leonard H van den Berg
- Brain Center Rudolf Magnus, Department of Neurology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Michael A van Es
- Brain Center Rudolf Magnus, Department of Neurology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Rik Vandenberghe
- University Hospitals Leuven, Department of Neurology, B-3000 Leuven, Belgium.,KU Leuven, Department of Neurosciences, Laboratory for Cognitive Neurology, B-3000 Leuven, Belgium
| | - Mathieu Vandenbulcke
- KU Leuven, Leuven Brain Institute, Laboratory for Translational Neuropsychiatry, B-3000 Leuven, Belgium.,KU Leuven, University Psychiatric Center, Geriatric Psychiatry, B-3000 Leuven, Belgium
| | - Jan Van den Stock
- KU Leuven, Leuven Brain Institute, Laboratory for Translational Neuropsychiatry, B-3000 Leuven, Belgium.,KU Leuven, University Psychiatric Center, Geriatric Psychiatry, B-3000 Leuven, Belgium
| | - Michel Koole
- KU Leuven, University Hospitals Leuven, Department of Imaging and Pathology, Division of Nuclear Medicine, B-3000 Leuven, Belgium
| | - Patrick Dupont
- KU Leuven, Department of Neurosciences, Laboratory for Cognitive Neurology, B-3000 Leuven, Belgium
| | - Koen Van Laere
- KU Leuven, University Hospitals Leuven, Department of Imaging and Pathology, Division of Nuclear Medicine, B-3000 Leuven, Belgium
| | - Philip Van Damme
- KU Leuven, Department of Neurosciences, Experimental Neurology, B-3000 Leuven, Belgium.,University Hospitals Leuven, Department of Neurology, B-3000 Leuven, Belgium.,VIB - Center of Brain & Disease Research, Laboratory of Neurobiology, B-3000 Leuven, Belgium
| |
Collapse
|
29
|
Karantali E, Kazis D, Chatzikonstantinou S, Petridis F, Mavroudis I. The role of neurofilament light chain in frontotemporal dementia: a meta-analysis. Aging Clin Exp Res 2021; 33:869-881. [PMID: 32306372 DOI: 10.1007/s40520-020-01554-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 04/02/2020] [Indexed: 12/13/2022]
Abstract
Frontotemporal dementia (FTD) is the second most frequent dementia, after Alzheimer's, in patients under the age of 65. It encompasses clinical entities characterized by behavioral, language, and executive control dysfunction. Neurofilament light chain (NfL) is a new, non-disease specific, widely studied biomarker indicative of axonal injury and degeneration. Various studies have previously explored the role of NfL in the diagnostic process, monitoring, and prognosis of dementia. The current systematic review and meta-analysis include all the available data concerning the role of NfL in frontotemporal dementia and its use as a potential biomarker in differentiating patients with FTD from (a) healthy individuals, (b) Alzheimer's dementia, (c) Dementia with Lewy bodies, (d) Motor Neuron disease, (e) Parkinsonian syndromes, and (f) psychiatric disorders. We also analyze the utility of NfL in distinguishing specific FTD subgroups. Neurofilament light chain has a potential role in differentiating patients with frontotemporal dementia from healthy controls, patients with Alzheimer's dementia, and psychiatric disorders. Higher NfL levels were also noted in patients with semantic primary progressive aphasia (PPA) when compared with behavioral FTD and non-fluent PPA patients. Further studies exploring the use of NfL in frontotemporal dementia are needed.
Collapse
Affiliation(s)
- Eleni Karantali
- Third Department of Neurology, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | - Dimitrios Kazis
- Third Department of Neurology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Fivos Petridis
- Third Department of Neurology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioannis Mavroudis
- Leeds Teaching Hospitals, Leeds, UK
- Medical School, Cyprus University, Nicosia, Cyprus
| |
Collapse
|
30
|
Rohrer JD, Boxer AL. The Frontotemporal Dementia Prevention Initiative: Linking Together Genetic Frontotemporal Dementia Cohort Studies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1281:113-121. [PMID: 33433872 DOI: 10.1007/978-3-030-51140-1_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Around one-third of frontotemporal dementia (FTD) is autosomal dominant with the major genetic causes being mutations in MAPT, GRN and C9orf72. Studying familial forms of FTD can provide a window into the earliest stages of the illness, many years before symptoms start. Large cohort studies have been set up in recent years to better understand this presymptomatic phase, including the Genetic FTD Initiative (GENFI) and the Advancing Research and Treatment for Frontotemporal Lobar Degeneration and Longitudinal Evaluation of Familial Frontotemporal Dementia Subjects (ARTFL/LEFFTDS) studies. Whilst these studies have focused on the investigation of a variety of aspects of genetic FTD, from understanding the molecular pathogenesis to developing biomarkers, they also have a common goal: finding a way to prevent FTD. Researchers from these cohort studies have therefore come together to form the FTD Prevention Initiative (FPI), which has the overarching aim of promoting clinical trials of new therapies to prevent FTD through creating an international database of participants eligible for trials and uniform standards for conducting such trials. This chapter outlines the work of the FPI so far and its future goals over the next few years.
Collapse
Affiliation(s)
- Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.
| | - Adam L Boxer
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
31
|
Fluid Biomarkers of Frontotemporal Lobar Degeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1281:123-139. [PMID: 33433873 DOI: 10.1007/978-3-030-51140-1_9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A timely diagnosis of frontotemporal degeneration (FTD) is frequently challenging due to the heterogeneous symptomatology and poor phenotype-pathological correlation. Fluid biomarkers that reflect FTD pathophysiology could be instrumental in both clinical practice and pharmaceutical trials. In recent years, significant progress has been made in developing biomarkers of neurodegenerative diseases: amyloid-β and tau in cerebrospinal fluid (CSF) can be used to exclude Alzheimer's disease, while neurofilament light chain (NfL) is emerging as a promising, albeit nonspecific, marker of neurodegeneration in both CSF and blood. Gene-specific biomarkers such as PGRN in GRN mutation carriers and dipeptide repeat proteins in C9orf72 mutation carriers are potential target engagement markers in genetic FTD trials. Novel techniques capable of measuring very low concentrations of brain-derived proteins in peripheral fluids are facilitating studies of blood biomarkers as a minimally invasive alternative to CSF. A major remaining challenge is the identification of a biomarker that can be used to predict the neuropathological substrate in sporadic FTD patients.
Collapse
|
32
|
Gagliardi D, Costamagna G, Taiana M, Andreoli L, Biella F, Bersani M, Bresolin N, Comi GP, Corti S. Insights into disease mechanisms and potential therapeutics for C9orf72-related amyotrophic lateral sclerosis/frontotemporal dementia. Ageing Res Rev 2020; 64:101172. [PMID: 32971256 DOI: 10.1016/j.arr.2020.101172] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 08/31/2020] [Indexed: 12/12/2022]
Abstract
In 2011, a hexanucleotide repeat expansion (HRE) in the noncoding region of C9orf72 was associated with the most frequent genetic cause of frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS). The main pathogenic mechanisms in C9-ALS/FTD are haploinsufficiency of the C9orf72 protein and gain of function toxicity from bidirectionally-transcribed repeat-containing RNAs and dipeptide repeat proteins (DPRs) resulting from non-canonical RNA translation. Additionally, abnormalities in different downstream cellular mechanisms, such as nucleocytoplasmic transport and autophagy, play a role in pathogenesis. Substantial research efforts using in vitro and in vivo models have provided valuable insights into the contribution of each mechanism in disease pathogenesis. However, conflicting evidence exists, and a unifying theory still lacks. Here, we provide an overview of the recently published literature on clinical, neuropathological and molecular features of C9-ALS/FTD. We highlight the supposed neuronal role of C9orf72 and the HRE pathogenic cascade, mainly focusing on the contribution of RNA foci and DPRs to neurodegeneration and discussing the several downstream mechanisms. We summarize the emerging biochemical and neuroimaging biomarkers, as well as the potential therapeutic approaches. Despite promising results, a specific disease-modifying treatment is still not available to date and greater insights into disease mechanisms may help in this direction.
Collapse
Affiliation(s)
- Delia Gagliardi
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Gianluca Costamagna
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Michela Taiana
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Luca Andreoli
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Fabio Biella
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Margherita Bersani
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Nereo Bresolin
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy; Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milan, Italy
| | - Giacomo Pietro Comi
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy; Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milan, Italy
| | - Stefania Corti
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy; Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milan, Italy.
| |
Collapse
|
33
|
Spotorno N, Lindberg O, Nilsson C, Landqvist Waldö M, van Westen D, Nilsson K, Vestberg S, Englund E, Zetterberg H, Blennow K, Lätt J, Markus N, Lars-Olof W, Alexander S. Plasma neurofilament light protein correlates with diffusion tensor imaging metrics in frontotemporal dementia. PLoS One 2020; 15:e0236384. [PMID: 33108404 PMCID: PMC7591030 DOI: 10.1371/journal.pone.0236384] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 10/12/2020] [Indexed: 12/02/2022] Open
Abstract
Neurofilaments are structural components of neurons and are particularly abundant in highly myelinated axons. The levels of neurofilament light chain (NfL) in both cerebrospinal fluid (CSF) and plasma have been related to degeneration in several neurodegenerative conditions including frontotemporal dementia (FTD) and NfL is currently considered as the most promising diagnostic and prognostic fluid biomarker in FTD. Although the location and function of filaments in the healthy nervous system suggests a link between increased NfL and white matter degeneration, such a claim has not been fully elucidated in vivo, especially in the context of FTD. The present study provides evidence of an association between the plasma levels of NfL and white matter involvement in behavioral variant FTD (bvFTD) by relating plasma concentration of NfL to diffusion tensor imaging (DTI) metrics in a group of 20 bvFTD patients. The results of both voxel-wise and tract specific analysis showed that increased plasma NfL concentration is associated with a reduction in fractional anisotropy (FA) in a widespread set of white matter tracts including the superior longitudinal fasciculus, the fronto-occipital fasciculus the anterior thalamic radiation and the dorsal cingulum bundle. Plasma NfL concentration also correlated with cortical thinning in a portion of the right medial prefrontal cortex and of the right lateral orbitofrontal cortex. These results support the hypothesis that blood NfL levels reflect the global level of neurodegeneration in bvFTD and help to advance our understanding of the association between this blood biomarker for FTD and the disease process.
Collapse
Affiliation(s)
- Nicola Spotorno
- Department of Neurology, Penn Frontotemporal Degeneration Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States of America
- Department of Clinical Sciences, Clinical Memory Research Unit, Lund University, Malmö, Sweden
| | - Olof Lindberg
- Division of Clinical Geriatrics, Karolinska Institute, Stockholm, Sweden
| | - Christer Nilsson
- Division of Neurology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Maria Landqvist Waldö
- Department of clinical Sciences, Clinical Sciences Helsingborg, Lund, Lund University, Lund, Sweden
| | - Danielle van Westen
- Department of Diagnostic Radiology, Clinical Sciences, Lund University, Lund, Sweden
| | - Karin Nilsson
- Department of Clinical Sciences, Clinical Memory Research Unit, Lund University, Malmö, Sweden
| | | | - Elisabet Englund
- Division of Pathology, Department of Clinical Sciences, Lund, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Jimmy Lätt
- Center for Medical Imaging and Physiology, Skåne University Hospital, Lund, Sweden
| | - Nilsson Markus
- Department of Diagnostic Radiology, Clinical Sciences, Lund University, Lund, Sweden
| | - Wahlund Lars-Olof
- Division of Clinical Geriatrics, Karolinska Institute, Stockholm, Sweden
| | - Santillo Alexander
- Department of Clinical Sciences, Clinical Memory Research Unit, Lund University, Malmö, Sweden
| |
Collapse
|
34
|
Zucchi E, Bonetto V, Sorarù G, Martinelli I, Parchi P, Liguori R, Mandrioli J. Neurofilaments in motor neuron disorders: towards promising diagnostic and prognostic biomarkers. Mol Neurodegener 2020; 15:58. [PMID: 33059698 PMCID: PMC7559190 DOI: 10.1186/s13024-020-00406-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 09/18/2020] [Indexed: 12/11/2022] Open
Abstract
Motor neuron diseases (MNDs) are etiologically and biologically heterogeneous diseases. The pathobiology of motor neuron degeneration is still largely unknown, and no effective therapy is available. Heterogeneity and lack of specific disease biomarkers have been appointed as leading reasons for past clinical trial failure, and biomarker discovery is pivotal in today's MND research agenda.In the last decade, neurofilaments (NFs) have emerged as promising biomarkers for the clinical assessment of neurodegeneration. NFs are scaffolding proteins with predominant structural functions contributing to the axonal cytoskeleton of myelinated axons. NFs are released in CSF and peripheral blood as a consequence of axonal degeneration, irrespective of the primary causal event. Due to the current availability of highly-sensitive automated technologies capable of precisely quantify proteins in biofluids in the femtomolar range, it is now possible to reliably measure NFs not only in CSF but also in blood.In this review, we will discuss how NFs are impacting research and clinical management in ALS and other MNDs. Besides contributing to the diagnosis at early stages by differentiating between MNDs with different clinical evolution and severity, NFs may provide a useful tool for the early enrolment of patients in clinical trials. Due to their stability across the disease, NFs convey prognostic information and, on a larger scale, help to stratify patients in homogenous groups. Shortcomings of NFs assessment in biofluids will also be discussed according to the available literature in the attempt to predict the most appropriate use of the biomarker in the MND clinic.
Collapse
Affiliation(s)
- Elisabetta Zucchi
- Department of Biomedical, Metabolic and Neural Science, University of Modena and Reggio Emilia, Modena, Italy
| | - Valentina Bonetto
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Gianni Sorarù
- Neuromuscular Center, Department of Neurosciences, University of Padova, Padua, Italy.,Clinica Neurologica, Azienda Ospedaliera di Padova, Padua, Italy
| | - Ilaria Martinelli
- Department of Neurosciences, Azienda Ospedaliero Universitaria Modena, Modena, Italy
| | - Piero Parchi
- IRCCS Istituto delle Scienze Neurologiche, Ospedale Bellaria, Bologna, Italy.,Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Rocco Liguori
- IRCCS Istituto delle Scienze Neurologiche, Ospedale Bellaria, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Jessica Mandrioli
- Department of Neurosciences, Azienda Ospedaliero Universitaria Modena, Modena, Italy.
| |
Collapse
|
35
|
Cajanus A, Katisko K, Kontkanen A, Jääskeläinen O, Hartikainen P, Haapasalo A, Herukka SK, Vanninen R, Solje E, Hall A, Remes AM. Serum neurofilament light chain in FTLD: association with C9orf72, clinical phenotype, and prognosis. Ann Clin Transl Neurol 2020; 7:903-910. [PMID: 32441885 PMCID: PMC7318100 DOI: 10.1002/acn3.51041] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/17/2020] [Accepted: 03/20/2020] [Indexed: 12/13/2022] Open
Abstract
Objective The aim of the present study was to compare the levels of serum neurofilament light chain (sNfL) in frontotemporal lobar degeneration (FTLD) patients of different clinical subtypes (bvFTD, PPA, and FTLD‐MND) and with or without the C9orf72 repeat expansion, and to correlate sNfL levels to disease progression, assessed by the brain atrophy rate and survival time. Methods The sNfL levels were determined from 78 FTLD patients (C9orf72 repeat expansion carriers [n = 26] and non‐carriers [n = 52]) with Single Molecule Array (SIMOA). The progression of brain atrophy was evaluated using repeated T1‐weighted MRI scans and the survival time from medical records. Results In the total FTLD cohort, sNfL levels were significantly higher in C9orf72 repeat expansion carriers compared to non‐carriers. Considering clinical phenotypes, sNfL levels were higher in the C9orf72 repeat expansion carriers than in the non‐carriers in bvFTD and PPA groups. Furthermore, sNfL levels were the highest in the FTLD‐MND group (median 105 pg/mL) and the lowest in the bvFTD group (median 27 pg/mL). Higher sNfL levels significantly correlated with frontal cortical atrophy rate and subcortical grey matter atrophy rate. The higher sNfL levels also associated with shorter survival time. Interpretation Our results indicate that the C9orf72 repeat expansion carriers show elevated sNFL levels compared to non‐carriers and that the levels differ among different clinical phenotypes of FTLD. Higher sNfL levels correlated with a shorter survival time and cortical and subcortical atrophy rates. Thus, sNfL could prove as a potential prognostic biomarker in FTLD.
Collapse
Affiliation(s)
- Antti Cajanus
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland
| | - Kasper Katisko
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland
| | - Aleksi Kontkanen
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland
| | - Olli Jääskeläinen
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland
| | - Päivi Hartikainen
- Neuro Center, Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Annakaisa Haapasalo
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Sanna-Kaisa Herukka
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland.,Neuro Center, Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Ritva Vanninen
- Department of Radiology, Kuopio University Hospital, Kuopio, Finland
| | - Eino Solje
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland.,Neuro Center, Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Anette Hall
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland
| | - Anne M Remes
- Unit of Clinical Neuroscience, Neurology, University of Oulu, Oulu, Finland.,Medical Research Center, Oulu University Hospital, Oulu, Finland
| |
Collapse
|
36
|
Baldacci F, Mazzucchi S, Della Vecchia A, Giampietri L, Giannini N, Koronyo-Hamaoui M, Ceravolo R, Siciliano G, Bonuccelli U, Elahi FM, Vergallo A, Lista S, Giorgi FS. The path to biomarker-based diagnostic criteria for the spectrum of neurodegenerative diseases. Expert Rev Mol Diagn 2020; 20:421-441. [PMID: 32066283 PMCID: PMC7445079 DOI: 10.1080/14737159.2020.1731306] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 02/14/2020] [Indexed: 12/21/2022]
Abstract
Introduction: The postmortem examination still represents the reference standard for detecting the pathological nature of chronic neurodegenerative diseases (NDD). This approach displays intrinsic conceptual limitations since NDD represent a dynamic spectrum of partially overlapping phenotypes, shared pathomechanistic alterations that often give rise to mixed pathologies.Areas covered: We scrutinized the international clinical diagnostic criteria of NDD and the literature to provide a roadmap toward a biomarker-based classification of the NDD spectrum. A few pathophysiological biomarkers have been established for NDD. These are time-consuming, invasive, and not suitable for preclinical detection. Candidate screening biomarkers are gaining momentum. Blood neurofilament light-chain represents a robust first-line tool to detect neurodegeneration tout court and serum progranulin helps detect genetic frontotemporal dementia. Ultrasensitive assays and retinal scans may identify Aβ pathology early, in blood and the eye, respectively. Ultrasound also represents a minimally invasive option to investigate the substantia nigra. Protein misfolding amplification assays may accurately detect α-synuclein in biofluids.Expert opinion: Data-driven strategies using quantitative rather than categorical variables may be more reliable for quantification of contributions from pathophysiological mechanisms and their spatial-temporal evolution. A systems biology approach is suitable to untangle the dynamics triggering loss of proteostasis, driving neurodegeneration and clinical evolution.
Collapse
Affiliation(s)
- Filippo Baldacci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l’hôpital, Paris, France
| | - Sonia Mazzucchi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Linda Giampietri
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Nicola Giannini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Roberto Ceravolo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Ubaldo Bonuccelli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Fanny M. Elahi
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Andrea Vergallo
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l’hôpital, Paris, France
- Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, Paris, France
- Department of Neurology, Institute of Memory and Alzheimer’s Disease (IM2A), Pitié-Salpêtrière Hospital, Paris, France
| | - Simone Lista
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l’hôpital, Paris, France
- Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, Paris, France
- Department of Neurology, Institute of Memory and Alzheimer’s Disease (IM2A), Pitié-Salpêtrière Hospital, Paris, France
| | - Filippo Sean Giorgi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | |
Collapse
|
37
|
Heller C, Foiani MS, Moore K, Convery R, Bocchetta M, Neason M, Cash DM, Thomas D, Greaves CV, Woollacott IO, Shafei R, Van Swieten JC, Moreno F, Sanchez-Valle R, Borroni B, Laforce R, Masellis M, Tartaglia MC, Graff C, Galimberti D, Rowe JB, Finger E, Synofzik M, Vandenberghe R, de Mendonca A, Tagliavini F, Santana I, Ducharme S, Butler CR, Gerhard A, Levin J, Danek A, Frisoni G, Sorbi S, Otto M, Heslegrave AJ, Zetterberg H, Rohrer JD. Plasma glial fibrillary acidic protein is raised in progranulin-associated frontotemporal dementia. J Neurol Neurosurg Psychiatry 2020; 91:263-270. [PMID: 31937580 DOI: 10.1136/jnnp-2019-321954] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 11/20/2019] [Accepted: 12/02/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND There are few validated fluid biomarkers in frontotemporal dementia (FTD). Glial fibrillary acidic protein (GFAP) is a measure of astrogliosis, a known pathological process of FTD, but has yet to be explored as potential biomarker. METHODS Plasma GFAP and neurofilament light chain (NfL) concentration were measured in 469 individuals enrolled in the Genetic FTD Initiative: 114 C9orf72 expansion carriers (74 presymptomatic, 40 symptomatic), 119 GRN mutation carriers (88 presymptomatic, 31 symptomatic), 53 MAPT mutation carriers (34 presymptomatic, 19 symptomatic) and 183 non-carrier controls. Biomarker measures were compared between groups using linear regression models adjusted for age and sex with family membership included as random effect. Participants underwent standardised clinical assessments including the Mini-Mental State Examination (MMSE), Frontotemporal Lobar Degeneration-Clinical Dementia Rating scale and MRI. Spearman's correlation coefficient was used to investigate the relationship of plasma GFAP to clinical and imaging measures. RESULTS Plasma GFAP concentration was significantly increased in symptomatic GRN mutation carriers (adjusted mean difference from controls 192.3 pg/mL, 95% CI 126.5 to 445.6), but not in those with C9orf72 expansions (9.0, -61.3 to 54.6), MAPT mutations (12.7, -33.3 to 90.4) or the presymptomatic groups. GFAP concentration was significantly positively correlated with age in both controls and the majority of the disease groups, as well as with NfL concentration. In the presymptomatic period, higher GFAP concentrations were correlated with a lower cognitive score (MMSE) and lower brain volume, while in the symptomatic period, higher concentrations were associated with faster rates of atrophy in the temporal lobe. CONCLUSIONS Raised GFAP concentrations appear to be unique to GRN-related FTD, with levels potentially increasing just prior to symptom onset, suggesting that GFAP may be an important marker of proximity to onset, and helpful for forthcoming therapeutic prevention trials.
Collapse
Affiliation(s)
- Carolin Heller
- UK Dementia Research Institute, Department of Neurodegenerative Disease, University College London, London, UK
| | - Martha S Foiani
- UK Dementia Research Institute, Department of Neurodegenerative Disease, University College London, London, UK
| | - Katrina Moore
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Rhian Convery
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Martina Bocchetta
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Mollie Neason
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - David M Cash
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK.,Centre for Medical Image Computing, University College London, London, UK
| | - David Thomas
- Neuradiological Academic Unit, UCL Queen Square Institute of Neurology, London, UK
| | - Caroline V Greaves
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Ione Oc Woollacott
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Rachelle Shafei
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - John C Van Swieten
- Department of Neurology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Fermin Moreno
- Cognitive Disorders Unit, Department of Neurology, Donostia University Hospital, San Sebastian, País Vasco, Spain
| | - Raquel Sanchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic, Barcelona, Spain
| | - Barbara Borroni
- Centre for Neurodegenerative Disorders, Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Robert Laforce
- Clinique Interdisciplinaire de Mémoire du CHU de Québec, Département des Sciences Neurologiques, Université Laval, Québec, Québec, Canada
| | - Mario Masellis
- Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Maria Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, Ontario, Canada
| | - Caroline Graff
- Department of Geriatric Medicine, Karolinska University Hospital-Huddinge, Stockholm, Sweden
| | - Daniela Galimberti
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Centro Dino Ferrari, Milan, Italy.,Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - James B Rowe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Elizabeth Finger
- Department of Clinical Neurological Sciences, University of Western Ontario, London, Ontario, Canada
| | - Matthis Synofzik
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, Tübingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), University of Tübingen, Tübingen, Germany
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | | | - Fabrizio Tagliavini
- Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Neurologico Carlo Besta, Milan, Italy
| | - Isabel Santana
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Simon Ducharme
- Department of Neurology and Neurosurgery, McGill University, Montreal, Québec, Canada
| | | | - Alex Gerhard
- Faculty of Medical and Human Sciences, Institute of Brain, Behaviour and Mental Health, University of Manchester, Manchester, UK
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Department of Neurology, Ludwig-Maximilians-University, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Adrian Danek
- Department of Neurology, Ludwig-Maximilians-University, Munich, Germany
| | | | - Sandro Sorbi
- Department of Neuroscience, Psychology, Drug Research, and Child Health, University of Florence, Florence, Italy
| | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Amanda J Heslegrave
- UK Dementia Research Institute, Department of Neurodegenerative Disease, University College London, London, UK
| | - Henrik Zetterberg
- UK Dementia Research Institute, Department of Neurodegenerative Disease, University College London, London, UK.,Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Jonathan D Rohrer
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | | |
Collapse
|
38
|
Abstract
Frontotemporal degeneration (FTD) is a heterogeneous spectrum of neurodegenerative disorders characterized by diverse clinical presentations, neuropathological characteristics, and underlying genetic causes. In the last few years, several advances in the knowledge of clinical and biological aspects have been accomplished and three major scenarios have emerged that will represent the core issues in the FTD scene over the next few years. Foremost, the development of cerebrospinal fluid and blood biomarkers as well as neuroimaging techniques will aid the pursuit of new diagnostic and prognostic markers able to identify the ongoing proteinopathy and predict disease progression, which is key in identifying and stratifying patients for enrolment in clinical trials as well as evaluating response to treatment. On the other hand, current research has focused on the first attempts to slow down or revert disease progression, with the identification of disease modulators associated with disease onset and the ongoing development of the first pharmacological treatments for both sporadic and genetic FTD. Future research will certainly improve our knowledge of FTD and possibly open up a new era of disease-modifying therapies for this still-orphan disorder.
Collapse
Affiliation(s)
- Barbara Borroni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, 25100, Italy
| | - Alberto Benussi
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, 25100, Italy
| |
Collapse
|
39
|
Forgrave LM, Ma M, Best JR, DeMarco ML. The diagnostic performance of neurofilament light chain in CSF and blood for Alzheimer's disease, frontotemporal dementia, and amyotrophic lateral sclerosis: A systematic review and meta-analysis. ALZHEIMER'S & DEMENTIA: DIAGNOSIS, ASSESSMENT & DISEASE MONITORING 2019; 11:730-743. [PMID: 31909174 PMCID: PMC6939029 DOI: 10.1016/j.dadm.2019.08.009] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction A systematic review and meta-analysis was performed regarding the diagnostic performance of neurofilament light chain (NfL) in CSF and blood. Methods A database search was conducted for NfL biomarker studies in the context of Alzheimer's disease (AD), frontotemporal dementia (FTD), and amyotrophic lateral sclerosis (ALS) compared with controls (i.e., cognitively unimpaired, mild cognitive impairment, or disease mimics). Results In groups with a sufficient number of studies, the performance of NfL in blood and CSF was similar. Compared with disease mimics, we observed that CSF NfL had strong discriminatory power for ALS, modest discriminatory power for FTD, and no discriminatory power for AD. NfL provided the greatest separation between ALS and cognitively unimpaired controls in both the blood and CSF, followed by FTD (CSF and blood), then AD (blood and CSF). Discussion Comparable performance of CSF and blood NfL in many groups demonstrates the promise of NfL as a noninvasive biomarker of neurodegeneration; however, its utility in clinically meaningful scenarios requires greater scrutiny. Toward clinical implementation, a more comprehensive understanding of NfL concentrations in disease subtypes with overlapping phenotypes and at defined stages of disease, and the development of a harmonization program, are warranted.
Collapse
Affiliation(s)
- Lauren M Forgrave
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Matthew Ma
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - John R Best
- Department of Psychiatry, University of British Columbia, Vancouver, Canada
| | - Mari L DeMarco
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada.,Department of Pathology and Laboratory Medicine, St. Paul's Hospital, Providence Health Care, Vancouver, Canada
| |
Collapse
|
40
|
Turner MR. The early biomarker challenge in neurodegenerative disorders. J Neurol Neurosurg Psychiatry 2019; 90:1190-1191. [PMID: 31243045 DOI: 10.1136/jnnp-2019-321145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 06/18/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Martin R Turner
- Nuffield Department of Clinical Neurosciences, Oxford University, Oxford, UK
| |
Collapse
|
41
|
Benatar M, Wuu J, Lombardi V, Jeromin A, Bowser R, Andersen PM, Malaspina A. Neurofilaments in pre-symptomatic ALS and the impact of genotype. Amyotroph Lateral Scler Frontotemporal Degener 2019; 20:538-548. [PMID: 31432691 DOI: 10.1080/21678421.2019.1646769] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Objective. To evaluate serum and cerebrospinal fluid (CSF) levels of phosphorylated neurofilament heavy (pNfH), and to compare these to levels of neurofilament light (NfL), as biomarkers of pre-symptomatic ALS. Design. The study population includes 34 controls, 79 individuals at-risk for ALS, 22 ALS patients, and 14 phenoconverters. At-risk individuals are enrolled through Pre-Symptomatic Familial ALS (Pre-fALS), a longitudinal natural history and biomarker study of individuals who are carriers of any ALS-associated gene mutation, but who demonstrate no clinical evidence of disease at the time of enrollment. pNfH and NfL in serum and CSF were quantified using established enzyme-linked immunosorbent assays. Results. There is a longitudinal increase in serum pNfH in advance of the emergence of clinically manifest ALS. A similar pattern is observed for NfL, but with the absolute levels also frequently exceeding a normative threshold. Although CSF data are more sparse, similar patterns are observed for both neurofilaments, with absolute levels exceeding a normative threshold prior to phenoconversion. In serum, these changes are observed in the 6-12 months prior to disease among SOD1 A4V mutation carriers, and as far back as 2 and 3.5 years, respectively, in individuals with a FUS c.521del6 mutation and a C9ORF72 hexanucleotide repeat expansion. Conclusions. Serum and CSF pNfH increase prior to phenoconversion. In CSF, the temporal course of these changes is similar to NfL. In serum, however, pNfH is less sensitive to pre-symptomatic disease than NfL. The duration of pre-symptomatic disease, as defined by changes in neurofilaments, may vary depending on underlying genotype.
Collapse
Affiliation(s)
- Michael Benatar
- Department of Neurology, University of Miami , Miami , FL , USA
| | - Joanne Wuu
- Department of Neurology, University of Miami , Miami , FL , USA
| | - Vittoria Lombardi
- Neuroscience Center, Blizard, Institute of Cell and Molecular Medicine, Barts & the London School of Medicine & Dentistry , London , UK
| | | | | | - Peter M Andersen
- Department of Pharmacology and Clinical Neuroscience, Umeå University , Umeå , Sweden
| | - Andrea Malaspina
- Neuroscience Center, Blizard, Institute of Cell and Molecular Medicine, Barts & the London School of Medicine & Dentistry , London , UK
| |
Collapse
|
42
|
An update on genetic frontotemporal dementia. J Neurol 2019; 266:2075-2086. [PMID: 31119452 PMCID: PMC6647117 DOI: 10.1007/s00415-019-09363-4] [Citation(s) in RCA: 242] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 05/01/2019] [Accepted: 05/03/2019] [Indexed: 12/12/2022]
Abstract
Frontotemporal dementia (FTD) is a highly heritable group of neurodegenerative disorders, with around 30% of patients having a strong family history. The majority of that heritability is accounted for by autosomal dominant mutations in the chromosome 9 open reading frame 72 (C9orf72), progranulin (GRN), and microtubule-associated protein tau (MAPT) genes, with mutations more rarely seen in a number of other genes. This review will discuss the recent updates in the field of genetic FTD. Age at symptom onset in genetic FTD is variable with recently identified genetic modifiers including TMEM106B (in GRN carriers particularly) and a polymorphism at a locus containing two overlapping genes LOC101929163 and C6orf10 (in C9orf72 carriers). Behavioural variant FTD (bvFTD) is the most common diagnosis in each of the genetic groups, although in C9orf72 carriers amyotrophic lateral sclerosis either alone, or with bvFTD, is also common. An atypical neuropsychiatric presentation is also seen in C9orf72 carriers and family members of carriers are at greater risk of psychiatric disorders including schizophrenia and autistic spectrum disorders. Large natural history studies of presymptomatic genetic FTD are now underway both in Europe/Canada (GENFI—the Genetic FTD Initiative) and in the US (ARTFL/LEFFTDS study), collaborating together under the banner of the FTD Prevention Initiative (FPI). These studies are taking forward the validation of cognitive, imaging and fluid biomarkers that aim to robustly measure disease onset, staging and progression in genetic FTD. Grey matter changes on MRI and hypometabolism on FDG-PET are seen at least 10 years before symptom onset with white matter abnormalities seen earlier, but the pattern and exact timing of changes differ between different genetic groups. In contrast, tau PET has yet to show promise in genetic FTD. Three key fluid biomarkers have been identified so far that are likely to be helpful in clinical trials—CSF or blood neurofilament light chain levels (in all groups), CSF or blood progranulin levels (in GRN carriers) and CSF poly(GP) dipeptide repeat protein levels (in C9orf72 carriers). Increased knowledge about genetic FTD has led to more clinical presymptomatic genetic testing but this has not yet been mirrored in the development of either an accepted FTD-specific testing protocol or provision of appropriate psychological support mechanisms for those living through the at-risk phase. This will become even more relevant as disease-modifying therapy trials start in each of the genetic groups over the next few years.
Collapse
|
43
|
Molecular Mechanisms of Neurodegeneration Related to C9orf72 Hexanucleotide Repeat Expansion. Behav Neurol 2019; 2019:2909168. [PMID: 30774737 PMCID: PMC6350563 DOI: 10.1155/2019/2909168] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/28/2018] [Accepted: 09/18/2018] [Indexed: 12/11/2022] Open
Abstract
Two clinically distinct diseases, amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), have recently been classified as two extremes of the FTD/ALS spectrum. The neuropathological correlate of FTD is frontotemporal lobar degeneration (FTLD), characterized by tau-, TDP-43-, and FUS-immunoreactive neuronal inclusions. An earlier discovery that a hexanucleotide repeat expansion mutation in chromosome 9 open reading frame 72 (C9orf72) gene causes ALS and FTD established a special subtype of ALS and FTLD with TDP-43 pathology (C9FTD/ALS). Normal individuals carry 2–10 hexanucleotide GGGGCC repeats in the C9orf72 gene, while more than a few hundred repeats represent a risk for ALS and FTD. The proposed molecular mechanisms by which C9orf72 repeat expansions induce neurodegenerative changes are C9orf72 loss-of-function through haploinsufficiency, RNA toxic gain-of-function, and gain-of-function through the accumulation of toxic dipeptide repeat proteins. However, many more cellular processes are affected by pathological processes in C9FTD/ALS, including nucleocytoplasmic transport, RNA processing, normal function of nucleolus, formation of membraneless organelles, translation, ubiquitin proteasome system, Notch signalling pathway, granule transport, and normal function of TAR DNA-binding protein 43 (TDP-43). Although the exact molecular mechanisms through which C9orf72 repeat expansions account for neurodegeneration have not been elucidated, some potential therapeutics, such as antisense oligonucleotides targeting hexanucleotide GGGGCC repeats in mRNA, were successful in preclinical trials and are awaiting phase 1 clinical trials. In this review, we critically discuss each proposed mechanism and provide insight into the most recent studies aiming to elucidate the molecular underpinnings of C9FTD/ALS.
Collapse
|
44
|
Panman JL, Jiskoot LC, Bouts MJRJ, Meeter LHH, van der Ende EL, Poos JM, Feis RA, Kievit AJA, van Minkelen R, Dopper EGP, Rombouts SARB, van Swieten JC, Papma JM. Gray and white matter changes in presymptomatic genetic frontotemporal dementia: a longitudinal MRI study. Neurobiol Aging 2019; 76:115-124. [PMID: 30711674 DOI: 10.1016/j.neurobiolaging.2018.12.017] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 12/19/2018] [Accepted: 12/27/2018] [Indexed: 10/27/2022]
Abstract
In genetic frontotemporal dementia, cross-sectional studies have identified profiles of presymptomatic neuroanatomical loss for C9orf72 repeat expansion, MAPT, and GRN mutations. In this study, we characterize longitudinal gray matter (GM) and white matter (WM) brain changes in presymptomatic frontotemporal dementia. We included healthy carriers of C9orf72 repeat expansion (n = 12), MAPT (n = 15), GRN (n = 33) mutations, and related noncarriers (n = 53), that underwent magnetic resonance imaging at baseline and 2-year follow-up. We analyzed cross-sectional baseline, follow-up, and longitudinal GM and WM changes using voxel-based morphometry and cortical thickness analysis in SPM and tract-based spatial statistics in FSL. Compared with noncarriers, C9orf72 repeat expansion carriers showed lower GM volume in the cerebellum and insula, and WM differences in the anterior thalamic radiation, at baseline and follow-up. MAPT mutation carriers showed emerging GM temporal lobe changes and longitudinal WM degeneration of the uncinate fasciculus. GRN mutation carriers did not show presymptomatic neurodegeneration. This study shows distinct presymptomatic cross-sectional and longitudinal patterns of GM and WM changes across C9orf72 repeat expansion, MAPT, and GRN mutation carriers compared with noncarriers.
Collapse
Affiliation(s)
- Jessica L Panman
- Department of Neurology, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Lize C Jiskoot
- Department of Neurology, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Mark J R J Bouts
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands; Institute of Psychology, Leiden University, Leiden, the Netherlands
| | - Lieke H H Meeter
- Department of Neurology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Emma L van der Ende
- Department of Neurology, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Jackie M Poos
- Department of Neurology, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Rogier A Feis
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands; Institute of Psychology, Leiden University, Leiden, the Netherlands; Leiden Institute for Brain and Cognition, Leiden University, Leiden, the Netherlands
| | - Anneke J A Kievit
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Rick van Minkelen
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Elise G P Dopper
- Department of Neurology, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands; Department of Neurology, VU medical Center, Amsterdam, the Netherlands
| | - Serge A R B Rombouts
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands; Institute of Psychology, Leiden University, Leiden, the Netherlands; Leiden Institute for Brain and Cognition, Leiden University, Leiden, the Netherlands
| | - John C van Swieten
- Department of Neurology, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Clinical Genetics, VU Medical Center, Amsterdam, the Netherlands
| | - Janne M Papma
- Department of Neurology, Erasmus Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
45
|
Floeter MK, Gendron TF. Biomarkers for Amyotrophic Lateral Sclerosis and Frontotemporal Dementia Associated With Hexanucleotide Expansion Mutations in C9orf72. Front Neurol 2018; 9:1063. [PMID: 30568632 PMCID: PMC6289985 DOI: 10.3389/fneur.2018.01063] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 11/21/2018] [Indexed: 12/11/2022] Open
Abstract
Now that genetic testing can identify persons at risk for developing amyotrophic lateral sclerosis (ALS) many decades before symptoms begin, there is a critical need for biomarkers that signal the onset and progression of degeneration. The search for candidate disease biomarkers in patients with mutations in the gene C9orf72 has included imaging, physiology, and biofluid measurements. In cross-sectional imaging studies, C9+ ALS patients display diffuse reductions of gray and white matter integrity compared to ALS patients without mutations. This structural imaging signature overlaps with frontotemporal dementia (FTD), reflecting the frequent co-occurrence of cognitive impairment, even frank FTD, in C9+ ALS patients. Changes in functional connectivity occur as critical components of the networks associated with cognition and behavior degenerate. In presymptomatic C9+carriers, subtle differences in volumes of subcortical structures and functional connectivity can be detected, often decades before the typical family age of symptom onset. Dipeptide repeat proteins produced by the repeat expansion mutation are also measurable in the cerebrospinal fluid (CSF) of presymptomatic gene carriers, possibly throughout their lives. In contrast, a rise in the level of neurofilament proteins in the CSF appears to presage the onset of degeneration in presymptomatic carriers in one longitudinal study. Cross-sectional studies indicate that neurofilament protein levels may provide prognostic information for survival in C9+ ALS patients. Longitudinal studies will be needed to validate the candidate biomarkers discussed here. Understanding how these candidate biomarkers change over time is critical if they are to be used in future therapeutic decisions.
Collapse
Affiliation(s)
- Mary Kay Floeter
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Tania F Gendron
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
| |
Collapse
|
46
|
Zetterberg H, van Swieten JC, Boxer AL, Rohrer JD. Review: Fluid biomarkers for frontotemporal dementias. Neuropathol Appl Neurobiol 2018; 45:81-87. [DOI: 10.1111/nan.12530] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 11/05/2018] [Indexed: 12/13/2022]
Affiliation(s)
- H. 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; UCL Queen Square Institute of Neurology; Queen Square London UK
- UK Dementia Research Institute at UCL; London UK
| | - J. C. van Swieten
- Department of Neurology; Erasmus Medical Center; Rotterdam The Netherlands
- Department of Clinical Genetics; VU University Medical Center; Amsterdam The Netherlands
| | - A. L. Boxer
- Memory and Aging Center; Department of Neurology; University of California; San Francisco CA USA
| | - J. D. Rohrer
- Department of Neurodegenerative Disease; UCL Queen Square Institute of Neurology; Queen Square London UK
| |
Collapse
|
47
|
Steinacker P, Barschke P, Otto M. Biomarkers for diseases with TDP-43 pathology. Mol Cell Neurosci 2018; 97:43-59. [PMID: 30399416 DOI: 10.1016/j.mcn.2018.10.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 10/26/2018] [Accepted: 10/29/2018] [Indexed: 01/01/2023] Open
Abstract
The discovery that aggregated transactive response DNA-binding protein 43 kDa (TDP-43) is the major component of pathological ubiquitinated inclusions in amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) caused seminal progress in the unveiling of the genetic bases and molecular characteristics of these now so-called TDP-43 proteinopathies. Substantial increase in the knowledge of clinic-pathological coherencies, especially for FTLD variants, could be made in the last decade, but also revealed a considerable complexity of TDP-43 pathology and often a poor correlation of clinical and molecular disease characteristics. To date, an underlying TDP-43 pathology can be predicted only for patients with mutations in the genes C9orf72 and GRN, but is dependent on neuropathological verification in patients without family history, which represent the majority of cases. As etiology-specific therapies for neurodegenerative proteinopathies are emerging, methods to forecast TDP-43 pathology at patients' lifetime are highly required. Here, we review the current status of research pursued to identify specific indicators to predict or exclude TDP-43 pathology in the ALS-FTLD spectrum disorders and findings on candidates for prognosis and monitoring of disease progression in TDP-43 proteinopathies with a focus on TDP-43 with its pathological forms, neurochemical and imaging biomarkers.
Collapse
Affiliation(s)
| | - Peggy Barschke
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany.
| |
Collapse
|
48
|
Kanata E, Golanska E, Villar-Piqué A, Karsanidou A, Dafou D, Xanthopoulos K, Schmitz M, Ferrer I, Karch A, Sikorska B, Liberski PP, Sklaviadis T, Zerr I, Llorens F. Cerebrospinal fluid neurofilament light in suspected sporadic Creutzfeldt-Jakob disease. J Clin Neurosci 2018; 60:124-127. [PMID: 30309804 DOI: 10.1016/j.jocn.2018.09.031] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 09/26/2018] [Indexed: 12/12/2022]
Abstract
Sporadic Creutzfeldt-Jakob disease (sCJD) is the most common form of human prion disease. It is invariably fatal and displays a short clinical disease stage. The key event in sCJD is the propagation of a beta-sheet rich conformer of the physiological PrPC protein, known as PrPSc. Neuropathological disease characteristics include gliosis, neuronal loss and spongiform degeneration; disease clinical manifestations refer to mental and visual disabilities, cognitive impairment, gait or limb ataxia, myoclonus and mutism. Definite sCJD diagnosis requires post-mortem brain material histopathological examination. However, highly certain pre-mortem differential diagnosis is desired to exclude other treatable disorders and to reduce disease transmission risks. Detection and/or quantification of cerebrospinal fluid (CSF) biomarkers reflecting neuronal damage and PrPC misfolding in the diseased brain significantly enhance pre-mortem diagnosis. Previously established and newly identified biomarkers are used towards this direction. Increased CSF Neurofilament light chain (NFL) concentrations have been reported in several neurological disorders, including prion diseases. In the present study, we analyzed CSF NFL levels in two independent patient cohorts, consisting of highly suspected sCJD cases that were further classified as sCJD or non-CJD according to established diagnostic criteria. CSF NFL concentrations were increased in sCJD compared to non-CJD cases in both cohorts (area under the curve (with 95% confidence interval) equal to 0.89 (0.82 to 0.97) and 0.86 (0.77 to 0.96), respectively. CSF NFL was associated neither to age nor to sex but correlated with total-tau concentrations in both cohorts. Overall, our data provide independent validation of CSF NFL utility in sCJD differential diagnosis.
Collapse
Affiliation(s)
- Eirini Kanata
- Laboratory of Pharmacology, School of Health Sciences, Department of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | - Ewa Golanska
- Department of Molecular Pathology and Neuropathology, Medical University of Lodz, Lodz, Poland
| | - Anna Villar-Piqué
- Department of Neurology, University Medical School, Göttingen, Germany
| | - Aikaterini Karsanidou
- Laboratory of Pharmacology, School of Health Sciences, Department of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimitra Dafou
- Department of Genetics, Development, and Molecular Biology, School of Biology, Aristotle University, Thessaloniki, Greece
| | - Konstantinos Xanthopoulos
- Laboratory of Pharmacology, School of Health Sciences, Department of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Matthias Schmitz
- Department of Neurology, University Medical School, Göttingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Isidro Ferrer
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Institute Carlos III, Ministry of Health, L'Hospitalet de Llobregat, Spain; Bellvitge University Hospital-IDIBELL, Department of Pathology and Experimental Therapeutics, University of Barcelona, Hospitalet de Llobregat, Spain
| | - André Karch
- Department of Epidemiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Beata Sikorska
- Department of Molecular Pathology and Neuropathology, Medical University of Lodz, Lodz, Poland
| | - Pawel P Liberski
- Department of Molecular Pathology and Neuropathology, Medical University of Lodz, Lodz, Poland
| | - Theodoros Sklaviadis
- Laboratory of Pharmacology, School of Health Sciences, Department of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Inga Zerr
- Department of Neurology, University Medical School, Göttingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Franc Llorens
- Department of Neurology, University Medical School, Göttingen, Germany; Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Institute Carlos III, Ministry of Health, L'Hospitalet de Llobregat, Spain; Bellvitge Biomedical Research Institute (IDBELL), L'Hospitalet de Llobregat, Spain.
| |
Collapse
|