1
|
Bampton A, McHutchison C, Talbot K, Benatar M, Thompson AG, Turner MR. The Basis of Cognitive and Behavioral Dysfunction in Amyotrophic Lateral Sclerosis. Brain Behav 2024; 14:e70115. [PMID: 39501538 PMCID: PMC11538089 DOI: 10.1002/brb3.70115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/29/2024] [Accepted: 10/05/2024] [Indexed: 11/09/2024] Open
Abstract
OBJECTIVE To summarize and evaluate evidence pertaining to the clinical, genetic, histopathological, and neuroimaging correlates of cognitive and behavioral dysfunction in amyotrophic lateral sclerosis (ALS). METHODOLOGY We comprehensively reviewed the literature on cognitive and behavioral manifestations of ALS, narrating findings from both cross-sectional and longitudinal studies. We discussed knowledge gaps in the evidence base and key limitations affecting studies to date, before formulating a framework for future research paradigms aimed at investigating clinicopathological correlates of neuropsychological dysfunction in ALS. RESULTS Studies have demonstrated clinical associations with cognitive dysfunction in ALS e.g., bulbar-onset of symptoms, pathological associations (extramotor TDP-43 deposition), and imaging associations (frontotemporal involvement). The most common behavioral deficit, apathy, is highly associated with verbal fluency, but longitudinal studies assessing behavioral dysfunction in ALS are comparatively lacking. CONCLUSION Longitudinal studies have been helpful in identifying several potential correlates of cognitive and behavioral dysfunction but have frequently been confounded by selection bias and inappropriate testing platforms. This review provides a framework for more robust assessment of clinicopathological associations of neuropsychological abnormalities in ALS in the future, advocating for greater utilization of pre-symptomatic C9orf72 repeat expansion-carrying cohorts.
Collapse
Affiliation(s)
- Alexander Bampton
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | | | - Kevin Talbot
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Michael Benatar
- Department of NeurologyUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | | | - Martin R. Turner
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| |
Collapse
|
2
|
Santos Silva C, Gormicho M, Simão S, Pronto-Laborinho AC, Alves I, Pinto S, Oliveira Santos M, de Carvalho M. C9orf72 gene repeat expansion phenotype profile of motor neurone disease in Portugal. J Neurol Sci 2024; 465:123208. [PMID: 39226712 DOI: 10.1016/j.jns.2024.123208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/18/2024] [Accepted: 08/29/2024] [Indexed: 09/05/2024]
Abstract
BACKGROUND C9orf72 gene repeat expansion (C9RE) is the most frequent gene variant associated with amyotrophic lateral sclerosis (ALS). We aimed to study the phenotype of motor neurone disease (MND) patients with C9RE in a Portuguese cohort. METHODS Demographical and clinical data of MND patients with (C9RE+) and without C9RE were compared. ALS al Rating Scale-Revised (ALSFRS-R) and Edinburgh Cognitive and Behavioural ALS Screen (ECAS) were used to evaluate functional and cognitive performance, respectively. Survival analysis was performed using Kaplan Meier log-rank test and Cox proportional hazards model. RESULTS We included 761 patients of whom 61 (8.0 %) were C9RE+. C9RE+ patients had a higher frequency of ALS (95.1 vs 78.4 %, p = 0.002), and lower frequency of progressive muscular atrophy (3.3 vs 16.7 %, p = 0.006). C9RE+ was associated with earlier age of onset (58.1 vs 62.6 years, p = 0.003) and more frequent MND family history (65.5 vs 11.4 %, p < 0.001). Gender, ethnicity, onset site, diagnostic delay, disease progression rate until diagnosis (ΔF), ALSFRS-R and time until non-invasive ventilation did not differ between groups. Cognitive/behavioural symptoms and ECAS did not differ between groups, except a worse visuospatial score in C9RE+ group (p = 0.035). Death rate was 1.8 and 1.6 times higher in C9RE+ patients with MND and ALS, respectively. Significant survival prognostic factors in C9RE+ group were diagnosis delay (HR = 0.96, 95 %CI 0.92-0.99, p = 0.008) and ΔF (HR = 1.93, 95 %CI 1.26-2.96, p = 0.002). CONCLUSION Our study corroborates most previous cohorts' findings, but harbours some singularities regarding onset site, phenotype, and cognitive profile, that contribute to a better understanding of C9RE epidemiology.
Collapse
Affiliation(s)
- Cláudia Santos Silva
- Department of Neurosciences and Mental Health, Unidade Local de Saúde de Santa Maria, Lisbon, Portugal; Faculdade de Medicina-Instituto de Medicina Molecular, Centro de Estudos Egas Moniz, Universidade de Lisboa, Lisboa, Portugal.
| | - Marta Gormicho
- Faculdade de Medicina-Instituto de Medicina Molecular, Centro de Estudos Egas Moniz, Universidade de Lisboa, Lisboa, Portugal
| | - Sara Simão
- Faculdade de Medicina-Instituto de Medicina Molecular, Centro de Estudos Egas Moniz, Universidade de Lisboa, Lisboa, Portugal
| | - Ana Catarina Pronto-Laborinho
- Faculdade de Medicina-Instituto de Medicina Molecular, Centro de Estudos Egas Moniz, Universidade de Lisboa, Lisboa, Portugal
| | - Inês Alves
- Faculdade de Medicina-Instituto de Medicina Molecular, Centro de Estudos Egas Moniz, Universidade de Lisboa, Lisboa, Portugal
| | - Susana Pinto
- Faculdade de Medicina-Instituto de Medicina Molecular, Centro de Estudos Egas Moniz, Universidade de Lisboa, Lisboa, Portugal
| | - Miguel Oliveira Santos
- Department of Neurosciences and Mental Health, Unidade Local de Saúde de Santa Maria, Lisbon, Portugal; Faculdade de Medicina-Instituto de Medicina Molecular, Centro de Estudos Egas Moniz, Universidade de Lisboa, Lisboa, Portugal
| | - Mamede de Carvalho
- Department of Neurosciences and Mental Health, Unidade Local de Saúde de Santa Maria, Lisbon, Portugal; Faculdade de Medicina-Instituto de Medicina Molecular, Centro de Estudos Egas Moniz, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
3
|
Denning AE, Ittyerah R, Levorse LM, Sadeghpour N, Athalye C, Chung E, Ravikumar S, Dong M, Duong MT, Li Y, Ilesanmi A, Sreepada LP, Sabatini P, Lowe M, Bahena A, Zablah J, Spencer BE, Watanabe R, Kim B, Sørensen MH, Khandelwal P, Brown C, Hrybouski S, Xie SX, de Flores R, Robinson JL, Schuck T, Ohm DT, Arezoumandan S, Porta S, Detre JA, Insausti R, Wisse LEM, Das SR, Irwin DJ, Lee EB, Wolk DA, Yushkevich PA. Association of quantitative histopathology measurements with antemortem medial temporal lobe cortical thickness in the Alzheimer's disease continuum. Acta Neuropathol 2024; 148:37. [PMID: 39227502 PMCID: PMC11371872 DOI: 10.1007/s00401-024-02789-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/07/2024] [Accepted: 08/15/2024] [Indexed: 09/05/2024]
Abstract
The medial temporal lobe (MTL) is a hotspot for neuropathology, and measurements of MTL atrophy are often used as a biomarker for cognitive decline associated with neurodegenerative disease. Due to the aggregation of multiple proteinopathies in this region, the specific relationship of MTL atrophy to distinct neuropathologies is not well understood. Here, we develop two quantitative algorithms using deep learning to measure phosphorylated tau (p-tau) and TDP-43 (pTDP-43) pathology, which are both known to accumulate in the MTL and are associated with MTL neurodegeneration. We focus on these pathologies in the context of Alzheimer's disease (AD) and limbic predominant age-related TDP-43 encephalopathy (LATE) and apply our deep learning algorithms to distinct histology sections, on which MTL subregions were digitally annotated. We demonstrate that both quantitative pathology measures show high agreement with expert visual ratings of pathology and discriminate well between pathology stages. In 140 cases with antemortem MR imaging, we compare the association of semi-quantitative and quantitative postmortem measures of these pathologies in the hippocampus with in vivo structural measures of the MTL and its subregions. We find widespread associations of p-tau pathology with MTL subregional structural measures, whereas pTDP-43 pathology had more limited associations with the hippocampus and entorhinal cortex. Quantitative measurements of p-tau pathology resulted in a significantly better model of antemortem structural measures than semi-quantitative ratings and showed strong associations with cortical thickness and volume. By providing a more granular measure of pathology, the quantitative p-tau measures also showed a significant negative association with structure in a severe AD subgroup where semi-quantitative ratings displayed a ceiling effect. Our findings demonstrate the advantages of using quantitative neuropathology to understand the relationship of pathology to structure, particularly for p-tau, and motivate the use of quantitative pathology measurements in future studies.
Collapse
Affiliation(s)
- Amanda E Denning
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA.
| | - Ranjit Ittyerah
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Lisa M Levorse
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Chinmayee Athalye
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Eunice Chung
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Sadhana Ravikumar
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Mengjin Dong
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael Tran Duong
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Yue Li
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Ademola Ilesanmi
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Lasya P Sreepada
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Philip Sabatini
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - MaKayla Lowe
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Alejandra Bahena
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jamila Zablah
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Barbara E Spencer
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Ryohei Watanabe
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Neurodegenerative Disease Research, Institute On Aging, University of Pennsylvania, Philadelphia, PA, USA
| | - Boram Kim
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Neurodegenerative Disease Research, Institute On Aging, University of Pennsylvania, Philadelphia, PA, USA
| | - Maja Højvang Sørensen
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Neurodegenerative Disease Research, Institute On Aging, University of Pennsylvania, Philadelphia, PA, USA
| | - Pulkit Khandelwal
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher Brown
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Sharon X Xie
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Robin de Flores
- UMR-S U1237, PhIND "Physiopathology and Imaging of Neurological Disorders", Institut Blood and Brain @ Caen-Normandie, INSERM, Caen-Normandie University, GIP Cyceron, Caen, France
| | - John L Robinson
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Neurodegenerative Disease Research, Institute On Aging, University of Pennsylvania, Philadelphia, PA, USA
| | - Theresa Schuck
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Neurodegenerative Disease Research, Institute On Aging, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel T Ohm
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Sanaz Arezoumandan
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Sílvia Porta
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Neurodegenerative Disease Research, Institute On Aging, University of Pennsylvania, Philadelphia, PA, USA
| | - John A Detre
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Ricardo Insausti
- Human Neuroanatomy Lab, University of Castilla La Mancha, Albacete, Spain
| | - Laura E M Wisse
- Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Sandhitsu R Das
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - David J Irwin
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward B Lee
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Neurodegenerative Disease Research, Institute On Aging, University of Pennsylvania, Philadelphia, PA, USA
| | - David A Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul A Yushkevich
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
4
|
Peters RM, Schieszler-Ockrassa CM, Gleason A, Patterson K. Phenocopy behavioral variant frontotemporal dementia: A case study. Clin Neuropsychol 2024; 38:1256-1271. [PMID: 38360583 DOI: 10.1080/13854046.2024.2315726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 11/30/2023] [Indexed: 02/17/2024]
Abstract
Objective: Behavioral variant frontotemporal dementia (bvFTD) is a neurodegenerative condition characterized by progressive changes in behavior, cognition, and day-to-day functioning. Progression of the disease usually leads to death 3-5 years after diagnosis. However, there are reports of individuals who are initially diagnosed with bvFTD but fail to progress. These individuals are thought to have what is becoming known as phenocopy bvFTD (phFTD). Methods: This manuscript reviews a single case study of a 68-year-old male Veteran who was diagnosed with bvFTD in 2010, which has not progressed over time. Results: Review of serial neuropsychological evaluations was broadly normal with mild evidence of executive dysfunction with minimal reliable change in his performances from 2015, 2017, and 2022 evaluations. He also has not developed neuroimaging evidence of FTD. Conclusions: This case illustrates the importance of monitoring individuals over time and incorporating neuroimaging data into the diagnosis. We believe this Veteran's presentation is most consistent with what has been described as phFTD.
Collapse
Affiliation(s)
| | | | - Angela Gleason
- Clement J. Zablocki VA Medical Center, Milwaukee, WI, USA
| | | |
Collapse
|
5
|
Nigri A, Stanziano M, Fedeli D, Manera U, Ferraro S, Medina Carrion JP, Palermo S, Lequio L, Denegri F, Agosta F, Spinelli EG, Filippi M, Grisoli M, Valentini MC, De Mattei F, Canosa A, Calvo A, Chiò A, Bruzzone MG, Moglia C. Distinct neural signatures of pulvinar in C9orf72 amyotrophic lateral sclerosis mutation carriers and noncarriers. Eur J Neurol 2024; 31:e16266. [PMID: 38469975 PMCID: PMC11235848 DOI: 10.1111/ene.16266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 01/30/2024] [Accepted: 02/15/2024] [Indexed: 03/13/2024]
Abstract
BACKGROUND AND PURPOSE Thalamic alterations have been reported as a major feature in presymptomatic and symptomatic patients carrying the C9orf72 mutation across the frontotemporal dementia-amyotrophic lateral sclerosis (ALS) spectrum. Specifically, the pulvinar, a high-order thalamic nucleus and timekeeper for large-scale cortical networks, has been hypothesized to be involved in C9orf72-related neurodegenerative diseases. We investigated whether pulvinar volume can be useful for differential diagnosis in ALS C9orf72 mutation carriers and noncarriers and how underlying functional connectivity changes affect this region. METHODS We studied 19 ALS C9orf72 mutation carriers (ALSC9+) accurately matched with wild-type ALS (ALSC9-) and ALS mimic (ALSmimic) patients using structural and resting-state functional magnetic resonance imaging data. Pulvinar volume was computed using automatic segmentation. Seed-to-voxel functional connectivity analyses were performed using seeds from a pulvinar functional parcellation. RESULTS Pulvinar structural integrity had high discriminative values for ALSC9+ patients compared to ALSmimic (area under the curve [AUC] = 0.86) and ALSC9- (AUC = 0.77) patients, yielding a volume cutpoint of approximately 0.23%. Compared to ALSmimic, ALSC9- showed increased anterior, inferior, and lateral pulvinar connections with bilateral occipital-temporal-parietal regions, whereas ALSC9+ showed no differences. ALSC9+ patients when compared to ALSC9- patients showed reduced pulvinar-occipital connectivity for anterior and inferior pulvinar seeds. CONCLUSIONS Pulvinar volume could be a differential biomarker closely related to the C9orf72 mutation. A pulvinar-cortical circuit dysfunction might play a critical role in disease progression and development, in both the genetic phenotype and ALS wild-type patients.
Collapse
Affiliation(s)
- Anna Nigri
- Neuroradiology UnitFondazione IRCCS Istituto Neurologico Carlo BestaMilanItaly
| | - Mario Stanziano
- Neuroradiology UnitFondazione IRCCS Istituto Neurologico Carlo BestaMilanItaly
- ALS Centre, “Rita Levi Montalcini” Department of NeuroscienceUniversity of TurinTurinItaly
| | - Davide Fedeli
- Neuroradiology UnitFondazione IRCCS Istituto Neurologico Carlo BestaMilanItaly
| | - Umberto Manera
- ALS Centre, “Rita Levi Montalcini” Department of NeuroscienceUniversity of TurinTurinItaly
- Azienda Ospedaliero‐Universitaria Città della Salute e della Scienza di Torino, SC Neurologia 1UTurinItaly
| | - Stefania Ferraro
- Neuroradiology UnitFondazione IRCCS Istituto Neurologico Carlo BestaMilanItaly
- School of Life Science and Technology, MOE Key Laboratory for NeuroinformationUniversity of Electronic Science and Technology of ChinaChengduChina
| | | | - Sara Palermo
- Neuroradiology UnitFondazione IRCCS Istituto Neurologico Carlo BestaMilanItaly
| | - Laura Lequio
- Neuroradiology UnitCTO Hospital, AOU Città della Salute e della Scienza di TorinoTurinItaly
| | - Federica Denegri
- Neuroradiology UnitCTO Hospital, AOU Città della Salute e della Scienza di TorinoTurinItaly
| | - Federica Agosta
- Neuroimaging Research Unit, Division of NeuroscienceIRCCS San Raffaele Scientific InstituteMilanItaly
- Neurology UnitIRCCS San Raffaele Scientific InstituteMilanItaly
- Vita‐Salute San Raffaele UniversityMilanItaly
| | - Edoardo Gioele Spinelli
- Neuroimaging Research Unit, Division of NeuroscienceIRCCS San Raffaele Scientific InstituteMilanItaly
- Neurology UnitIRCCS San Raffaele Scientific InstituteMilanItaly
- Neurorehabilitation UnitIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Massimo Filippi
- Neuroimaging Research Unit, Division of NeuroscienceIRCCS San Raffaele Scientific InstituteMilanItaly
- Neurology UnitIRCCS San Raffaele Scientific InstituteMilanItaly
- Vita‐Salute San Raffaele UniversityMilanItaly
- Neurorehabilitation UnitIRCCS San Raffaele Scientific InstituteMilanItaly
- Neurophysiology ServiceIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Marina Grisoli
- Neuroradiology UnitFondazione IRCCS Istituto Neurologico Carlo BestaMilanItaly
| | | | - Filippo De Mattei
- ALS Centre, “Rita Levi Montalcini” Department of NeuroscienceUniversity of TurinTurinItaly
- Azienda Ospedaliero‐Universitaria Città della Salute e della Scienza di Torino, SC Neurologia 1UTurinItaly
| | - Antonio Canosa
- ALS Centre, “Rita Levi Montalcini” Department of NeuroscienceUniversity of TurinTurinItaly
- Azienda Ospedaliero‐Universitaria Città della Salute e della Scienza di Torino, SC Neurologia 1UTurinItaly
| | - Andrea Calvo
- ALS Centre, “Rita Levi Montalcini” Department of NeuroscienceUniversity of TurinTurinItaly
- Azienda Ospedaliero‐Universitaria Città della Salute e della Scienza di Torino, SC Neurologia 1UTurinItaly
| | - Adriano Chiò
- ALS Centre, “Rita Levi Montalcini” Department of NeuroscienceUniversity of TurinTurinItaly
- Azienda Ospedaliero‐Universitaria Città della Salute e della Scienza di Torino, SC Neurologia 1UTurinItaly
- Institute of Cognitive Sciences and TechnologiesNational Council of ResearchRomeItaly
| | | | - Cristina Moglia
- ALS Centre, “Rita Levi Montalcini” Department of NeuroscienceUniversity of TurinTurinItaly
- Azienda Ospedaliero‐Universitaria Città della Salute e della Scienza di Torino, SC Neurologia 1UTurinItaly
| |
Collapse
|
6
|
Shen T, Vogel JW, Duda J, Phillips JS, Cook PA, Gee J, Elman L, Quinn C, Amado DA, Baer M, Massimo L, Grossman M, Irwin DJ, McMillan CT. Novel data-driven subtypes and stages of brain atrophy in the ALS-FTD spectrum. Transl Neurodegener 2023; 12:57. [PMID: 38062485 PMCID: PMC10701950 DOI: 10.1186/s40035-023-00389-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 11/15/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND TDP-43 proteinopathies represent a spectrum of neurological disorders, anchored clinically on either end by amyotrophic lateral sclerosis (ALS) and frontotemporal degeneration (FTD). The ALS-FTD spectrum exhibits a diverse range of clinical presentations with overlapping phenotypes, highlighting its heterogeneity. This study was aimed to use disease progression modeling to identify novel data-driven spatial and temporal subtypes of brain atrophy and its progression in the ALS-FTD spectrum. METHODS We used a data-driven procedure to identify 13 anatomic clusters of brain volume for 57 behavioral variant FTD (bvFTD; with either autopsy-confirmed TDP-43 or TDP-43 proteinopathy-associated genetic variants), 103 ALS, and 47 ALS-FTD patients with likely TDP-43. A Subtype and Stage Inference (SuStaIn) model was trained to identify subtypes of individuals along the ALS-FTD spectrum with distinct brain atrophy patterns, and we related subtypes and stages to clinical, genetic, and neuropathological features of disease. RESULTS SuStaIn identified three novel subtypes: two disease subtypes with predominant brain atrophy in either prefrontal/somatomotor regions or limbic-related regions, and a normal-appearing group without obvious brain atrophy. The limbic-predominant subtype tended to present with more impaired cognition, higher frequencies of pathogenic variants in TBK1 and TARDBP genes, and a higher proportion of TDP-43 types B, E and C. In contrast, the prefrontal/somatomotor-predominant subtype had higher frequencies of pathogenic variants in C9orf72 and GRN genes and higher proportion of TDP-43 type A. The normal-appearing brain group showed higher frequency of ALS relative to ALS-FTD and bvFTD patients, higher cognitive capacity, higher proportion of lower motor neuron onset, milder motor symptoms, and lower frequencies of genetic pathogenic variants. The overall SuStaIn stages also correlated with evidence for clinical progression including longer disease duration, higher King's stage, and cognitive decline. Additionally, SuStaIn stages differed across clinical phenotypes, genotypes and types of TDP-43 pathology. CONCLUSIONS Our findings suggest distinct neurodegenerative subtypes of disease along the ALS-FTD spectrum that can be identified in vivo, each with distinct brain atrophy, clinical, genetic and pathological patterns.
Collapse
Affiliation(s)
- Ting Shen
- Penn Frontotemporal Degeneration Center, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jacob W Vogel
- Department of Clinical Sciences, SciLifeLab, Lund University, 222 42, Lund, Sweden
| | - Jeffrey Duda
- Penn Image Computing and Science Lab (PICSL), Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jeffrey S Phillips
- Penn Frontotemporal Degeneration Center, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Philip A Cook
- Penn Image Computing and Science Lab (PICSL), Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - James Gee
- Penn Image Computing and Science Lab (PICSL), Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Lauren Elman
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Colin Quinn
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Defne A Amado
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Michael Baer
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Lauren Massimo
- Penn Frontotemporal Degeneration Center, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Murray Grossman
- Penn Frontotemporal Degeneration Center, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - David J Irwin
- Penn Frontotemporal Degeneration Center, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Digital Neuropathology Laboratory, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Corey T McMillan
- Penn Frontotemporal Degeneration Center, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
7
|
White LM, Boardman J, Lilleker J, Chaouch A, Kargwell H, Ealing J, Hamdalla H. Phenotypical differences of C9ORF72 gene-positive and negative amyotrophic lateral sclerosis: a comparative case series. J Med Genet 2023; 60:1016-1020. [PMID: 37173134 DOI: 10.1136/jmg-2022-109016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 05/01/2023] [Indexed: 05/15/2023]
Abstract
BACKGROUND Hexanucleotide repeat expansions of C9ORF72 account for a significant proportion of autosomal dominant neurodegenerative diseases in the amyotrophic lateral sclerosis (ALS)-frontotemporal dementia spectrum. In the absence of a family history, clinical identification of such patients remains difficult. We aimed to identify differences in demographics and clinical presentation between patients with C9ORF72 gene-positive ALS (C9pALS) versus C9ORF72 gene-negative ALS (C9nALS), to aid identification of these patients in the clinic and examine differences in outcomes including survival. METHODS We retrospectively reviewed the clinical presentations of 32 patients with C9pALS and compared their characteristics with a cohort of 46 patients with C9nALS from the same tertiary neurosciences centre. RESULTS Patients with C9pALS more commonly presented with mixed upper and lower motor signs (C9pALS 87.5%, C9nALS 65.2%; p=0.0352), but less frequently presented with purely upper motor neuron signs (C9pALS 3.1%, C9nALS 21.7%; p=0.0226). The C9pALS cohort had a higher frequency of cognitive impairment (C9pALS 31.3%, C9nALS 10.9%; p=0.0394) and bulbar disease (C9pALS 56.3%, C9nALS 28.3%; p=0.0186). There were no differences between cohorts in age at diagnosis, gender, limb weakness, respiratory symptoms, presentation with predominantly lower motor neuron signs or overall survival. DISCUSSION Analysis of this ALS clinic cohort at a UK tertiary neurosciences centre adds to the small but growing understanding of the unique clinical features of patients with C9pALS. In the age of precision medicine with expanding opportunities to manage genetic diseases with disease-modifying therapies, clinical identification of such patients is increasingly important as focused therapeutic strategies become available.
Collapse
Affiliation(s)
- Laura Michelle White
- Manchester Centre for Clinical Neurosciences, Salford Royal Hospital, Salford, UK
| | | | - James Lilleker
- Manchester Centre for Clinical Neurosciences, Salford Royal Hospital, Salford, UK
- Centre for Musculoskeletal Research, The University of Manchester School of Biological Sciences, Manchester, UK
| | - Amina Chaouch
- Manchester Centre for Clinical Neurosciences, Salford Royal Hospital, Salford, UK
| | - Haga Kargwell
- Manchester Centre for Clinical Neurosciences, Salford Royal Hospital, Salford, UK
| | - John Ealing
- Manchester Centre for Clinical Neurosciences, Salford Royal Hospital, Salford, UK
| | - Hisham Hamdalla
- Manchester Centre for Clinical Neurosciences, Salford Royal Hospital, Salford, UK
| |
Collapse
|
8
|
Jellinger KA. The Spectrum of Cognitive Dysfunction in Amyotrophic Lateral Sclerosis: An Update. Int J Mol Sci 2023; 24:14647. [PMID: 37834094 PMCID: PMC10572320 DOI: 10.3390/ijms241914647] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/21/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
Cognitive dysfunction is an important non-motor symptom in amyotrophic lateral sclerosis (ALS) that has a negative impact on survival and caregiver burden. It shows a wide spectrum ranging from subjective cognitive decline to frontotemporal dementia (FTD) and covers various cognitive domains, mainly executive/attention, language and verbal memory deficits. The frequency of cognitive impairment across the different ALS phenotypes ranges from 30% to 75%, with up to 45% fulfilling the criteria of FTD. Significant genetic, clinical, and pathological heterogeneity reflects deficits in various cognitive domains. Modern neuroimaging studies revealed frontotemporal degeneration and widespread involvement of limbic and white matter systems, with hypometabolism of the relevant areas. Morphological substrates are frontotemporal and hippocampal atrophy with synaptic loss, associated with TDP-43 and other co-pathologies, including tau deposition. Widespread functional disruptions of motor and extramotor networks, as well as of frontoparietal, frontostriatal and other connectivities, are markers for cognitive deficits in ALS. Cognitive reserve may moderate the effect of brain damage but is not protective against cognitive decline. The natural history of cognitive dysfunction in ALS and its relationship to FTD are not fully understood, although there is an overlap between the ALS variants and ALS-related frontotemporal syndromes, suggesting a differential vulnerability of motor and non-motor networks. An assessment of risks or the early detection of brain connectivity signatures before structural changes may be helpful in investigating the pathophysiological mechanisms of cognitive impairment in ALS, which might even serve as novel targets for effective disease-modifying therapies.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, A-1150 Vienna, Austria
| |
Collapse
|
9
|
Shen T, Vogel JW, Duda J, Phillips JS, Cook PA, Gee J, Elman L, Quinn C, Amado DA, Baer M, Massimo L, Grossman M, Irwin DJ, McMillan CT. Novel data-driven subtypes and stages of brain atrophy in the ALS-FTD spectrum. RESEARCH SQUARE 2023:rs.3.rs-3183113. [PMID: 37609205 PMCID: PMC10441467 DOI: 10.21203/rs.3.rs-3183113/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Background TDP-43 proteinopathies represents a spectrum of neurological disorders, anchored clinically on either end by amyotrophic lateral sclerosis (ALS) and frontotemporal degeneration (FTD). The ALS-FTD spectrum exhibits a diverse range of clinical presentations with overlapping phenotypes, highlighting its heterogeneity. This study aimed to use disease progression modeling to identify novel data-driven spatial and temporal subtypes of brain atrophy and its progression in the ALS-FTD spectrum. Methods We used a data-driven procedure to identify 13 anatomic clusters of brain volumes for 57 behavioral variant FTD (bvFTD; with either autopsy-confirmed TDP-43 or TDP-43 proteinopathy-associated genetic variants), 103 ALS, and 47 ALS-FTD patients with likely TDP-43. A Subtype and Stage Inference (SuStaIn) model was trained to identify subtypes of individuals along the ALS-FTD spectrum with distinct brain atrophy patterns, and we related subtypes and stages to clinical, genetic, and neuropathological features of disease. Results SuStaIn identified three novel subtypes: two disease subtypes with predominant brain atrophy either in prefrontal/somatomotor regions or limbic-related regions, and a normal-appearing group without obvious brain atrophy. The Limbic-predominant subtype tended to present with more impaired cognition, higher frequencies of pathogenic variants in TBK1 and TARDBP genes, and a higher proportion of TDP-43 type B, E and C. In contrast, the Prefrontal/Somatomotor-predominant subtype had higher frequencies of pathogenic variants in C9orf72 and GRN genes and higher proportion of TDP-43 type A. The normal-appearing brain group showed higher frequency of ALS relative to ALS-FTD and bvFTD patients, higher cognitive capacity, higher proportion of lower motor neuron onset, milder motor symptoms, and lower frequencies of genetic pathogenic variants. Overall SuStaIn stages also correlated with evidence for clinical progression including longer disease duration, higher King's stage, and cognitive decline. Additionally, SuStaIn stages differed across clinical phenotypes, genotypes and types of TDP-43 pathology. Conclusions Our findings suggest distinct neurodegenerative subtypes of disease along the ALS-FTD spectrum that can be identified in vivo, each with distinct brain atrophy, clinical, genetic and pathological patterns.
Collapse
Affiliation(s)
- Ting Shen
- University of Pennsylvania Perelman School of Medicine
| | | | - Jeffrey Duda
- University of Pennsylvania Perelman School of Medicine
| | | | - Philip A Cook
- University of Pennsylvania Perelman School of Medicine
| | - James Gee
- University of Pennsylvania Perelman School of Medicine
| | - Lauren Elman
- University of Pennsylvania Perelman School of Medicine
| | - Colin Quinn
- University of Pennsylvania Perelman School of Medicine
| | - Defne A Amado
- University of Pennsylvania Perelman School of Medicine
| | - Michael Baer
- University of Pennsylvania Perelman School of Medicine
| | | | | | - David J Irwin
- University of Pennsylvania Perelman School of Medicine
| | | |
Collapse
|
10
|
De Vocht J, Van Weehaeghe D, Ombelet F, Masrori P, Lamaire N, Devrome M, Van Esch H, Moisse M, Koole M, Dupont P, Van Laere K, Van Damme P. Differences in Cerebral Glucose Metabolism in ALS Patients with and without C9orf72 and SOD1 Mutations. Cells 2023; 12:cells12060933. [PMID: 36980274 PMCID: PMC10047407 DOI: 10.3390/cells12060933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/22/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is characterized by progressive loss of upper and lower motor neurons. In 10% of patients, the disorder runs in the family. Our aim was to study the impact of ALS-causing gene mutations on cerebral glucose metabolism. Between October 2010 and October 2022, 538 patients underwent genetic testing for mutations with strong evidence of causality for ALS and 18F-2-fluoro-2-deoxy-D-glucose-PET (FDG PET), at University Hospitals Leuven. We identified 48 C9orf72-ALS and 22 SOD1-ALS patients. After propensity score matching, two cohorts of 48 and 21 matched sporadic ALS patients, as well as 20 healthy controls were included. FDG PET images were assessed using a voxel-based and volume-of-interest approach. We observed widespread frontotemporal involvement in all ALS groups, in comparison to healthy controls. The degree of relative glucose metabolism in SOD1-ALS in motor and extra-motor regions did not differ significantly from matched sporadic ALS patients. In C9orf72-ALS, we found more pronounced hypometabolism in the peri-rolandic region and thalamus, and hypermetabolism in the medulla extending to the pons, in comparison to matched sporadic ALS patients. Our study revealed C9orf72-dependent differences in glucose metabolism in the peri-rolandic region, thalamus, and brainstem (i.e., medulla, extending to the pons) in relation to matched sporadic ALS patients.
Collapse
Affiliation(s)
- Joke De Vocht
- Division of Psychiatry, Division of Neurology, University Hospitals Leuven, VIB-KULeuven Center for Brain & Disease Research, Laboratory of Neurobiology, Department of Neurosciences, Leuven Brain Institute (LBI), Katholieke Universiteit Leuven, 3000 Leuven, Belgium
- Correspondence: ; Tel.: +32-16-34-13-73
| | | | - Fouke Ombelet
- Division of Neurology, University Hospitals Leuven, VIB-KULeuven Center for Brain & Disease Research, Laboratory of Neurobiology, Department of Neurosciences, Leuven Brain Institute (LBI), Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Pegah Masrori
- Division of Neurology, University Hospitals Leuven, VIB-KULeuven Center for Brain & Disease Research, Laboratory of Neurobiology, Department of Neurosciences, Leuven Brain Institute (LBI), Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Nikita Lamaire
- Division of Neurology, University Hospitals Leuven, VIB-KULeuven Center for Brain & Disease Research, Laboratory of Neurobiology, Department of Neurosciences, Leuven Brain Institute (LBI), Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Martijn Devrome
- Department of Imaging and Pathology, Nuclear Medicine and Molecular Imaging, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Hilde Van Esch
- Center for Human Genetics, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Mathieu Moisse
- VIB-KU Leuven Center for Brain & Disease Research, Laboratory of Neurobiology, Department of Neurosciences, Leuven Brain Institute (LBI), Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Michel Koole
- Department of Imaging and Pathology, Nuclear Medicine and Molecular Imaging, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Patrick Dupont
- Laboratory of Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute (LBI), Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Koen Van Laere
- Department of Imaging and Pathology, Nuclear Medicine and Molecular Imaging, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Philip Van Damme
- Division of Neurology, University Hospitals Leuven, VIB-KULeuven Center for Brain & Disease Research, Laboratory of Neurobiology, Department of Neurosciences, Leuven Brain Institute (LBI), Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| |
Collapse
|
11
|
Motor, cognitive and behavioural profiles of C9orf72 expansion-related amyotrophic lateral sclerosis. J Neurol 2023; 270:898-908. [PMID: 36308529 PMCID: PMC9886586 DOI: 10.1007/s00415-022-11433-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/16/2022] [Accepted: 10/17/2022] [Indexed: 02/03/2023]
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) individuals carrying the hexanucleotide repeat expansion (HRE) in the C9orf72 gene (C9Pos) have been described as presenting distinct features compared to the general ALS population (C9Neg). We aim to identify the phenotypic traits more closely associated with the HRE and analyse the role of the repeat length as a modifier factor. METHODS We studied a cohort of 960 ALS patients (101 familial and 859 sporadic cases). Motor phenotype was determined using the MRC scale, the lower motor neuron score (LMNS) and the Penn upper motor neuron score (PUMNS). Neuropsychological profile was studied using the Italian version of the Edinburgh Cognitive and Behavioral ALS Screen (ECAS), the Frontal Behavioral Inventory (FBI), the Beck Depression Inventory-II (BDI-II) and the State-Trait Anxiety Inventory (STAI). A two-step PCR protocol and Southern blotting were performed to determine the presence and the size of C9orf72 HRE, respectively. RESULTS C9orf72 HRE was detected in 55/960 ALS patients. C9Pos patients showed a younger onset, higher odds of bulbar onset, increased burden of UMN signs, reduced survival and higher frequency of concurrent dementia. We found an inverse correlation between the HRE length and the performance at ECAS ALS-specific tasks (P = 0.031). Patients also showed higher burden of behavioural disinhibition (P = 1.6 × 10-4), lower degrees of depression (P = 0.015) and anxiety (P = 0.008) compared to C9Neg cases. CONCLUSIONS Our study provides an extensive characterization of motor, cognitive and behavioural features of C9orf72-related ALS, indicating that the C9orf72 HRE size may represent a modifier of the cognitive phenotype.
Collapse
|
12
|
Factors predicting disease progression in C9ORF72 ALS patients. J Neurol 2023; 270:877-890. [PMID: 36280624 DOI: 10.1007/s00415-022-11426-y] [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: 07/13/2022] [Revised: 10/10/2022] [Accepted: 10/13/2022] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To unveil clinical features, comorbidities, disease progression and prognostic factors in a population-based cohort of ALS patients carrying C9ORF72 expansion (C9 + ALS). METHODS This is a retrospective observational study on ALS patients residing in Emilia Romagna and Piedmont-Valle D'Aosta regions whose data are available through population based registers. We analysed patients who underwent genetic testing, focusing on C9 + ALS subgroup. RESULTS Among 2204 genotyped patients of the two registers, 150 were C9 + ALS. In comparison with patients without mutation, a higher proportion of family history (12.85 vs 68%, p < 0.001) and frontotemporal dementia (3.93% vs 10.67%, p < 0.001) was detected in C9 + ALS. C9 + ALS presented a faster disease progression as measured by monthly decline in ALS Functional Rating Scale-Revised (1.86 ± 3.30 vs 1.45 ± 2.35, p < 0.01) and in forced vital capacity (5.90 ± 5.24 vs 2.97 ± 3.47, p < 0.01), a shorter diagnostic delay (8.93 ± 6.74 vs 12.68 ± 12.86 months, p < 0.01) and earlier onset (58.91 ± 9.02 vs 65.04 ± 11.55 years, p < 0.01). Consistently, they reached death or tracheostomy earlier than other patients (31 vs 37 months, HR = 1.52, 95% C.I. 1.27-1.82, p < 0.001). With respect to other genotyped patients, C9 + ALS patients did not present a significantly higher prevalence of concomitant diseases. Independent prognostic factors of survival of C9 + ALS included sex, age, progression rate, presence of frontotemporal dementia and thyroid disorders, with the latter being associated with prolonged ALS survival (43 vs 29 months, HR = 0.42, 95% C.I. 0.24-0.74, p = 0.003). CONCLUSION Even in the context of a more aggressive disease, C9 + ALS had a longer survival in presence of thyroid disorders. This finding may suggest protective pathogenic pathways in C9 + ALS to be explored, looking for therapeutic strategies to slow disease course.
Collapse
|
13
|
Yildiz O, Schroth J, Tree T, Turner MR, Shaw PJ, Henson SM, Malaspina A. Senescent-like Blood Lymphocytes and Disease Progression in Amyotrophic Lateral Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:e200042. [PMID: 36323511 PMCID: PMC9673751 DOI: 10.1212/nxi.0000000000200042] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 08/22/2022] [Indexed: 03/11/2023]
Abstract
BACKGROUND AND OBJECTIVES Aging is known to exacerbate neuroinflammation, and in the neurodegenerative disorder amyotrophic lateral sclerosis (ALS), an older age is associated with a worse prognosis. We have previously shown the activation of cell senescence pathways in the proteome of peripheral blood mononuclear cells and the increase of proinflammatory cytokines in blood from individuals living with ALS. In this single-center, retrospective study, we investigated the expression of senescent-like blood mononuclear cells in ALS. METHODS We first applied multidimensional cytometry by time-of-flight (CyTOF) to study the senescent immunophenotype of blood mononuclear cells from 21 patients with ALS and 10 healthy controls (HCs). We then used targeted flow cytometry (FC) to investigate frequencies of senescent blood lymphocytes in 40 patients with ALS and 20 HCs. Longitudinal analysis included 2 additional time points in 17 patients with ALS. Frequencies of senescent-like lymphocytes were analyzed in relation to survival. RESULTS Unsupervised clustering of CyTOF data showed higher frequencies of senescent CD4+CD27-CD57+ T cells in patients with ALS compared with those in HCs (p = 0.0017, false discovery (FDR)-adjusted p = 0.029). Moderate to strong negative correlations were identified between CD4 T central memory-cell frequencies and survival (R = -061, p = 0.01; FDR-adjusted p < 0.1) and between CD95 CD8 cells and ALS functional rating scale revised at baseline (R = -0.72, p = 0.001; FDR-adjusted p < 0.1).Targeted FC analysis showed higher memory T regulatory cells (p = 0.0052) and memory CD8+ T cell (M-Tc; p = 0.0006) in bulbar ALS (A-B) compared with those in limb ALS (A-L), while late memory B cells (LM-B) were also elevated in A-B and fast-progressing ALS (p = 0.0059). Higher M-Tc levels separated A-B from A-L (AUC: 0.887; p < 0.0001). A linear regression model with prespecified clinical independent variables and neurofilament light chain plasma concentration showed that higher frequencies of LM-B predicted a shorter survival (hazard ratio: 1.094, CI: 1.026-1.167; p = 0.006). DISCUSSION Our data suggest that a systemic elevation of senescent and late memory T and B lymphocytes is a feature of faster progressing ALS and of ALS individuals with bulbar involvement. Lymphocyte senescence and their memory state may be central to the immune dysregulation known to drive disease progression in ALS and a target for biomarkers and therapeutics discovery.
Collapse
Affiliation(s)
- Ozlem Yildiz
- From the Neuroscience and Trauma Centre (O.Y., A.M.), Blizard Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London; Queen Square Motor Neuron Disease Centre (A.M.), Neuromuscular Department, Institute of Neurology, University College London; Translational Medicine and Therapeutics (J.S., S.M.H.), William Harvey Research Institute, Barts and the London, Queen Mary University of London; Department of Immunobiology (T.T.), School of Immunology & Microbial Sciences, King's College London; Nuffield Department of Clinical Neurosciences (M.R.T.), University of Oxford; and Sheffield Institute for Translational Neuroscience (P.J.S.), University of Sheffield, UK
| | - Johannes Schroth
- From the Neuroscience and Trauma Centre (O.Y., A.M.), Blizard Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London; Queen Square Motor Neuron Disease Centre (A.M.), Neuromuscular Department, Institute of Neurology, University College London; Translational Medicine and Therapeutics (J.S., S.M.H.), William Harvey Research Institute, Barts and the London, Queen Mary University of London; Department of Immunobiology (T.T.), School of Immunology & Microbial Sciences, King's College London; Nuffield Department of Clinical Neurosciences (M.R.T.), University of Oxford; and Sheffield Institute for Translational Neuroscience (P.J.S.), University of Sheffield, UK
| | - Timothy Tree
- From the Neuroscience and Trauma Centre (O.Y., A.M.), Blizard Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London; Queen Square Motor Neuron Disease Centre (A.M.), Neuromuscular Department, Institute of Neurology, University College London; Translational Medicine and Therapeutics (J.S., S.M.H.), William Harvey Research Institute, Barts and the London, Queen Mary University of London; Department of Immunobiology (T.T.), School of Immunology & Microbial Sciences, King's College London; Nuffield Department of Clinical Neurosciences (M.R.T.), University of Oxford; and Sheffield Institute for Translational Neuroscience (P.J.S.), University of Sheffield, UK
| | - Martin R Turner
- From the Neuroscience and Trauma Centre (O.Y., A.M.), Blizard Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London; Queen Square Motor Neuron Disease Centre (A.M.), Neuromuscular Department, Institute of Neurology, University College London; Translational Medicine and Therapeutics (J.S., S.M.H.), William Harvey Research Institute, Barts and the London, Queen Mary University of London; Department of Immunobiology (T.T.), School of Immunology & Microbial Sciences, King's College London; Nuffield Department of Clinical Neurosciences (M.R.T.), University of Oxford; and Sheffield Institute for Translational Neuroscience (P.J.S.), University of Sheffield, UK
| | - Pamela J Shaw
- From the Neuroscience and Trauma Centre (O.Y., A.M.), Blizard Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London; Queen Square Motor Neuron Disease Centre (A.M.), Neuromuscular Department, Institute of Neurology, University College London; Translational Medicine and Therapeutics (J.S., S.M.H.), William Harvey Research Institute, Barts and the London, Queen Mary University of London; Department of Immunobiology (T.T.), School of Immunology & Microbial Sciences, King's College London; Nuffield Department of Clinical Neurosciences (M.R.T.), University of Oxford; and Sheffield Institute for Translational Neuroscience (P.J.S.), University of Sheffield, UK
| | - Sian M Henson
- From the Neuroscience and Trauma Centre (O.Y., A.M.), Blizard Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London; Queen Square Motor Neuron Disease Centre (A.M.), Neuromuscular Department, Institute of Neurology, University College London; Translational Medicine and Therapeutics (J.S., S.M.H.), William Harvey Research Institute, Barts and the London, Queen Mary University of London; Department of Immunobiology (T.T.), School of Immunology & Microbial Sciences, King's College London; Nuffield Department of Clinical Neurosciences (M.R.T.), University of Oxford; and Sheffield Institute for Translational Neuroscience (P.J.S.), University of Sheffield, UK
| | - Andrea Malaspina
- From the Neuroscience and Trauma Centre (O.Y., A.M.), Blizard Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London; Queen Square Motor Neuron Disease Centre (A.M.), Neuromuscular Department, Institute of Neurology, University College London; Translational Medicine and Therapeutics (J.S., S.M.H.), William Harvey Research Institute, Barts and the London, Queen Mary University of London; Department of Immunobiology (T.T.), School of Immunology & Microbial Sciences, King's College London; Nuffield Department of Clinical Neurosciences (M.R.T.), University of Oxford; and Sheffield Institute for Translational Neuroscience (P.J.S.), University of Sheffield, UK.
| |
Collapse
|
14
|
Zecca C, Tortelli R, Carrera P, Dell'Abate MT, Logroscino G, Ferrari M. Genotype-phenotype correlation in the spectrum of frontotemporal dementia-parkinsonian syndromes and advanced diagnostic approaches. Crit Rev Clin Lab Sci 2022; 60:171-188. [PMID: 36510705 DOI: 10.1080/10408363.2022.2150833] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The term frontotemporal dementia (FTD) refers to a group of progressive neurodegenerative disorders characterized mainly by atrophy of the frontal and anterior temporal lobes. Based on clinical presentation, three main clinical syndromes have traditionally been described: behavioral variant frontotemporal dementia (bvFTD), non-fluent/agrammatic primary progressive aphasia (nfPPA), and semantic variant PPA (svPPA). However, over the last 20 years, it has been recognized that cognitive phenotypes often overlap with motor phenotypes, either motor neuron diseases or parkinsonian signs and/or syndromes like progressive supranuclear palsy (PSP) and cortico-basal syndrome (CBS). Furthermore, FTD-related genes are characterized by genetic pleiotropy and can cause, even in the same family, pure motor phenotypes, findings that underlie the clinical continuum of the spectrum, which has pure cognitive and pure motor phenotypes as the extremes. The genotype-phenotype correlation of the spectrum, FTD-motor neuron disease, has been well defined and extensively investigated, while the continuum, FTD-parkinsonism, lacks a comprehensive review. In this narrative review, we describe the current knowledge about the genotype-phenotype correlation of the spectrum, FTD-parkinsonism, focusing on the phenotypes that are less frequent than bvFTD, namely nfPPA, svPPA, PSP, CBS, and cognitive-motor overlapping phenotypes (i.e. PPA + PSP). From a pathological point of view, they are characterized mainly by the presence of phosphorylated-tau inclusions, either 4 R or 3 R. The genetic correlate of the spectrum can be heterogeneous, although some variants seem to lead preferentially to specific clinical syndromes. Furthermore, we critically review the contribution of genome-wide association studies (GWAS) and next-generation sequencing (NGS) in disentangling the complex heritability of the FTD-parkinsonism spectrum and in defining the genotype-phenotype correlation of the entire clinical scenario, owing to the ability of these techniques to test multiple genes, and so to allow detailed investigations of the overlapping phenotypes. Finally, we conclude with the importance of a detailed genetic characterization and we offer to patients and families the chance to be included in future randomized clinical trials focused on autosomal dominant forms of FTLD.
Collapse
Affiliation(s)
- Chiara Zecca
- Department of Clinical Research in Neurology, Center for Neurodegenerative Diseases and the Aging Brain, University of Bari "Aldo Moro", Pia Fondazione Card G. Panico Hospital, Tricase, Italy
| | - Rosanna Tortelli
- Neuroscience and Rare Diseases Discovery and Translational Area, Roche Pharma Research and Early Development, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Paola Carrera
- Unit of Genomics for Human Disease Diagnosis and Clinical Molecular Biology Laboratory, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria Teresa Dell'Abate
- Department of Clinical Research in Neurology, Center for Neurodegenerative Diseases and the Aging Brain, University of Bari "Aldo Moro", Pia Fondazione Card G. Panico Hospital, Tricase, Italy
| | - Giancarlo Logroscino
- Department of Clinical Research in Neurology, Center for Neurodegenerative Diseases and the Aging Brain, University of Bari "Aldo Moro", Pia Fondazione Card G. Panico Hospital, Tricase, Italy.,Department of Basic Medicine Sciences, Neuroscience, and Sense Organs, University of Bari "Aldo Moro", Bari, Italy
| | | |
Collapse
|
15
|
Carvalho TA, Vasconcelos MMDA, de Bessa J, Bastos JM, Dutra MF, Guimarães ICDO, Lima EM, Silva ACSE, Mrad FCDC. Relationship between primary monosymptomatic enuresis and process toilet training: a case-control. Int Braz J Urol 2022; 48:944-951. [PMID: 36173406 PMCID: PMC9747032 DOI: 10.1590/s1677-5538.ibju.2022.0381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 09/05/2022] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE Primary monosymptomatic nocturnal enuresis (PMNE) is a prevalent condition in childhood, and the pathophysiology is multifactorial. This study investigated the relationship between the toilet training process (TT) and PMNE in children and adolescents. PATIENTS AND METHODS A case-control study was carried out from 2015 to 2020. The presence of PMNE was identified according to International Children's Continence Society criteria. A semi-structured questionnaire was applied to assess TT. RESULTS The study included 103 children and adolescents with PMNE and 269 participants with normal psychomotor development without PMNE (control group [CG]). Readiness signals were more remembered and less frequent in participants with PMNE (p=0.001) when compared to control group. No differences were found between the groups regarding the onset age of the daytime TT (p= 0.10), the nocturnal TT (p=0.08), the acquisition of daytime continence (p=0.06), and the type of equipment used for the TT (p=0.99). The use of Child-Oriented approach in group of children with enuresis was lower than in controls [87.4% (90/103) versus 94% (250/266)], respectively (OR= 0.44, 95% CI 0.21-0.94, p = 0.039). CONCLUSIONS The age of onset of TT, acquisition of daytime continence, and the type of equipment were not associated with higher occurrence of PMNE. On the other hand, the Child-Oriented approach was a protective factor for the occurrence of PMNE.
Collapse
Affiliation(s)
- Tânia Antunes Carvalho
- Universidade Federal de Minas GeraisFaculdade de MedicinaDepartamento de PediatriaBelo HorizonteMGBrasilDepartamento de Pediatria, Unidade de Nefrologia Pediátrica, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brasil
| | - Mônica Maria de Almeida Vasconcelos
- Universidade Federal de Minas GeraisFaculdade de MedicinaDepartamento de PediatriaBelo HorizonteMGBrasilDepartamento de Pediatria, Unidade de Nefrologia Pediátrica, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brasil
| | - José de Bessa
- Universidade Estadual de Feira de SantanaDepartamento de UrologiaFeira de SantanaBABrasilDepartamento de Urologia, Universidade Estadual de Feira de Santana (UFFS), Feira de Santana, BA, Brasil
| | - José Murillo Bastos
- Universidade Federal de Juiz de ForaFaculdade de MedicinaDepartamento de UrologiaJuiz de ForaMGBrasilDepartamento de Urologia, Faculdade de Medicina, Universidade Federal de Juiz de Fora (UFJF), Juiz de Fora, MG, Brasil,Faculdade de Ciências Médicas de Juiz de ForaMaternidade Therezinha de JesusDepartamento de UrologiaJuiz de ForaMGBrasilDepartamento de Urologia, Maternidade Therezinha de Jesus, Faculdade de Ciências Médicas de Juiz de Fora, Juiz de Fora, MG, Brasil
| | - Melissa Faria Dutra
- Universidade Federal de Minas GeraisFaculdade de MedicinaDepartamento de PediatriaBelo HorizonteMGBrasilDepartamento de Pediatria, Unidade de Nefrologia Pediátrica, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brasil
| | - Isabela Cristina de Oliveira Guimarães
- Universidade Federal de Minas GeraisFaculdade de MedicinaDepartamento de PediatriaBelo HorizonteMGBrasilDepartamento de Pediatria, Unidade de Nefrologia Pediátrica, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brasil
| | - Eleonora Moreira Lima
- Universidade Federal de Minas GeraisFaculdade de MedicinaDepartamento de PediatriaBelo HorizonteMGBrasilDepartamento de Pediatria, Unidade de Nefrologia Pediátrica, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brasil
| | - Ana Cristina Simões e Silva
- Universidade Federal de Minas GeraisFaculdade de MedicinaDepartamento de PediatriaBelo HorizonteMGBrasilDepartamento de Pediatria, Unidade de Nefrologia Pediátrica, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brasil,Universidade Federal de Minas GeraisLaboratório Interdisciplinar de Investigação MédicaBelo HorizonteMGBrasilLaboratório Interdisciplinar de Investigação Médica, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Flávia Cristina de Carvalho Mrad
- Universidade Federal de Minas GeraisFaculdade de MedicinaDepartamento de PediatriaBelo HorizonteMGBrasilDepartamento de Pediatria, Unidade de Nefrologia Pediátrica, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brasil,Correspondence address: Flávia Cristina de Carvalho Mrad, MD Depart. de Pediatria, Unidade de Nefrologia Pediátrica, Fac. de Med., Univ. Federal de Minas Gerais (UFMG) Avenida Alfredo Balena 190. sala 267, Santa Efigênia Belo Horizonte, MG, 30130-100, Brasil E-mail:
| |
Collapse
|
16
|
Ramamoorthy D, Severson K, Ghosh S, Sachs K, Glass JD, Fournier CN, Herrington TM, Berry JD, Ng K, Fraenkel E. Identifying patterns in amyotrophic lateral sclerosis progression from sparse longitudinal data. NATURE COMPUTATIONAL SCIENCE 2022; 2:605-616. [PMID: 38177466 PMCID: PMC10766562 DOI: 10.1038/s43588-022-00299-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 07/14/2022] [Indexed: 01/06/2024]
Abstract
The clinical presentation of amyotrophic lateral sclerosis (ALS), a fatal neurodegenerative disease, varies widely across patients, making it challenging to determine if potential therapeutics slow progression. We sought to determine whether there were common patterns of disease progression that could aid in the design and analysis of clinical trials. We developed an approach based on a mixture of Gaussian processes to identify clusters of patients sharing similar disease progression patterns, modeling their average trajectories and the variability in each cluster. We show that ALS progression is frequently nonlinear, with periods of stable disease preceded or followed by rapid decline. We also show that our approach can be extended to Alzheimer's and Parkinson's diseases. Our results advance the characterization of disease progression of ALS and provide a flexible modeling approach that can be applied to other progressive diseases.
Collapse
Affiliation(s)
| | - Kristen Severson
- Center for Computational Health and MIT-IBM Watson AI Lab, IBM Research, Cambridge, MA, USA
| | - Soumya Ghosh
- Center for Computational Health and MIT-IBM Watson AI Lab, IBM Research, Cambridge, MA, USA
| | - Karen Sachs
- Department of Biological Engineering, MIT, Cambridge, MA, USA
- Next Generation Analytics, Palo Alto, CA, USA
| | - Jonathan D Glass
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Todd M Herrington
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - James D Berry
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Kenney Ng
- Center for Computational Health and MIT-IBM Watson AI Lab, IBM Research, Cambridge, MA, USA
| | - Ernest Fraenkel
- Department of Biological Engineering, MIT, Cambridge, MA, USA.
| |
Collapse
|
17
|
Sista SRS, Shelly S, Oskarsson B, Rubin DI, Martinez-Thompson JM, Parra-Cantu C, Staff NP, Laughlin RS. Clinical and electrophysiological findings in C9ORF72 ALS. Muscle Nerve 2022; 66:270-275. [PMID: 35727129 DOI: 10.1002/mus.27665] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 11/10/2022]
Abstract
INTRODUCTION/AIMS Amyotrophic lateral sclerosis (ALS) is a progressive, fatal, neurodegenerative disorder of motor neurons in which the cause is mostly unknown. Early identification of genetic ALS cases, of which C9ORF72 (C9ALS) is the most frequent, can have important implications for evaluation, prognosis, and therapeutics. Here, we aimed to characterize the clinical and electrophysiological hallmarks of C9ALS and investigate differences from C9ORF72 negative ALS (non-C9ALS). METHODS We retrospectively reviewed clinical and electrodiagnostic (EDX) data for all genetically confirmed C9ALS cases seen between 1/1/2012 and 10/1/2020 who met Gold Coast criteria and compared them 1:1 with non-C9ALS patients within the same time frame. RESULTS A total of 99 C9ALS and 99 non-C9ALS cases were identified. Compared to non-C9ALS, C9ALS demonstrated higher prevalence in women, lesser racial variability, stronger family history of ALS, and higher frequency of upper motor neuron signs. EDX testing of C9ALS showed higher median sensory nerve and lower fibular compound muscle action potential amplitudes. DISCUSSION Although the differences between C9ALS and non-C9ALS reached statistical significance in certain nerve conduction parameters, they were not sufficient to discriminate between groups on a case-by-case basis. Genetic testing is required to identify C9ALS patients.
Collapse
Affiliation(s)
| | - Shahar Shelly
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA.,Department of Neurology, Sheba Medical Center, Tel HaShomer, Sackler Faculty of Medicine, Tel Aviv University, Middle East, Israel
| | - Björn Oskarsson
- Department of Neurology, Mayo Clinic, Jacksonville, Florida, USA
| | - Devon I Rubin
- Department of Neurology, Mayo Clinic, Jacksonville, Florida, USA
| | | | | | - Nathan P Staff
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | | |
Collapse
|
18
|
Zejlon C, Nakhostin D, Winklhofer S, Pangalu A, Kulcsar Z, Lewandowski S, Finnsson J, Piehl F, Ingre C, Granberg T, Ineichen BV. Structural magnetic resonance imaging findings and histopathological correlations in motor neuron diseases—A systematic review and meta-analysis. Front Neurol 2022; 13:947347. [PMID: 36110394 PMCID: PMC9468579 DOI: 10.3389/fneur.2022.947347] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectivesThe lack of systematic evidence on neuroimaging findings in motor neuron diseases (MND) hampers the diagnostic utility of magnetic resonance imaging (MRI). Thus, we aimed at performing a systematic review and meta-analysis of MRI features in MND including their histopathological correlation.MethodsIn a comprehensive literature search, out of 5941 unique publications, 223 records assessing brain and spinal cord MRI findings in MND were eligible for a qualitative synthesis. 21 records were included in a random effect model meta-analysis.ResultsOur meta-analysis shows that both T2-hyperintensities along the corticospinal tracts (CST) and motor cortex T2*-hypointensitites, also called “motor band sign”, are more prevalent in ALS patients compared to controls [OR 2.21 (95%-CI: 1.40–3.49) and 10.85 (95%-CI: 3.74–31.44), respectively]. These two imaging findings correlate to focal axonal degeneration/myelin pallor or glial iron deposition on histopathology, respectively. Additionally, certain clinical MND phenotypes such as amyotrophic lateral sclerosis (ALS) seem to present with distinct CNS atrophy patterns.ConclusionsAlthough CST T2-hyperintensities and the “motor band sign” are non-specific imaging features, they can be leveraged for diagnostic workup of suspected MND cases, together with certain brain atrophy patterns. Collectively, this study provides high-grade evidence for the usefulness of MRI in the diagnostic workup of suspected MND cases.Systematic review registrationhttps://www.crd.york.ac.uk/PROSPERO/, identifier: CRD42020182682.
Collapse
Affiliation(s)
- Charlotte Zejlon
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Dominik Nakhostin
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zürich, Switzerland
| | - Sebastian Winklhofer
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zürich, Switzerland
| | - Athina Pangalu
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zürich, Switzerland
| | - Zsolt Kulcsar
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zürich, Switzerland
| | | | - Johannes Finnsson
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Center of Neurology, Academic Specialist Center, Stockholm Health Services, Stockholm, Sweden
| | - Caroline Ingre
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Tobias Granberg
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Benjamin Victor Ineichen
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zürich, Switzerland
- *Correspondence: Benjamin Victor Ineichen
| |
Collapse
|
19
|
Liu Y, Xing H, Ernst AF, Liu C, Maugee C, Yokoi F, Lakshmana M, Li Y. Hyperactivity of Purkinje cell and motor deficits in C9orf72 knockout mice. Mol Cell Neurosci 2022; 121:103756. [PMID: 35843530 PMCID: PMC10369482 DOI: 10.1016/j.mcn.2022.103756] [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: 07/08/2022] [Accepted: 07/11/2022] [Indexed: 10/17/2022] Open
Abstract
A hexanucleotide (GGGGCC) repeat expansion in the first intron of the C9ORF72 gene is the most frequently reported genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). The cerebellum has not traditionally been thought to be involved in the pathogenesis of C9ORF72-associated ALS/FTD, but recent evidence suggested a potential role. C9ORF72 is highly expressed in the cerebellum. Decreased C9ORF72 transcript and protein levels were detected in the postmortem cerebellum, suggesting a loss-of-function effect of C9ORF72 mutation. This study investigated the role of loss of C9ORF72 function using a C9orf72 knockout mouse line. C9orf72 deficiency led to motor impairment in rotarod, beam-walking, paw-print, open-field, and grip-strength tests. Purkinje cells are the sole output neurons in the cerebellum, and we next determined their involvement in the motor phenotypes. We found hyperactivity of Purkinje cells in the C9orf72 knockout mouse accompanied by a significant increase of the large-conductance calcium-activated potassium channel (BK) protein in the cerebellum. The link between BK and Purkinje cell firing was demonstrated by the acute application of the BK activator that increased the firing frequency of the Purkinje cells ex vivo. In vivo chemogenetic activation of Purkinje cells in wild-type mice led to similar motor deficits in rotarod and beam-walking tests. Our results highlight that C9ORF72 loss alters the activity of the Purkinje cell and potentially the pathogenesis of the disease. Manipulating the Purkinje cell firing or cerebellar output may contribute to C9ORF72-associated ALS/FTD treatment.
Collapse
Affiliation(s)
- Yuning Liu
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, United States; Genetics Institute, University of Florida, Gainesville, FL, United States
| | - Hong Xing
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Alexis F Ernst
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Canna Liu
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Christian Maugee
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, United States; Genetics Institute, University of Florida, Gainesville, FL, United States
| | - Fumiaki Yokoi
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Madepalli Lakshmana
- Department of Immunology and Nano-Medicine, The Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
| | - Yuqing Li
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, United States.
| |
Collapse
|
20
|
Gene Therapy in Amyotrophic Lateral Sclerosis. Cells 2022; 11:cells11132066. [PMID: 35805149 PMCID: PMC9265980 DOI: 10.3390/cells11132066] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 12/30/2022] Open
Abstract
Since the discovery of Cu/Zn superoxide dismutase (SOD1) gene mutation, in 1993, as the first genetic abnormality in amyotrophic lateral sclerosis (ALS), over 50 genes have been identified as either cause or modifier in ALS and ALS/frontotemporal dementia (FTD) spectrum disease. Mutations in C9orf72, SOD1, TAR DNA binding protein 43 (TARDBP), and fused in sarcoma (FUS) genes are the four most common ones. During the last three decades, tremendous effort has been made worldwide to reveal biological pathways underlying the pathogenesis of these gene mutations in ALS/FTD. Accordingly, targeting etiologic genes (i.e., gene therapies) to suppress their toxic effects have been investigated widely. It includes four major strategies: (i) removal or inhibition of abnormal transcribed RNA using microRNA or antisense oligonucleotides (ASOs), (ii) degradation of abnormal mRNA using RNA interference (RNAi), (iii) decrease or inhibition of mutant proteins (e.g., using antibodies against misfolded proteins), and (iv) DNA genome editing with methods such as clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein (CRISPR/Cas). The promising results of these studies have led to the application of some of these strategies into ALS clinical trials, especially for C9orf72 and SOD1. In this paper, we will overview advances in gene therapy in ALS/FTD, focusing on C9orf72, SOD1, TARDBP, and FUS genes.
Collapse
|
21
|
Su WM, Gu XJ, Duan QQ, Jiang Z, Gao X, Shang HF, Chen YP. Genetic factors for survival in amyotrophic lateral sclerosis: an integrated approach combining a systematic review, pairwise and network meta-analysis. BMC Med 2022; 20:209. [PMID: 35754054 PMCID: PMC9235235 DOI: 10.1186/s12916-022-02411-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/18/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The time of survival in patients with amyotrophic lateral sclerosis (ALS) varies greatly, and the genetic factors that contribute to the survival of ALS are not well studied. There is a lack of a comprehensive study to elucidate the role of genetic factors in the survival of ALS. METHODS The published studies were systematically searched and obtained from PubMed, EMBASE, and the Cochrane Library without any language restrictions from inception to Oct 27, 2021. A network meta-analysis for ALS causative/risk genes and a systematic review and pairwise meta-analysis for other genetic modifiers were conducted. The PROSPERO registration number: CRD42022311646. RESULTS A total of 29,764 potentially relevant references were identified, and 71 papers were eligible for analysis based on pre-decided criteria, including 35 articles in network meta-analysis for 9 ALS causative/risk genes, 17 articles in pairwise meta-analysis for four genetic modifiers, and 19 articles described in the systematic review. Variants in three genes, including ATXN2 (HR: 3.6), C9orf72 (HR: 1.6), and FUS (HR:1.8), were associated with short survival of ALS, but such association was not identified in SOD1, TARDBP, TBK1, NEK1, UBQLN2, and CCNF. In addition, UNC13A rs12608932 CC genotype and ZNF521B rs2275294 C allele also caused a shorter survival of ALS; however, APOE ε4 allele and KIFAP3 rs1541160 did not be found to have any effect on the survival of ALS. CONCLUSIONS Our study summarized and contrasted evidence for prognostic genetic factors in ALS and would help to understand ALS pathogenesis and guide clinical trials and drug development.
Collapse
Affiliation(s)
- Wei-Ming Su
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Lab of Neurodegenerative Disorders, Institute of Inflammation and Immunology (III), Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Centre for Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xiao-Jing Gu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Lab of Neurodegenerative Disorders, Institute of Inflammation and Immunology (III), Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Centre for Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Qing-Qing Duan
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Lab of Neurodegenerative Disorders, Institute of Inflammation and Immunology (III), Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Centre for Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zheng Jiang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Lab of Neurodegenerative Disorders, Institute of Inflammation and Immunology (III), Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Centre for Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xia Gao
- Department of Geriatrics, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Hui-Fang Shang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Lab of Neurodegenerative Disorders, Institute of Inflammation and Immunology (III), Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Centre for Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yong-Ping Chen
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
- Lab of Neurodegenerative Disorders, Institute of Inflammation and Immunology (III), Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
- Centre for Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
22
|
Hou X, Li W, Liu P, Liu Z, Yuan Y, Ni J, Shen L, Tang B, Wang J. The Clinical and Ploynucleotide Repeat Expansion Analysis of ATXN2, NOP56, AR and C9orf72 in Patients With ALS From Mainland China. Front Neurol 2022; 13:811202. [PMID: 35599735 PMCID: PMC9120572 DOI: 10.3389/fneur.2022.811202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Background Repeat expansions, including those in C9orf72 and ATXN2, have been implicated in amyotrophic lateral sclerosis (ALS). However, there have been few studies on the association of AR and NOP56 repeat expansion with ALS, especially in China. Accordingly, we aimed to evaluate the frequency of C9orf72 and ATXN2 repeat mutations and investigate whether NOP56 and AR repeat expansion are risk factors for ALS. Methods In this study, 736 ALS patients and several hundred healthy controls were recruited. Polymerase chain reaction (PCR) and repeat-primed PCR (RP-PCR) were performed to determine the repeat lengths in C9orf72, ATXN2, AR, and NOP56. Results GGGGCC repeats in C9orf72 were observed in six ALS patients (0.8%, 6/736) but not in any of the controls (0/365). The patients with pathogenic GGGGCC repeats showed shorter median survival times than those with a normal genotype (p = 0.006). Regarding ATXN2 CAG repeats, we identified that intermediate repeat lengths (29–34 copies) were associated with ALS (p = 0.033), and there was no difference in clinical characteristics between the groups with and without intermediate repeats (p > 0.05). Meanwhile, we observed that there was no association between the repeat size in AR and NOP56 and ALS (p > 0.05). Conclusions Our results demonstrated that pathogenetic repeats in C9orf72 are rare in China, while intermediate CAG repeats in ATXN2 are more frequent but have no effect on disease phenotypes; the repeat size in AR and NOP56 may not be a risk factor for ALS.
Collapse
Affiliation(s)
- Xiaorong Hou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Wanzhen Li
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Pan Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhen Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Yanchun Yuan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Jie Ni
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Lu Shen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- Laboratory of Medical Genetics, Central South University, Changsha, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- Laboratory of Medical Genetics, Central South University, Changsha, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
| | - Junling Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- Laboratory of Medical Genetics, Central South University, Changsha, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
- *Correspondence: Junling Wang
| |
Collapse
|
23
|
Zhang S, Shen L, Jiao B. Cognitive Dysfunction in Repeat Expansion Diseases: A Review. Front Aging Neurosci 2022; 14:841711. [PMID: 35478698 PMCID: PMC9036481 DOI: 10.3389/fnagi.2022.841711] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/24/2022] [Indexed: 11/16/2022] Open
Abstract
With the development of the sequencing technique, more than 40 repeat expansion diseases (REDs) have been identified during the past two decades. Moreover, the clinical features of these diseases show some commonality, and the nervous system, especially the cognitive function was affected in part by these diseases. However, the specific cognitive domains impaired in different diseases were inconsistent. Here, we survey literature on the cognitive consequences of the following disorders presenting cognitive dysfunction and summarizing the pathogenic genes, epidemiology, and different domains affected by these diseases. We found that the cognitive domains affected in neuronal intranuclear inclusion disease (NIID) were widespread including the executive function, memory, information processing speed, attention, visuospatial function, and language. Patients with C9ORF72-frontotemporal dementia (FTD) showed impairment in executive function, memory, language, and visuospatial function. While in Huntington's disease (HD), the executive function, memory, and information processing speed were affected, in the fragile X-associated tremor/ataxia syndrome (FXTAS), executive function, memory, information processing speed, and attention were impaired. Moreover, the spinocerebellar ataxias showed broad damage in almost all the cognitive domains except for the relatively intact language ability. Some other diseases with relatively rare clinical data also indicated cognitive dysfunction, such as myotonic dystrophy type 1 (DM1), progressive myoclonus epilepsy (PME), Friedreich ataxia (FRDA), Huntington disease like-2 (HDL2), and cerebellar ataxia, neuropathy, vestibular areflexia syndrome (CANVAS). We drew a cognitive function landscape of the related REDs that might provide an aspect for differential diagnosis through cognitive domains and effective non-specific interventions for these diseases.
Collapse
Affiliation(s)
- Sizhe Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Lu Shen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Bin Jiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- *Correspondence: Bin Jiao
| |
Collapse
|
24
|
Thalamic and Cerebellar Regional Involvement across the ALS-FTD Spectrum and the Effect of C9orf72. Brain Sci 2022; 12:brainsci12030336. [PMID: 35326292 PMCID: PMC8945983 DOI: 10.3390/brainsci12030336] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/23/2022] [Accepted: 02/27/2022] [Indexed: 02/01/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are part of the same disease spectrum. While thalamic−cerebellar degeneration has been observed in C9orf72 expansion carriers, the exact subregions involved across the clinical phenotypes of the ALS−FTD spectrum remain unclear. Using MRIs from 58 bvFTD, 41 ALS−FTD and 52 ALS patients compared to 57 controls, we aimed to delineate thalamic and cerebellar subregional changes across the ALS−FTD spectrum and to contrast these profiles between cases with and without C9orf72 expansions. Thalamic involvement was evident across all ALS−FTD clinical phenotypes, with the laterodorsal nucleus commonly affected across all groups (values below the 2.5th control percentile). The mediodorsal nucleus was disproportionately affected in bvFTD and ALS−FTD but not in ALS. Cerebellar changes were only observed in bvFTD and ALS−FTD predominantly in the superior−posterior region. Comparison of genetic versus sporadic cases revealed significantly lower volumes exclusively in the pulvinar in C9orf72 expansion carriers compared to non-carriers, irrespective of clinical syndrome. Overall, bvFTD showed significant correlations between thalamic subregions, level of cognitive dysfunction and severity of behavioural symptoms. Notably, strong associations were evident between mediodorsal nucleus atrophy and severity of behavioural changes in C9orf72-bvFTD (r = −0.9, p < 0.0005). Our findings reveal distinct thalamic and cerebellar atrophy profiles across the ALS−FTD spectrum, with differential impacts on behaviour and cognition, and point to a unique contribution of C9orf72 expansions in the clinical profiles of these patients.
Collapse
|
25
|
Chen Y, Landin-Romero R, Kumfor F, Irish M, Dobson-Stone C, Kwok JB, Halliday GM, Hodges JR, Piguet O. Cerebellar integrity and contributions to cognition in C9orf72-mediated frontotemporal dementia. Cortex 2022; 149:73-84. [DOI: 10.1016/j.cortex.2021.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 10/07/2021] [Accepted: 12/22/2021] [Indexed: 11/03/2022]
|
26
|
Common genetic variation is associated with longitudinal decline and network features in behavioral variant frontotemporal degeneration. Neurobiol Aging 2021; 108:16-23. [PMID: 34474300 PMCID: PMC8616801 DOI: 10.1016/j.neurobiolaging.2021.07.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 07/25/2021] [Accepted: 07/28/2021] [Indexed: 01/28/2023]
Abstract
The T allele in rs1768208 located in or near the myelin oligodendrocyte basic protein gene (MOBP) is a risk factor for frontotemporal degeneration pathology. We evaluated the hypothesis that the presence of a T allele in rs1768208 will be associated with rate of cognitive decline in behavioral variant frontotemporal degeneration (bvFTD) related to compromised frontal networks. We studied 81 individuals clinically diagnosed with bvFTD who were genotyped for rs1768208 and coded using a dominant model reflecting the presence (i.e., MOBP +) or absence (MOBP -) of the T risk allele. Linear mixed-effects models assessed the association of genotype on neuropsychological performance over time. Regression analyses examined differences in network structure by MOBP genotype. We found a genotype by time interaction for declining cognitive performance, whereby MOBP + individuals demonstrated faster rates of decline in executive function. The presence of a MOBP risk allele was associated with degradation of white matter network features in the frontal lobe. These findings suggest that individual genetic variation may contribute to heterogeneity in clinical progression.
Collapse
|
27
|
Bocchetta M, Malpetti M, Todd EG, Rowe JB, Rohrer JD. Looking beneath the surface: the importance of subcortical structures in frontotemporal dementia. Brain Commun 2021; 3:fcab158. [PMID: 34458729 PMCID: PMC8390477 DOI: 10.1093/braincomms/fcab158] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2021] [Indexed: 12/15/2022] Open
Abstract
Whilst initial anatomical studies of frontotemporal dementia focussed on cortical involvement, the relevance of subcortical structures to the pathophysiology of frontotemporal dementia has been increasingly recognized over recent years. Key structures affected include the caudate, putamen, nucleus accumbens, and globus pallidus within the basal ganglia, the hippocampus and amygdala within the medial temporal lobe, the basal forebrain, and the diencephalon structures of the thalamus, hypothalamus and habenula. At the most posterior aspect of the brain, focal involvement of brainstem and cerebellum has recently also been shown in certain subtypes of frontotemporal dementia. Many of the neuroimaging studies on subcortical structures in frontotemporal dementia have been performed in clinically defined sporadic cases. However, investigations of genetically- and pathologically-confirmed forms of frontotemporal dementia are increasingly common and provide molecular specificity to the changes observed. Furthermore, detailed analyses of sub-nuclei and subregions within each subcortical structure are being added to the literature, allowing refinement of the patterns of subcortical involvement. This review focuses on the existing literature on structural imaging and neuropathological studies of subcortical anatomy across the spectrum of frontotemporal dementia, along with investigations of brain–behaviour correlates that examine the cognitive sequelae of specific subcortical involvement: it aims to ‘look beneath the surface’ and summarize the patterns of subcortical involvement have been described in frontotemporal dementia.
Collapse
Affiliation(s)
- Martina Bocchetta
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Maura Malpetti
- Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, University of Cambridge, Cambridge, UK
| | - Emily G Todd
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - James B Rowe
- Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, University of Cambridge, Cambridge, UK.,Medical Research Council Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
28
|
Cognitive dysfunction in amyotrophic lateral sclerosis: can we predict it? Neurol Sci 2021; 42:2211-2222. [PMID: 33772353 PMCID: PMC8159827 DOI: 10.1007/s10072-021-05188-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/15/2021] [Indexed: 01/26/2023]
Abstract
Background and aim Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder characterized by the degeneration of both upper and lower motoneurons in the brain and spinal cord leading to motor and extra-motor symptoms. Although traditionally considered a pure motor disease, recent evidences suggest that ALS is a multisystem disorder. Neuropsychological alterations, in fact, are observed in more than 50% of patients: while executive dysfunctions have been firstly identified, alterations in verbal fluency, behavior, and pragmatic and social cognition have also been described. Detecting and monitoring ALS cognitive and behavioral impairment even at early disease stages is likely to have staging and prognostic implications, and it may impact the enrollment in future clinical trials. During the last 10 years, humoral, radiological, neurophysiological, and genetic biomarkers have been reported in ALS, and some of them seem to potentially correlate to cognitive and behavioral impairment of patients. In this review, we sought to give an up-to-date state of the art of neuropsychological alterations in ALS: we will describe tests used to detect cognitive and behavioral impairment, and we will focus on promising non-invasive biomarkers to detect pre-clinical cognitive decline. Conclusions To date, the research on humoral, radiological, neurophysiological, and genetic correlates of neuropsychological alterations is at the early stage, and no conclusive longitudinal data have been published. Further and longitudinal studies on easily accessible and quantifiable biomarkers are needed to clarify the time course and the evolution of cognitive and behavioral impairments of ALS patients.
Collapse
|
29
|
Temp AGM, Prudlo J, Vielhaber S, Machts J, Hermann A, Teipel SJ, Kasper E. Cognitive reserve and regional brain volume in amyotrophic lateral sclerosis. Cortex 2021; 139:240-248. [PMID: 33892294 DOI: 10.1016/j.cortex.2021.03.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 01/07/2021] [Accepted: 03/04/2021] [Indexed: 12/29/2022]
Abstract
OBJECTIVE We investigated whether cognitive reserve measured by education and premorbid IQ allows amyotrophic lateral sclerosis patients to compensate for regional brain volume loss. METHODS This was a cross-sectional study. We recruited sixty patients with amyotrophic lateral sclerosis from two specialist out-patient clinics. All participants underwent neuropsychological assessment; the outcomes were standardized z-scores reflecting verbal fluency, executive functions (shifting, planning, working memory), verbal memory and visuo-constructive ability. The predictor was regional brain volume. The moderating proxies of cognitive reserve were premorbid IQ (estimated by vocabulary) and educational years. We hypothesized that higher cognitive reserve would correlate with better performance on a cognitive test battery, and tested this hypothesis with Bayesian analysis of covariance. RESULTS The analyses provided moderate to very strong evidence in favor of our hypothesis with regard to verbal fluency functions, working memory, verbal learning and recognition, and visuo-constructive ability (all BF01 > 3): higher cognitive reserve was associated with a mild increase in performance. For shifting and planning ability, the evidence was anecdotal. CONCLUSIONS These results indicate that cognitive reserve moderates the effect of brain morphology on cognition in ALS. Patients draw small but meaningful benefits from higher reserve, preserving fluency, memory and visuo-constructive functions. Executive functions presented a dissociation: verbally assessed functions benefitted from cognitive reserve, non-verbally assessed functions did not. This motivates future research into cognitive reserve in ALS and practical implications, such as strengthening reserve to delay decline.
Collapse
Affiliation(s)
- Anna G M Temp
- German Centre for Neurodegenerative Diseases (DZNE), Rostock, Germany.
| | - Johannes Prudlo
- German Centre for Neurodegenerative Diseases (DZNE), Rostock, Germany; Department of Neurology, University of Rostock, Rostock, Germany.
| | - Stefan Vielhaber
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany; German Centre for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.
| | - Judith Machts
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany; German Centre for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.
| | - Andreas Hermann
- German Centre for Neurodegenerative Diseases (DZNE), Rostock, Germany; Translational Neurodegeneration Section "Albrecht-Kossel", Department of Neurology, University of Rostock, Rostock, Germany.
| | - Stefan J Teipel
- German Centre for Neurodegenerative Diseases (DZNE), Rostock, Germany; Department of Psychosomatic Medicine, University of Rostock, Rostock, Germany.
| | - Elisabeth Kasper
- German Centre for Neurodegenerative Diseases (DZNE), Rostock, Germany; Department of Neurology, University of Rostock, Rostock, Germany.
| |
Collapse
|
30
|
Kinney NG, Bove J, Phillips JS, Cousins KAQ, Olm CA, Wakeman DG, McMillan CT, Massimo L. Social and leisure activity are associated with attenuated cortical loss in behavioral variant frontotemporal degeneration. Neuroimage Clin 2021; 30:102629. [PMID: 33770546 PMCID: PMC8024767 DOI: 10.1016/j.nicl.2021.102629] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 02/16/2021] [Accepted: 03/07/2021] [Indexed: 12/04/2022]
Abstract
Behavioral variant frontotemporal degeneration (bvFTD) is clinically characterized by progressive decline in social and executive domains. Previous work suggests that early lifestyle factors such as education and occupational attainment may relate to structural integrity and moderate the rate of cognitive decline in bvFTD, but the role of other cognitively stimulating activities is understudied. We sought to investigate the effect of such activities on cortical thickness (CT) in bvFTD. bvFTD patients (n = 31) completed a baseline MRI scan, and informants for the patients completed the Lifetime of Experiences Questionnaire (LEQ), which measures specific activities considered to be undertaken primarily within one particular life phase, such as education (young-life), occupation (mid-life), and social/leisure activity (late-life). At baseline, linear models assessed the effect of LEQ scores from each life phase on regional CT. A subset (n = 19) of patients completed longitudinal MRI, and to evaluate the association of LEQ with longitudinal rates of CT decline, we derived individualized slopes of decline using linear mixed effects models and these were related to LEQ scores from each life phase. At baseline, a higher late-life LEQ score was associated with less atrophy in left superior and inferior anterior temporal regions as well as right middle temporal gyrus. Longitudinally, we observed that higher late-life LEQ scores were associated with an attenuated rate of CT loss in insular cortex. Late-life LEQ score was positively associated with both relatively preserved CT early in bvFTD and a slower rate of cortical loss in regions important for social functioning. These findings suggest that social and leisure activities may contribute to a form of resilience against pathologic effects of disease.
Collapse
Affiliation(s)
- Nikolas G Kinney
- Frontotemporal Degeneration Center, Perelman School of Medicine, Department of Neurology, Philadelphia, PA, United States
| | - Jessica Bove
- Frontotemporal Degeneration Center, Perelman School of Medicine, Department of Neurology, Philadelphia, PA, United States
| | - Jeffrey S Phillips
- Frontotemporal Degeneration Center, Perelman School of Medicine, Department of Neurology, Philadelphia, PA, United States
| | - Katheryn A Q Cousins
- Frontotemporal Degeneration Center, Perelman School of Medicine, Department of Neurology, Philadelphia, PA, United States
| | - Christopher A Olm
- Frontotemporal Degeneration Center, Perelman School of Medicine, Department of Neurology, Philadelphia, PA, United States
| | - Daniel G Wakeman
- Frontotemporal Degeneration Center, Perelman School of Medicine, Department of Neurology, Philadelphia, PA, United States
| | - Corey T McMillan
- Frontotemporal Degeneration Center, Perelman School of Medicine, Department of Neurology, Philadelphia, PA, United States
| | - Lauren Massimo
- Frontotemporal Degeneration Center, Perelman School of Medicine, Department of Neurology, Philadelphia, PA, United States; School of Nursing, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
31
|
Mitochondrial dysfunction and traffic jams in amyotrophic lateral sclerosis. Mitochondrion 2021; 58:102-110. [PMID: 33639271 DOI: 10.1016/j.mito.2021.02.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/07/2021] [Accepted: 02/12/2021] [Indexed: 12/11/2022]
Abstract
Neurodegenerative diseases are characterized by progressive neuronal loss anatomically or physiologically and accumulation of protein in the cells. Mitochondria provide energy to these neuronal cells consuming 20% of the body's oxygen. Mitochondria are the dynamic membrane-bound cell organelles that function to generate ATP, regulate calcium homeostasis, and produce reactive oxygen species. Because of alterations in the electron transport chain, mutation, and environmental toxins, there is reduced ATP production, calcium dyshomeostasis, and increased oxidative stress, resulting in mitochondrial dysfunction, leading to the pathogenesis of neurodegenerative diseases such as ALS. ALS is described as the loss of upper and lower motor neurons resulting in progressive muscle denervation and loss of voluntary movements. There are multiple shreds of evidence in the literature regarding the mechanism involved in mitochondrial dysfunction and possible therapeutic targets to treat the condition. Moreover, different studies reported the role of different gene mutations and malfunctions in transport system responsible for the accumulation and aggregation of the proteins inside the brain cells. This accumulation and/or aggregation of proteins in the neuronal cells is known as neuronal traffic jam, which also plays the leading role in the progressive neurodegenerative diseases. In this review, we have elucidated the critical insights into mitochondrial dysfunction and neuronal traffic jam; and its role in the initiation and progression of ALS. Moreover, the pharmacological targets and possible conducts to this scenario are also brought together.
Collapse
|
32
|
Glasmacher SA, Wong C, Pearson IE, Pal S. Survival and Prognostic Factors in C9orf72 Repeat Expansion Carriers: A Systematic Review and Meta-analysis. JAMA Neurol 2021; 77:367-376. [PMID: 31738367 DOI: 10.1001/jamaneurol.2019.3924] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Importance The c9orf72 repeat expansion (c9 or c9orf72RE) confers a survival disadvantage in amyotrophic lateral sclerosis (ALS); its effect on prognosis in frontotemporal dementia (FTD) remains uncertain. Data on prognostic factors in c9orf72RE disorders could inform patient care, genetic counseling, and trial design. Objective To examine prognostic factors in c9ALS, c9FTD, c9ALS-FTD, and atypical phenotypes. Data Sources The MEDLINE, Embase, Amed, ProQuest, PsychINFO, CINAHL, and LILACS databases were searched between January 2011 and January 2019. Keywords used were c9orf72 and chromosome 9 open reading frame 72. Reference lists, citations of eligible studies, and review articles were also searched by hand. Study Selection Studies reporting disease duration for patients with a confirmed c9orf72RE and a neurological and/or psychiatric disorder were included. A second author independently reviewed studies classified as irrelevant by the first author. Analysis began in January 2019. Data Extraction and Synthesis Data were extracted by 1 author; a further author independently extracted 10% of data. Data were synthesized in univariate and multivariable Cox regression and are displayed as hazard ratios (HR) and 95% confidence intervals. Main Outcomes and Measures Survival after symptom onset. Results Overall, 206 studies reporting on 1060 patients were included from 2878 publications identified (c9ALS: n = 455; c9FTD: n = 296; c9ALS-FTD: n = 198; atypical phenotypes: n = 111); 197 duplicate cases were excluded. The median (95% CI) survival (in years) differed significantly between patients with c9ALS (2.8 [2.67-3.00]), c9FTD (9.0 [8.09-9.91]), and c9ALS-FTD (3.0 [2.73-3.27]); survival in atypical phenotypes varied substantially. Older age at onset was associated with shorter survival in c9ALS (HR, 1.03; 95% CI, 1.02-1.04; P < .001), c9FTD (HR, 1.04; 95% CI, 1.02-1.06; P < .001), and c9ALS-FTD (HR, 1.02; 95% CI, 1.004-1.04; P = .016). Bulbar onset was associated with shorter survival in c9ALS (HR, 1.64; 95% CI, 1.27-2.08; P < .001). Age at onset and bulbar onset ALS remained significant in multivariable regression including variables indicating potential diagnostic ascertainment bias, selection bias, and reporting bias. Family history, sex, study continent, FTD subtype, or the presence of additional pathogenic sequence variants were not significantly associated with survival. Clinical phenotypes in patients with neuropathologically confirmed frontotemporal lobar degeneration-TDP-43, motor neuron disease-TDP-43 and frontotemporal lobar degeneration-motor neuron disease-TDP-43 were heterogenous and impacted on survival. Conclusions and Relevance Several factors associated with survival in c9orf72RE disorders were identified. The inherent limitations of our methodological approach must be considered; nonetheless, the reported prognostic factors were not significantly associated with the bias indicators examined.
Collapse
Affiliation(s)
- Stella A Glasmacher
- College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Charis Wong
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom.,Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, United Kingdom.,Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Iona E Pearson
- College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Suvankar Pal
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom.,Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, United Kingdom.,Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
33
|
Ghasemi M, Keyhanian K, Douthwright C. Glial Cell Dysfunction in C9orf72-Related Amyotrophic Lateral Sclerosis and Frontotemporal Dementia. Cells 2021; 10:cells10020249. [PMID: 33525344 PMCID: PMC7912327 DOI: 10.3390/cells10020249] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/14/2021] [Accepted: 01/25/2021] [Indexed: 12/17/2022] Open
Abstract
Since the discovery of the chromosome 9 open reading frame 72 (C9orf72) repeat expansion mutation in 2011 as the most common genetic abnormality in amyotrophic lateral sclerosis (ALS, also known as Lou Gehrig's disease) and frontotemporal dementia (FTD), progress in understanding the signaling pathways related to this mutation can only be described as intriguing. Two major theories have been suggested-(i) loss of function or haploinsufficiency and (ii) toxic gain of function from either C9orf72 repeat RNA or dipeptide repeat proteins (DPRs) generated from repeat-associated non-ATG (RAN) translation. Each theory has provided various signaling pathways that potentially participate in the disease progression. Dysregulation of the immune system, particularly glial cell dysfunction (mainly microglia and astrocytes), is demonstrated to play a pivotal role in both loss and gain of function theories of C9orf72 pathogenesis. In this review, we discuss the pathogenic roles of glial cells in C9orf72 ALS/FTD as evidenced by pre-clinical and clinical studies showing the presence of gliosis in C9orf72 ALS/FTD, pathologic hallmarks in glial cells, including TAR DNA-binding protein 43 (TDP-43) and p62 aggregates, and toxicity of C9orf72 glial cells. A better understanding of these pathways can provide new insights into the development of therapies targeting glial cell abnormalities in C9orf72 ALS/FTD.
Collapse
Affiliation(s)
- Mehdi Ghasemi
- Correspondence: ; Tel.: +1-774-441-7726; Fax: +1-508-856-4485
| | | | | |
Collapse
|
34
|
El-Wahsh S, Finger EC, Piguet O, Mok V, Rohrer JD, Kiernan MC, Ahmed RM. Predictors of survival in frontotemporal lobar degeneration syndromes. J Neurol Neurosurg Psychiatry 2021; 92:jnnp-2020-324349. [PMID: 33441385 DOI: 10.1136/jnnp-2020-324349] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/26/2020] [Accepted: 12/14/2020] [Indexed: 12/11/2022]
Abstract
After decades of research, large-scale clinical trials in patients diagnosed with frontotemporal lobar degeneration (FTLD) are now underway across multiple centres worldwide. As such, refining the determinants of survival in FTLD represents a timely and important challenge. Specifically, disease outcome measures need greater clarity of definition to enable accurate tracking of therapeutic interventions in both clinical and research settings. Multiple factors potentially determine survival, including the clinical phenotype at presentation; radiological patterns of atrophy including markers on both structural and functional imaging; metabolic factors including eating behaviour and lipid metabolism; biomarkers including both serum and cerebrospinal fluid markers of underlying pathology; as well as genetic factors, including both dominantly inherited genes, but also genetic modifiers. The present review synthesises the effect of these factors on disease survival across the syndromes of frontotemporal dementia, with comparison to amyotrophic lateral sclerosis, progressive supranuclear palsy and corticobasal syndrome. A pathway is presented that outlines the utility of these varied survival factors for future clinical trials and drug development. Given the complexity of the FTLD spectrum, it seems unlikely that any single factor may predict overall survival in individual patients, further suggesting that a precision medicine approach will need to be developed in predicting disease survival in FTLD, to enhance drug target development and future clinical trial methodologies.
Collapse
Affiliation(s)
- Shadi El-Wahsh
- Department of Neurology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Elizabeth C Finger
- Department of Clinicial Neurological Sciences, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Olivier Piguet
- Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Vincent Mok
- Gerald Choa Neuroscience Centre, Lui Che Woo Institute of Innovative Medicine, Margaret K.L. Cheung Research Centre for Management of Parkinsonism, Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Matthew C Kiernan
- Department of Neurology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Rebekah M Ahmed
- Department of Neurology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
35
|
Li Hi Shing S, McKenna MC, Siah WF, Chipika RH, Hardiman O, Bede P. The imaging signature of C9orf72 hexanucleotide repeat expansions: implications for clinical trials and therapy development. Brain Imaging Behav 2021; 15:2693-2719. [PMID: 33398779 DOI: 10.1007/s11682-020-00429-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2020] [Indexed: 01/14/2023]
Abstract
While C9orf72-specific imaging signatures have been proposed by both ALS and FTD research groups and considerable presymptomatic alterations have also been confirmed in young mutation carriers, considerable inconsistencies exist in the literature. Accordingly, a systematic review of C9orf72-imaging studies has been performed to identify consensus findings, stereotyped shortcomings, and unique contributions to outline future directions. A formal literature review was conducted according to the STROBE guidelines. All identified papers were individually reviewed for sample size, choice of controls, study design, imaging modalities, statistical models, clinical profiling, and identified genotype-associated pathological patterns. A total of 74 imaging papers were systematically reviewed. ALS patients with GGGGCC repeat expansions exhibit relatively limited motor cortex involvement and widespread extra-motor pathology. C9orf72 positive FTD patients often show preferential posterior involvement. Reports of thalamic involvement are relatively consistent across the various phenotypes. Asymptomatic hexanucleotide repeat carriers often exhibit structural and functional changes decades prior to symptom onset. Common shortcomings included sample size limitations, lack of disease-controls, limited clinical profiling, lack of genetic testing in healthy controls, and absence of post mortem validation. There is a striking paucity of longitudinal studies and existing presymptomatic studies have not evaluated the predictive value of radiological changes with regard to age of onset and phenoconversion. With the advent of antisense oligonucleotide therapies, the meticulous characterisation of C9orf72-associated changes has gained practical relevance. Neuroimaging offers non-invasive biomarkers for future clinical trials, presymptomatic ascertainment, diagnostic and prognostic applications.
Collapse
Affiliation(s)
- Stacey Li Hi Shing
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Mary Clare McKenna
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - We Fong Siah
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Rangariroyashe H Chipika
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Orla Hardiman
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Peter Bede
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
36
|
Pasquini L, Nana AL, Toller G, Brown JA, Deng J, Staffaroni A, Kim EJ, Hwang JHL, Li L, Park Y, Gaus SE, Allen I, Sturm VE, Spina S, Grinberg LT, Rankin KP, Kramer JH, Rosen HJ, Miller BL, Seeley WW. Salience Network Atrophy Links Neuron Type-Specific Pathobiology to Loss of Empathy in Frontotemporal Dementia. Cereb Cortex 2020; 30:5387-5399. [PMID: 32500143 PMCID: PMC7566683 DOI: 10.1093/cercor/bhaa119] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/21/2020] [Accepted: 04/16/2020] [Indexed: 12/12/2022] Open
Abstract
Each neurodegenerative syndrome reflects a stereotyped pattern of cellular, regional, and large-scale brain network degeneration. In behavioral variant of frontotemporal dementia (bvFTD), a disorder of social-emotional function, von Economo neurons (VENs), and fork cells are among the initial neuronal targets. These large layer 5 projection neurons are concentrated in the anterior cingulate and frontoinsular (FI) cortices, regions that anchor the salience network, a large-scale system linked to social-emotional function. Here, we studied patients with bvFTD, amyotrophic lateral sclerosis (ALS), or both, given that these syndromes share common pathobiological and genetic factors. Our goal was to determine how neuron type-specific TAR DNA-binding protein of 43 kDa (TDP-43) pathobiology relates to atrophy in specific brain structures and to loss of emotional empathy, a cardinal feature of bvFTD. We combined questionnaire-based empathy assessments, in vivo structural MR imaging, and quantitative histopathological data from 16 patients across the bvFTD/ALS spectrum. We show that TDP-43 pathobiology within right FI VENs and fork cells is associated with salience network atrophy spanning insular, medial frontal, and thalamic regions. Gray matter degeneration within these structures mediated loss of emotional empathy, suggesting a chain of influence linking the cellular, regional/network, and behavioral levels in producing signature bvFTD clinical features.
Collapse
Affiliation(s)
- Lorenzo Pasquini
- Department of Neurology, Memory and Aging Center, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, California 94158, USA
| | - Alissa L Nana
- Department of Neurology, Memory and Aging Center, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, California 94158, USA
| | - Gianina Toller
- Department of Neurology, Memory and Aging Center, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, California 94158, USA
| | - Jesse A Brown
- Department of Neurology, Memory and Aging Center, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, California 94158, USA
| | - Jersey Deng
- Department of Neurology, Memory and Aging Center, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, California 94158, USA
| | - Adam Staffaroni
- Department of Neurology, Memory and Aging Center, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, California 94158, USA
| | - Eun-Joo Kim
- Department of Neurology, Memory and Aging Center, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, California 94158, USA
| | - Ji-Hye L Hwang
- Department of Neurology, Memory and Aging Center, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, California 94158, USA
| | - Libo Li
- Department of Neurology, Memory and Aging Center, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, California 94158, USA
- Department of Psychopharmacology, Qiqihar Medical University, 333 Bukui N St, Qiqihar 161006, China
| | - Youngsoon Park
- Department of Neurology, Memory and Aging Center, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, California 94158, USA
| | - Stephanie E Gaus
- Department of Neurology, Memory and Aging Center, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, California 94158, USA
| | - Isabel Allen
- Department of Neurology, Memory and Aging Center, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, California 94158, USA
| | - Virginia E Sturm
- Department of Neurology, Memory and Aging Center, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, California 94158, USA
| | - Salvatore Spina
- Department of Neurology, Memory and Aging Center, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, California 94158, USA
| | - Lea T Grinberg
- Department of Neurology, Memory and Aging Center, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, California 94158, USA
- Department of Pathology, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, California 94158, USA
| | - Katherine P Rankin
- Department of Neurology, Memory and Aging Center, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, California 94158, USA
| | - Joel H Kramer
- Department of Neurology, Memory and Aging Center, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, California 94158, USA
| | - Howard J Rosen
- Department of Neurology, Memory and Aging Center, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, California 94158, USA
| | - Bruce L Miller
- Department of Neurology, Memory and Aging Center, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, California 94158, USA
| | - William W Seeley
- Department of Neurology, Memory and Aging Center, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, California 94158, USA
- Department of Pathology, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, California 94158, USA
| |
Collapse
|
37
|
Häkkinen S, Chu SA, Lee SE. Neuroimaging in genetic frontotemporal dementia and amyotrophic lateral sclerosis. Neurobiol Dis 2020; 145:105063. [PMID: 32890771 DOI: 10.1016/j.nbd.2020.105063] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/30/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023] Open
Abstract
Frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) have a strong clinical, genetic and pathological overlap. This review focuses on the current understanding of structural, functional and molecular neuroimaging signatures of genetic FTD and ALS. We overview quantitative neuroimaging studies on the most common genes associated with FTD (MAPT, GRN), ALS (SOD1), and both (C9orf72), and summarize visual observations of images reported in the rarer genes (CHMP2B, TARDBP, FUS, OPTN, VCP, UBQLN2, SQSTM1, TREM2, CHCHD10, TBK1).
Collapse
Affiliation(s)
- Suvi Häkkinen
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Stephanie A Chu
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Suzee E Lee
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
38
|
"Switchboard" malfunction in motor neuron diseases: Selective pathology of thalamic nuclei in amyotrophic lateral sclerosis and primary lateral sclerosis. NEUROIMAGE-CLINICAL 2020; 27:102300. [PMID: 32554322 PMCID: PMC7303672 DOI: 10.1016/j.nicl.2020.102300] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/22/2020] [Accepted: 05/23/2020] [Indexed: 02/06/2023]
Abstract
The thalamus is a key cerebral hub relaying a multitude of corticoefferent and corticoafferent connections and mediating distinct extrapyramidal, sensory, cognitive and behavioural functions. While the thalamus consists of dozens of anatomically well-defined nuclei with distinctive physiological roles, existing imaging studies in motor neuron diseases typically evaluate the thalamus as a single structure. Based on the unique cortical signatures observed in ALS and PLS, we hypothesised that similarly focal thalamic involvement may be observed if the nuclei are individually evaluated. A prospective imaging study was undertaken with 100 patients with ALS, 33 patients with PLS and 117 healthy controls to characterise the integrity of thalamic nuclei. ALS patients were further stratified for the presence of GGGGCC hexanucleotide repeat expansions in C9orf72. The thalamus was segmented into individual nuclei to examine their volumetric profile. Additionally, thalamic shape deformations were evaluated by vertex analyses and focal density alterations were examined by region-of-interest morphometry. Our data indicate that C9orf72 negative ALS patients and PLS patients exhibit ventral lateral and ventral anterior involvement, consistent with the ‘motor’ thalamus. Degeneration of the sensory nuclei was also detected in C9orf72 negative ALS and PLS. Both ALS groups and the PLS cohort showed focal changes in the mediodorsal-paratenial-reuniens nuclei, which mediate memory and executive functions. PLS patients exhibited distinctive thalamic changes with marked pulvinar and lateral geniculate atrophy compared to both controls and C9orf72 negative ALS. The considerable ventral lateral and ventral anterior pathology detected in both ALS and PLS support the emerging literature of extrapyramidal dysfunction in MND. The involvement of sensory nuclei is consistent with sporadic reports of sensory impairment in MND. The unique thalamic signature of PLS is in line with the distinctive clinical features of the phenotype. Our data confirm phenotype-specific patterns of thalamus involvement in motor neuron diseases with the preferential involvement of nuclei mediating motor and cognitive functions. Given the selective involvement of thalamic nuclei in ALS and PLS, future biomarker and natural history studies in MND should evaluate individual thalamic regions instead overall thalamic changes.
Collapse
|
39
|
Trojsi F, D’Alvano G, Bonavita S, Tedeschi G. Genetics and Sex in the Pathogenesis of Amyotrophic Lateral Sclerosis (ALS): Is There a Link? Int J Mol Sci 2020; 21:ijms21103647. [PMID: 32455692 PMCID: PMC7279172 DOI: 10.3390/ijms21103647] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/17/2020] [Accepted: 05/18/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease with no known cure. Approximately 90% of ALS cases are sporadic, although multiple genetic risk factors have been recently revealed also in sporadic ALS (SALS). The pathological expansion of a hexanucleotide repeat in chromosome 9 open reading frame 72 (C9orf72) is the most common genetic mutation identified in familial ALS, detected also in 5–10% of SALS patients. C9orf72-related ALS phenotype appears to be dependent on several modifiers, including demographic factors. Sex has been reported as an independent factor influencing ALS development, with men found to be more susceptible than women. Exposure to both female and male sex hormones have been shown to influence disease risk or progression. Moreover, interplay between genetics and sex has been widely investigated in ALS preclinical models and in large populations of ALS patients carrying C9orf72 repeat expansion. In light of the current need for reclassifying ALS patients into pathologically homogenous subgroups potentially responsive to targeted personalized therapies, we aimed to review the recent literature on the role of genetics and sex as both independent and synergic factors, in the pathophysiology, clinical presentation, and prognosis of ALS. Sex-dependent outcomes may lead to optimizing clinical trials for developing patient-specific therapies for ALS.
Collapse
|
40
|
Ahmed RM, Devenney EM, Strikwerda-Brown C, Hodges JR, Piguet O, Kiernan MC. Phenotypic variability in ALS-FTD and effect on survival. Neurology 2020; 94:e2005-e2013. [PMID: 32277059 DOI: 10.1212/wnl.0000000000009398] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 11/16/2019] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE To determine if survival and cognitive profile is affected by initial presentation in amyotrophic lateral sclerosis-frontotemporal dementia (ALS-FTD) (motor vs cognitive), we compared survival patterns in ALS-FTD based on initial phenotypic presentation and their cognitive profile compared to behavioral variant FTD (bvFTD). METHODS Cognitive/behavioral profiles were examined in 98 patients (59 ALS-FTD and 39 bvFTD). The initial presentation of ALS-FTD was categorized into either motor or cognitive. Survival was calculated from initial symptom onset. MRI brain atrophy patterns were examined using a validated visual rating scale. RESULTS In the ALS-FTD group, 41 (69%) patients were categorized as having an initial cognitive presentation and 18 (31%) a motor presentation. Patients with motor presentation experienced a significantly shorter median survival of 2.7 years compared to 4.4 years (p < 0.001) in those with a cognitive presentation. No differences between motor vs cognitive onset ALS-FTD were found on cognitive testing. When compared to bvFTD, ALS-FTD-cognitive presentation was characterized by reduced language function (p < 0.001), verbal fluency (p = 0.001), and naming (p = 0.007). Both motor and cognitive onset ALS-FTD showed reduced emotion processing (p = 0.01) and exhibited greater motor cortex and dorsal lateral prefrontal cortex atrophy than bvFTD. Increased motor cortex atrophy was associated with 1.5-fold reduction in survival. CONCLUSIONS Initial motor presentation in ALS-FTD leads to faster progression than in those with a cognitive presentation, despite similar overall cognitive deficits. These findings suggest that disease progression in ALS-FTD may be critically linked to physiologic and motor changes.
Collapse
Affiliation(s)
- Rebekah M Ahmed
- From the Memory and Cognition Clinic, Department of Clinical Neurosciences (R.M.A., M.C.K.), Royal Prince Alfred Hospital; Central Sydney Medical School and Brain & Mind Centre (R.M.A., E.M.D., J.R.H., M.C.K.) and School of Psychology and Brain & Mind Centre (C.S.-B., O.P.), The University of Sydney; and ARC Centre of Excellence of Cognition and its Disorders (C.S.-B., O.P.), Sydney, Australia.
| | - Emma M Devenney
- From the Memory and Cognition Clinic, Department of Clinical Neurosciences (R.M.A., M.C.K.), Royal Prince Alfred Hospital; Central Sydney Medical School and Brain & Mind Centre (R.M.A., E.M.D., J.R.H., M.C.K.) and School of Psychology and Brain & Mind Centre (C.S.-B., O.P.), The University of Sydney; and ARC Centre of Excellence of Cognition and its Disorders (C.S.-B., O.P.), Sydney, Australia
| | - Cherie Strikwerda-Brown
- From the Memory and Cognition Clinic, Department of Clinical Neurosciences (R.M.A., M.C.K.), Royal Prince Alfred Hospital; Central Sydney Medical School and Brain & Mind Centre (R.M.A., E.M.D., J.R.H., M.C.K.) and School of Psychology and Brain & Mind Centre (C.S.-B., O.P.), The University of Sydney; and ARC Centre of Excellence of Cognition and its Disorders (C.S.-B., O.P.), Sydney, Australia
| | - John R Hodges
- From the Memory and Cognition Clinic, Department of Clinical Neurosciences (R.M.A., M.C.K.), Royal Prince Alfred Hospital; Central Sydney Medical School and Brain & Mind Centre (R.M.A., E.M.D., J.R.H., M.C.K.) and School of Psychology and Brain & Mind Centre (C.S.-B., O.P.), The University of Sydney; and ARC Centre of Excellence of Cognition and its Disorders (C.S.-B., O.P.), Sydney, Australia
| | - Olivier Piguet
- From the Memory and Cognition Clinic, Department of Clinical Neurosciences (R.M.A., M.C.K.), Royal Prince Alfred Hospital; Central Sydney Medical School and Brain & Mind Centre (R.M.A., E.M.D., J.R.H., M.C.K.) and School of Psychology and Brain & Mind Centre (C.S.-B., O.P.), The University of Sydney; and ARC Centre of Excellence of Cognition and its Disorders (C.S.-B., O.P.), Sydney, Australia
| | - Matthew C Kiernan
- From the Memory and Cognition Clinic, Department of Clinical Neurosciences (R.M.A., M.C.K.), Royal Prince Alfred Hospital; Central Sydney Medical School and Brain & Mind Centre (R.M.A., E.M.D., J.R.H., M.C.K.) and School of Psychology and Brain & Mind Centre (C.S.-B., O.P.), The University of Sydney; and ARC Centre of Excellence of Cognition and its Disorders (C.S.-B., O.P.), Sydney, Australia
| |
Collapse
|
41
|
Montembeault M, Sayah S, Rinaldi D, Le Toullec B, Bertrand A, Funkiewiez A, Saracino D, Camuzat A, Couratier P, Chouly M, Hannequin D, Aubier-Girard C, Pasquier F, Delbeuck X, Colliot O, Batrancourt B, Azuar C, Lévy R, Dubois B, Le Ber I, Migliaccio R. Cognitive inhibition impairments in presymptomatic C9orf72 carriers. J Neurol Neurosurg Psychiatry 2020; 91:366-372. [PMID: 32054668 DOI: 10.1136/jnnp-2019-322242] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 01/13/2020] [Accepted: 01/15/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To investigate cognitive inhibition in presymptomatic C9orf72 mutation carriers (C9+) and its associated neuroanatomical correlates. METHODS Thirty-eight presymptomatic C9orf72 mutation carriers (C9+, mean age 38.2±8.0 years) and 22 C9- controls from the PREV-DEMALS cohort were included in this study. They underwent a cognitive inhibition assessment with the Hayling Sentence Completion Test (HSCT; time to completion (part B-part A); error score in part B) as well as a 3D MRI. RESULTS C9+ individuals younger than 40 years had higher error scores (part B) but equivalent HSCT time to completion (part B-part A) compared to C9- individuals. C9+ individuals older than 40 years had both higher error scores and longer time to completion. HSCT time to completion significantly predicted the proximity to estimated clinical conversion from presymptomatic to symptomatic phase in C9+ individuals (based on the average age at onset of affected relatives in the family). Anatomically, we found that HSCT time to completion was associated with the integrity of the cerebellum. CONCLUSION The HSCT represents a good marker of cognitive inhibition impairments in C9+ and of proximity to clinical conversion. This study also highlights the key role of the cerebellum in cognitive inhibition.
Collapse
Affiliation(s)
- Maxime Montembeault
- Inserm U1127, Institut du Cerveau, Hôpital Pitié-Salpêtrière, Paris, France.,Sorbonne University, Paris, France.,FrontLab, Paris, France.,Department of Neurology, University of California San Francisco, Memory and Aging Center, San Francisco, California, USA
| | - Sabrina Sayah
- Inserm U1127, Institut du Cerveau, Hôpital Pitié-Salpêtrière, Paris, France
| | - Daisy Rinaldi
- Inserm U1127, Institut du Cerveau, Hôpital Pitié-Salpêtrière, Paris, France.,Reference Centre for Rare of Early Onset Dementias, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Benjamin Le Toullec
- Inserm U1127, Institut du Cerveau, Hôpital Pitié-Salpêtrière, Paris, France.,Reference Centre for Rare of Early Onset Dementias, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Anne Bertrand
- Sorbonne University, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie Paris 06, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Institut du Cerveau et la Moelle Épinière, FrontLAB, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France.,Aramis Project Team, Inria Research Center of Paris, Paris, France
| | - Aurélie Funkiewiez
- Reference Centre for Rare of Early Onset Dementias, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France.,Institute of Memory and Alzheimer's Disease, Centre of Excellence of Neurodegenerative Disease, Department of Neurology, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Dario Saracino
- Inserm U1127, Institut du Cerveau, Hôpital Pitié-Salpêtrière, Paris, France.,Reference Centre for Rare of Early Onset Dementias, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France.,Aramis Project Team, Inria Research Center of Paris, Paris, France
| | - Agnès Camuzat
- Inserm U1127, Institut du Cerveau, Hôpital Pitié-Salpêtrière, Paris, France
| | - Philippe Couratier
- Centre de Référence SLA et autres maladies du motoneurone, Centre Hospitalier Universitaire de Limoges, Limoges, France.,Centre de Compétence Démences Rares, Centre Hospitalier Universitaire de Limoges, Limoges, France
| | - Marianne Chouly
- Centre de Référence SLA et autres maladies du motoneurone, Centre Hospitalier Universitaire de Limoges, Limoges, France.,Centre de Compétence Démences Rares, Centre Hospitalier Universitaire de Limoges, Limoges, France
| | - Didier Hannequin
- Centre National de Référence pour les Malades Alzheimer Jeunes, Centre Hospitalier Universitaire de Rouen, INSERM 1245, Rouen, France.,Department of Neurology, Centre Hospitalier Universitaire de Rouen, Rouen, France
| | - Carole Aubier-Girard
- Centre National de Référence pour les Malades Alzheimer Jeunes, Centre Hospitalier Universitaire de Rouen, INSERM 1245, Rouen, France.,Department of Neurology, Centre Hospitalier Universitaire de Rouen, Rouen, France
| | - Florence Pasquier
- Université de Lille, INSERM U1171, Centre de la mémoire (CMRR), Centre national de référence pour les malades Alzheimer jeunes (CNRMAJ), CHU Lille, Development of Innovative Strategies for a Transdisciplinary approach to ALZheimer's disease (DistAlz), Lille, France
| | - Xavier Delbeuck
- Université de Lille, INSERM U1171, Centre de la mémoire (CMRR), Centre national de référence pour les malades Alzheimer jeunes (CNRMAJ), CHU Lille, Development of Innovative Strategies for a Transdisciplinary approach to ALZheimer's disease (DistAlz), Lille, France
| | - Olivier Colliot
- Inserm U1127, Institut du Cerveau, Hôpital Pitié-Salpêtrière, Paris, France.,Aramis Project Team, Inria Research Center of Paris, Paris, France
| | - Bénédicte Batrancourt
- Inserm U1127, Institut du Cerveau, Hôpital Pitié-Salpêtrière, Paris, France.,Sorbonne University, Paris, France.,FrontLab, Paris, France
| | - Carole Azuar
- Inserm U1127, Institut du Cerveau, Hôpital Pitié-Salpêtrière, Paris, France.,FrontLab, Paris, France.,Reference Centre for Rare of Early Onset Dementias, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Richard Lévy
- Inserm U1127, Institut du Cerveau, Hôpital Pitié-Salpêtrière, Paris, France.,Sorbonne University, Paris, France.,FrontLab, Paris, France.,Reference Centre for Rare of Early Onset Dementias, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Bruno Dubois
- Inserm U1127, Institut du Cerveau, Hôpital Pitié-Salpêtrière, Paris, France.,Sorbonne University, Paris, France.,Reference Centre for Rare of Early Onset Dementias, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Isabelle Le Ber
- Inserm U1127, Institut du Cerveau, Hôpital Pitié-Salpêtrière, Paris, France.,Sorbonne University, Paris, France.,FrontLab, Paris, France.,Reference Centre for Rare of Early Onset Dementias, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Raffaella Migliaccio
- Inserm U1127, Institut du Cerveau, Hôpital Pitié-Salpêtrière, Paris, France .,Sorbonne University, Paris, France.,FrontLab, Paris, France.,Reference Centre for Rare of Early Onset Dementias, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | | |
Collapse
|
42
|
Gosselt IK, Nijboer TCW, Van Es MA. An overview of screening instruments for cognition and behavior in patients with ALS: selecting the appropriate tool for clinical practice. Amyotroph Lateral Scler Frontotemporal Degener 2020; 21:324-336. [PMID: 32157912 DOI: 10.1080/21678421.2020.1732424] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Objective: Patients with amyotrophic lateral sclerosis (ALS) not only show motor deficits, but may also have cognitive and/or behavioral impairments. Recognizing these impairments is crucial as they are associated with lower quality of life, shorter survival, and increased caregiver burden. Therefore, ALS-specific neuropsychological screening instruments have been developed that can account for motor and speech difficulties. This study provides an overview and comparison of these screeners. Methods: A systematic review was conducted using Medline and Embase. Articles describing cognitive/behavioral screening instruments assessed in ALS patients were included. Screening instruments were compared on multiple factors, such as domains, adaptability, required time, and validation. Results: We included 99 articles, reporting on nine cognitive screeners (i.e. ACE-R, ALS-BCA, ALS-CBS, ECAS, FAB, MMSE, MoCA, PSSFTS, and UCSF-SB), of which five ALS-specific. Furthermore, eight behavioral screeners (i.e. ALS-FTD-Q, AES, BBI, DAS, FBI, FrSBe, MiND-B, and NPI) were reported on, of which three ALS-specific. Conclusion: Considering the broad range of cognitive domains, adaptability, and satisfying validity, the ALS-CBS and ECAS appear to be the most suitable screeners to detect cognitive and behavioral changes in ALS. The BBI appears to be the best option to screen for behavioral changes in ALS, since all relevant domains are assessed, motor-related problems are considered, and has a satisfactory validity. The MiND-B and ALS-FTD-Q are promising as well. In general, all screening instruments would benefit from additional validation research to gain greater insights into test characteristics and to aid clinicians in selecting screening tools for use in clinical practice.
Collapse
Affiliation(s)
- Isabel K Gosselt
- Center of Excellence for Rehabilitation Medicine, UMC Utrecht Brain Center, University Medical Center Utrecht, and De Hoogstraat Rehabilitation, Utrecht, Netherlands
| | - Tanja C W Nijboer
- Center of Excellence for Rehabilitation Medicine, UMC Utrecht Brain Center, University Medical Center Utrecht, and De Hoogstraat Rehabilitation, Utrecht, Netherlands.,Department of Experimental Psychology, Helmholtz Institute, Utrecht University, Utrecht, The Netherlands
| | - Michael A Van Es
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
43
|
Hanstock C, Sun K, Choi C, Eurich D, Camicioli R, Johnston W, Kalra S. Spectroscopic markers of neurodegeneration in the mesial prefrontal cortex predict survival in ALS. Amyotroph Lateral Scler Frontotemporal Degener 2020; 21:246-251. [PMID: 32067510 DOI: 10.1080/21678421.2020.1727926] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background and objective: N-acetylaspartate (NAA) and myo-inositol (mIns) are spectroscopic markers of neuronal integrity and astrogliosis, respectively. We performed a survival analysis to determine the prognostic value of the NAA/mIns metabolite ratio in ALS after a period of two and five years. Methods: Twenty-four patients with ALS (two with ALS-FTD) were recruited to participate in a high-field MR spectroscopy study of the mesial prefrontal cortex. Univariate and multivariate Cox proportional hazards analyses were used to assess NAA/mIns as a predictor of survival alongside other demographic and clinical measures. Census dates were set at two and five years after the time of MR scan for each patient. Survival curves were calculated using the Kaplan-Meier method. Results: After a five-year observation period, 19 patients had died and five were still alive. Median survival time from date of scan was 1.95 years. Univariate and multivariate Cox analysis showed NAA/mIns to be a significant independent predictor of survival at two years after scanning, but not at five years. Conclusion: Cerebral degeneration in the mesial prefrontal cortex as detected by the NAA/mIns metabolite ratio is predictive of survival in ALS in a time-dependent manner.
Collapse
Affiliation(s)
- Chris Hanstock
- Department of Biomedical Engineering, University of Alberta, Edmonton, AB, Canada
| | - Kerry Sun
- Department of Medicine, Division of Neurology, University of Alberta, Edmonton, AB, Canada
| | - Changho Choi
- South-Western Medical Center, University of Texas, Dallas, TX, USA
| | - Dean Eurich
- School of Public Health, University of Alberta, Edmonton, AB, Canada, and
| | - Richard Camicioli
- Department of Medicine, Division of Neurology, University of Alberta, Edmonton, AB, Canada
| | - Wendy Johnston
- Department of Medicine, Division of Neurology, University of Alberta, Edmonton, AB, Canada
| | - Sanjay Kalra
- Department of Biomedical Engineering, University of Alberta, Edmonton, AB, Canada.,Department of Medicine, Division of Neurology, University of Alberta, Edmonton, AB, Canada.,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
44
|
Moore KM, Nicholas J, Grossman M, McMillan CT, Irwin DJ, Massimo L, Van Deerlin VM, Warren JD, Fox NC, Rossor MN, Mead S, Bocchetta M, Boeve BF, Knopman DS, Graff-Radford NR, Forsberg LK, Rademakers R, Wszolek ZK, van Swieten JC, Jiskoot LC, Meeter LH, Dopper EG, Papma JM, Snowden JS, Saxon J, Jones M, Pickering-Brown S, Le Ber I, Camuzat A, Brice A, Caroppo P, Ghidoni R, Pievani M, Benussi L, Binetti G, Dickerson BC, Lucente D, Krivensky S, Graff C, Öijerstedt L, Fallström M, Thonberg H, Ghoshal N, Morris JC, Borroni B, Benussi A, Padovani A, Galimberti D, Scarpini E, Fumagalli GG, Mackenzie IR, Hsiung GYR, Sengdy P, Boxer AL, Rosen H, Taylor JB, Synofzik M, Wilke C, Sulzer P, Hodges JR, Halliday G, Kwok J, Sanchez-Valle R, Lladó A, Borrego-Ecija S, Santana I, Almeida MR, Tábuas-Pereira M, Moreno F, Barandiaran M, Indakoetxea B, Levin J, Danek A, Rowe JB, Cope TE, Otto M, Anderl-Straub S, de Mendonça A, Maruta C, Masellis M, Black SE, Couratier P, Lautrette G, Huey ED, Sorbi S, Nacmias B, Laforce R, Tremblay MPL, Vandenberghe R, Damme PV, Rogalski EJ, Weintraub S, Gerhard A, Onyike CU, Ducharme S, Papageorgiou SG, Ng ASL, Brodtmann A, Finger E, Guerreiro R, Bras J, Rohrer JD. Age at symptom onset and death and disease duration in genetic frontotemporal dementia: an international retrospective cohort study. Lancet Neurol 2020; 19:145-156. [PMID: 31810826 PMCID: PMC7007771 DOI: 10.1016/s1474-4422(19)30394-1] [Citation(s) in RCA: 167] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 09/04/2019] [Accepted: 09/13/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Frontotemporal dementia is a heterogenous neurodegenerative disorder, with about a third of cases being genetic. Most of this genetic component is accounted for by mutations in GRN, MAPT, and C9orf72. In this study, we aimed to complement previous phenotypic studies by doing an international study of age at symptom onset, age at death, and disease duration in individuals with mutations in GRN, MAPT, and C9orf72. METHODS In this international, retrospective cohort study, we collected data on age at symptom onset, age at death, and disease duration for patients with pathogenic mutations in the GRN and MAPT genes and pathological expansions in the C9orf72 gene through the Frontotemporal Dementia Prevention Initiative and from published papers. We used mixed effects models to explore differences in age at onset, age at death, and disease duration between genetic groups and individual mutations. We also assessed correlations between the age at onset and at death of each individual and the age at onset and at death of their parents and the mean age at onset and at death of their family members. Lastly, we used mixed effects models to investigate the extent to which variability in age at onset and at death could be accounted for by family membership and the specific mutation carried. FINDINGS Data were available from 3403 individuals from 1492 families: 1433 with C9orf72 expansions (755 families), 1179 with GRN mutations (483 families, 130 different mutations), and 791 with MAPT mutations (254 families, 67 different mutations). Mean age at symptom onset and at death was 49·5 years (SD 10·0; onset) and 58·5 years (11·3; death) in the MAPT group, 58·2 years (9·8; onset) and 65·3 years (10·9; death) in the C9orf72 group, and 61·3 years (8·8; onset) and 68·8 years (9·7; death) in the GRN group. Mean disease duration was 6·4 years (SD 4·9) in the C9orf72 group, 7·1 years (3·9) in the GRN group, and 9·3 years (6·4) in the MAPT group. Individual age at onset and at death was significantly correlated with both parental age at onset and at death and with mean family age at onset and at death in all three groups, with a stronger correlation observed in the MAPT group (r=0·45 between individual and parental age at onset, r=0·63 between individual and mean family age at onset, r=0·58 between individual and parental age at death, and r=0·69 between individual and mean family age at death) than in either the C9orf72 group (r=0·32 individual and parental age at onset, r=0·36 individual and mean family age at onset, r=0·38 individual and parental age at death, and r=0·40 individual and mean family age at death) or the GRN group (r=0·22 individual and parental age at onset, r=0·18 individual and mean family age at onset, r=0·22 individual and parental age at death, and r=0·32 individual and mean family age at death). Modelling showed that the variability in age at onset and at death in the MAPT group was explained partly by the specific mutation (48%, 95% CI 35-62, for age at onset; 61%, 47-73, for age at death), and even more by family membership (66%, 56-75, for age at onset; 74%, 65-82, for age at death). In the GRN group, only 2% (0-10) of the variability of age at onset and 9% (3-21) of that of age of death was explained by the specific mutation, whereas 14% (9-22) of the variability of age at onset and 20% (12-30) of that of age at death was explained by family membership. In the C9orf72 group, family membership explained 17% (11-26) of the variability of age at onset and 19% (12-29) of that of age at death. INTERPRETATION Our study showed that age at symptom onset and at death of people with genetic frontotemporal dementia is influenced by genetic group and, particularly for MAPT mutations, by the specific mutation carried and by family membership. Although estimation of age at onset will be an important factor in future pre-symptomatic therapeutic trials for all three genetic groups, our study suggests that data from other members of the family will be particularly helpful only for individuals with MAPT mutations. Further work in identifying both genetic and environmental factors that modify phenotype in all groups will be important to improve such estimates. FUNDING UK Medical Research Council, National Institute for Health Research, and Alzheimer's Society.
Collapse
Affiliation(s)
- Katrina M Moore
- Dementia Research Centre, Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, UK
| | - Jennifer Nicholas
- Department of Medical Statistics, London School of Hygiene & Tropical Medicine, London, UK
| | - Murray Grossman
- Department of Neurology, Penn Frontotemporal Degeneration Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Corey T McMillan
- Department of Neurology, Penn Frontotemporal Degeneration Center, University of Pennsylvania, Philadelphia, PA, USA
| | - David J Irwin
- Department of Neurology, Penn Frontotemporal Degeneration Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Lauren Massimo
- Department of Neurology, Penn Frontotemporal Degeneration Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Vivianna M Van Deerlin
- Department of Neurology, Penn Frontotemporal Degeneration Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Jason D Warren
- Dementia Research Centre, Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, UK
| | - Nick C Fox
- Dementia Research Centre, Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, UK
| | - Martin N Rossor
- Dementia Research Centre, Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, UK
| | - Simon Mead
- Institute of Prion Diseases, University College London, London, UK
| | - Martina Bocchetta
- Dementia Research Centre, Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, UK
| | | | | | | | | | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | | | - Lize C Jiskoot
- Department of Neurology, Erasmus Medical Centre, Rotterdam, Netherlands
| | - Lieke H Meeter
- Department of Neurology, Erasmus Medical Centre, Rotterdam, Netherlands
| | - Elise Gp Dopper
- Department of Neurology, Erasmus Medical Centre, Rotterdam, Netherlands
| | - Janne M Papma
- Department of Neurology, Erasmus Medical Centre, Rotterdam, Netherlands
| | - Julie S Snowden
- Cerebral Function Unit, Salford Royal NHS Foundation Trust and Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, UK
| | - Jennifer Saxon
- Cerebral Function Unit, Salford Royal NHS Foundation Trust and Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, UK
| | - Matthew Jones
- Cerebral Function Unit, Salford Royal NHS Foundation Trust and Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, UK
| | - Stuart Pickering-Brown
- Cerebral Function Unit, Salford Royal NHS Foundation Trust and Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, UK
| | - Isabelle Le Ber
- Institut du Cerveau et de la Moelle épinière & Centre de Référence des Démences Rares ou précoces, Institut de la Mémoire et de la Maladie d'Alzheimer, Assistance Publique-Hôpitaux de Paris, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Agnès Camuzat
- Institut du Cerveau et de la Moelle épinière & Centre de Référence des Démences Rares ou précoces, Institut de la Mémoire et de la Maladie d'Alzheimer, Assistance Publique-Hôpitaux de Paris, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Alexis Brice
- Institut du Cerveau et de la Moelle épinière & Centre de Référence des Démences Rares ou précoces, Institut de la Mémoire et de la Maladie d'Alzheimer, Assistance Publique-Hôpitaux de Paris, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Paola Caroppo
- Institut du Cerveau et de la Moelle épinière & Centre de Référence des Démences Rares ou précoces, Institut de la Mémoire et de la Maladie d'Alzheimer, Assistance Publique-Hôpitaux de Paris, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Roberta Ghidoni
- Molecular Markers Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Michela Pievani
- Alzheimer's Neuroimaging & Epidemiology Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Luisa Benussi
- Molecular Markers Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Giuliano Binetti
- Molecular Markers Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Bradford C Dickerson
- Frontotemporal Disorders Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Diane Lucente
- Frontotemporal Disorders Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Samantha Krivensky
- Frontotemporal Disorders Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Caroline Graff
- Center for Alzheimer Research, Division of Neurogenetics, Department of Neurobiology, Care Sciences and Society, Bioclinicum, Karolinska Institutet, Solna, Sweden; Unit for Hereditary Dementias, Theme Aging, Karolinska University Hospital, Solna, Sweden
| | - Linn Öijerstedt
- Center for Alzheimer Research, Division of Neurogenetics, Department of Neurobiology, Care Sciences and Society, Bioclinicum, Karolinska Institutet, Solna, Sweden; Unit for Hereditary Dementias, Theme Aging, Karolinska University Hospital, Solna, Sweden
| | - Marie Fallström
- Center for Alzheimer Research, Division of Neurogenetics, Department of Neurobiology, Care Sciences and Society, Bioclinicum, Karolinska Institutet, Solna, Sweden; Unit for Hereditary Dementias, Theme Aging, Karolinska University Hospital, Solna, Sweden
| | - Håkan Thonberg
- Center for Alzheimer Research, Division of Neurogenetics, Department of Neurobiology, Care Sciences and Society, Bioclinicum, Karolinska Institutet, Solna, Sweden; Unit for Hereditary Dementias, Theme Aging, Karolinska University Hospital, Solna, Sweden
| | - Nupur Ghoshal
- Department of Neurology, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St Louis, MO, USA
| | - John C Morris
- Department of Neurology, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St Louis, MO, USA
| | - Barbara Borroni
- Centre for Neurodegenerative Disorders, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Alberto Benussi
- Centre for Neurodegenerative Disorders, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Alessandro Padovani
- Centre for Neurodegenerative Disorders, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Daniela Galimberti
- Department of Biomedical, Surgical and Dental Sciences, Centro Dino Ferrari, University of Milan, Milan, Italy; Istituto di Ricovero e Cura a Carattere Scientifico Fondazione Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Elio Scarpini
- Department of Pathophysiology and Transplantation, Centro Dino Ferrari, University of Milan, Milan, Italy; Istituto di Ricovero e Cura a Carattere Scientifico Fondazione Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Giorgio G Fumagalli
- Department of Pathophysiology and Transplantation, Centro Dino Ferrari, University of Milan, Milan, Italy; Istituto di Ricovero e Cura a Carattere Scientifico Fondazione Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy; Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Ian R Mackenzie
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Ging-Yuek R Hsiung
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Pheth Sengdy
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Adam L Boxer
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Howie Rosen
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Joanne B Taylor
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Matthis Synofzik
- Department of Neurodegenerative Diseases, Center for Neurology and Hertie-Institute for Clinical Brain Research, Tübingen, Germany; German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Carlo Wilke
- Department of Neurodegenerative Diseases, Center for Neurology and Hertie-Institute for Clinical Brain Research, Tübingen, Germany; German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Patricia Sulzer
- Department of Neurodegenerative Diseases, Center for Neurology and Hertie-Institute for Clinical Brain Research, Tübingen, Germany; German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - John R Hodges
- Brain and Mind Centre & Central Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Glenda Halliday
- Brain and Mind Centre & Central Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - John Kwok
- Brain and Mind Centre & Central Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Raquel Sanchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Albert Lladó
- Alzheimer's Disease and Other Cognitive Disorders Unit, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Sergi Borrego-Ecija
- Alzheimer's Disease and Other Cognitive Disorders Unit, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Isabel Santana
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | | | - Miguel Tábuas-Pereira
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Fermin Moreno
- Cognitive Disorders Unit, Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neuroscience Area, Biodonostia Health Research Institute, San Sebastian, Spain; Center for Networked Biomedical Research on Neurodegenerative Disease, Carlos III Health Institute, Madrid, Spain
| | - Myriam Barandiaran
- Cognitive Disorders Unit, Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neuroscience Area, Biodonostia Health Research Institute, San Sebastian, Spain; Center for Networked Biomedical Research on Neurodegenerative Disease, Carlos III Health Institute, Madrid, Spain
| | - Begoña Indakoetxea
- Cognitive Disorders Unit, Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neuroscience Area, Biodonostia Health Research Institute, San Sebastian, Spain; Center for Networked Biomedical Research on Neurodegenerative Disease, Carlos III Health Institute, Madrid, Spain
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany; German Center for Neurodegenerative Diseases, Munich, Germany; Munich Cluster for Systems Neurology, Munich, Germany
| | - Adrian Danek
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - James B Rowe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Thomas E Cope
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany
| | | | | | | | - Mario Masellis
- Division of Neurology, Department of Medicine, Hurvitz Brain Sciences Program, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Sandra E Black
- Division of Neurology, Department of Medicine, Hurvitz Brain Sciences Program, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Philippe Couratier
- Centre de Compétence Démences Rares, Centre Hospitalier et Universitaire Limoges, Limoges, France
| | - Geraldine Lautrette
- Centre de Compétence Démences Rares, Centre Hospitalier et Universitaire Limoges, Limoges, France
| | - Edward D Huey
- Departments of Psychiatry and Neurology, Columbia University, New York, NY, USA
| | - Sandro Sorbi
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy; Istituto di Ricovero e Cura a Carattere Scientifico Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Benedetta Nacmias
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Robert Laforce
- Clinique Interdisciplinaire de Mémoire, Département des Sciences Neurologiques, Hôpital de l'Enfant-Jésus, and Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Marie-Pier L Tremblay
- Clinique Interdisciplinaire de Mémoire, Département des Sciences Neurologiques, Hôpital de l'Enfant-Jésus, and Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Rik Vandenberghe
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Philip Van Damme
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium; Center for Brain & Disease Research, VIB, Leuven, Belgium
| | - Emily J Rogalski
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University, Chicago, IL, USA
| | - Sandra Weintraub
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University, Chicago, IL, USA
| | - Alexander Gerhard
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK; Departments of Nuclear Medicine and Geriatric Medicine, University Hospital Essen, Essen, Germany
| | - Chiadi U Onyike
- Division of Geriatric Psychiatry and Neuropsychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Simon Ducharme
- Montreal Neurological Institute, McConnell Brain Imaging Centre, McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Sokratis G Papageorgiou
- Cognitive Disorders/Dementia Unit, 2nd Department of Neurology, National and Kapodistrian University of Athens, Attikon University General Hospital, Athens, Greece
| | - Adeline Su Lyn Ng
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore
| | - Amy Brodtmann
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | - Elizabeth Finger
- Department of Clinical Neurological Sciences, University of Western Ontario, London, ON, Canada
| | - Rita Guerreiro
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Jose Bras
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, UK.
| |
Collapse
|
45
|
Kawakami I, Arai T, Hasegawa M. The basis of clinicopathological heterogeneity in TDP-43 proteinopathy. Acta Neuropathol 2019; 138:751-770. [PMID: 31555895 PMCID: PMC6800885 DOI: 10.1007/s00401-019-02077-x] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 08/27/2019] [Accepted: 09/13/2019] [Indexed: 12/15/2022]
Abstract
Transactive response DNA-binding protein 43 kDa (TDP-43) was identified as a major disease-associated component in the brain of patients with amyotrophic lateral sclerosis (ALS), as well as the largest subset of patients with frontotemporal lobar degeneration with ubiquitinated inclusions (FTLD-U), which characteristically exhibits cytoplasmic inclusions that are positive for ubiquitin but negative for tau and α-synuclein. TDP-43 pathology occurs in distinct brain regions, involves disparate brain networks, and features accumulation of misfolded proteins in various cell types and in different neuroanatomical regions. The clinical phenotypes of ALS and FTLD-TDP (FTLD with abnormal intracellular accumulations of TDP-43) correlate with characteristic distribution patterns of the underlying pathology across specific brain regions with disease progression. Recent studies support the idea that pathological protein spreads from neuron to neuron via axonal transport in a hierarchical manner. However, little is known to date about the basis of the selective cellular and regional vulnerability, although the information would have important implications for the development of targeted and personalized therapies. Here, we aim to summarize recent advances in the neuropathology, genetics and animal models of TDP-43 proteinopathy, and their relationship to clinical phenotypes for the underlying selective neuronal and regional susceptibilities. Finally, we attempt to integrate these findings into the emerging picture of TDP-43 proteinopathy, and to highlight key issues for future therapy and research.
Collapse
Affiliation(s)
- Ito Kawakami
- Dementia Research Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6, Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan
- Department of Neuropathology, Tokyo Metropolitan Geriatric Hospital and Institute, Tokyo, Japan
| | - Tetsuaki Arai
- Dementia Research Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6, Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan.
- Department of Psychiatry, Division of Clinical Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.
| | - Masato Hasegawa
- Dementia Research Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6, Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan.
| |
Collapse
|
46
|
Cammack AJ, Atassi N, Hyman T, van den Berg LH, Harms M, Baloh RH, Brown RH, van Es MA, Veldink JH, de Vries BS, Rothstein JD, Drain C, Jockel-Balsarotti J, Malcolm A, Boodram S, Salter A, Wightman N, Yu H, Sherman AV, Esparza TJ, McKenna-Yasek D, Owegi MA, Douthwright C, McCampbell A, Ferguson T, Cruchaga C, Cudkowicz M, Miller TM. Prospective natural history study of C9orf72 ALS clinical characteristics and biomarkers. Neurology 2019; 93:e1605-e1617. [PMID: 31578300 DOI: 10.1212/wnl.0000000000008359] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 05/20/2019] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE To define the natural history of the C9orf72 amyotrophic lateral sclerosis (C9ALS) patient population, develop disease biomarkers, and characterize patient pathologies. METHODS We prospectively collected clinical and demographic data from 116 symptomatic C9ALS and 12 non-amyotrophic lateral sclerosis (ALS) full expansion carriers across 7 institutions in the United States and the Netherlands. In addition, we collected blood samples for DNA repeat size assessment, CSF samples for biomarker identification, and autopsy samples for dipeptide repeat protein (DPR) size determination. Finally, we collected retrospective clinical data via chart review from 208 individuals with C9ALS and 450 individuals with singleton ALS. RESULTS The mean age at onset in the symptomatic prospective cohort was 57.9 ± 8.3 years, and median duration of survival after onset was 36.9 months. The monthly change was -1.8 ± 1.7 for ALS Functional Rating Scale-Revised and -1.4% ± 3.24% of predicted for slow vital capacity. In blood DNA, we found that G4C2 repeat size correlates positively with age. In CSF, we observed that concentrations of poly(GP) negatively correlate with DNA expansion size but do not correlate with measures of disease progression. Finally, we found that size of poly(GP) dipeptides in the brain can reach large sizes similar to that of their DNA repeat derivatives. CONCLUSIONS We present a thorough investigation of C9ALS natural history, providing the basis for C9ALS clinical trial design. We found that clinical features of this genetic subset are less variant than in singleton ALS. In addition, we identified important correlations of C9ALS patient pathologies with clinical and demographic data.
Collapse
Affiliation(s)
- Alexander J Cammack
- From the Department of Neurology (A.J.C., T.H., C.D., J.J.-B., A.M., S.B., A.S., T.J.E., C.C., T.M.M.), Washington University School of Medicine, St. Louis, MO; Department of Neurology (N.A., H.Y., A.V.S., M.C.), Neurological Clinical Research Institute, Massachusetts General Hospital, Boston; Department of Neurology (L.H.v.d.B., M.A.v.E., J.H.V., B.S.d.V.), Brain Center Rudolf Magnus, University Medical Center Utrecht, University Utrecht, the Netherlands; Department of Neurology (M.H.), Columbia University, New York, NY; Department of Neurology (R.H. Baloh), Cedars-Sinai Medical Center, Los Angeles, CA; Department of Neurology (R.H. Brown, N.W., D.M.-Y., M.A.O., C.D.), University of Massachusetts, Worcester; Department of Neurology (J.D.R.), Johns Hopkins University, Baltimore, MD; and Biogen Inc. (A.M., T.F.), Boston, MA
| | - Nazem Atassi
- From the Department of Neurology (A.J.C., T.H., C.D., J.J.-B., A.M., S.B., A.S., T.J.E., C.C., T.M.M.), Washington University School of Medicine, St. Louis, MO; Department of Neurology (N.A., H.Y., A.V.S., M.C.), Neurological Clinical Research Institute, Massachusetts General Hospital, Boston; Department of Neurology (L.H.v.d.B., M.A.v.E., J.H.V., B.S.d.V.), Brain Center Rudolf Magnus, University Medical Center Utrecht, University Utrecht, the Netherlands; Department of Neurology (M.H.), Columbia University, New York, NY; Department of Neurology (R.H. Baloh), Cedars-Sinai Medical Center, Los Angeles, CA; Department of Neurology (R.H. Brown, N.W., D.M.-Y., M.A.O., C.D.), University of Massachusetts, Worcester; Department of Neurology (J.D.R.), Johns Hopkins University, Baltimore, MD; and Biogen Inc. (A.M., T.F.), Boston, MA
| | - Theodore Hyman
- From the Department of Neurology (A.J.C., T.H., C.D., J.J.-B., A.M., S.B., A.S., T.J.E., C.C., T.M.M.), Washington University School of Medicine, St. Louis, MO; Department of Neurology (N.A., H.Y., A.V.S., M.C.), Neurological Clinical Research Institute, Massachusetts General Hospital, Boston; Department of Neurology (L.H.v.d.B., M.A.v.E., J.H.V., B.S.d.V.), Brain Center Rudolf Magnus, University Medical Center Utrecht, University Utrecht, the Netherlands; Department of Neurology (M.H.), Columbia University, New York, NY; Department of Neurology (R.H. Baloh), Cedars-Sinai Medical Center, Los Angeles, CA; Department of Neurology (R.H. Brown, N.W., D.M.-Y., M.A.O., C.D.), University of Massachusetts, Worcester; Department of Neurology (J.D.R.), Johns Hopkins University, Baltimore, MD; and Biogen Inc. (A.M., T.F.), Boston, MA
| | - Leonard H van den Berg
- From the Department of Neurology (A.J.C., T.H., C.D., J.J.-B., A.M., S.B., A.S., T.J.E., C.C., T.M.M.), Washington University School of Medicine, St. Louis, MO; Department of Neurology (N.A., H.Y., A.V.S., M.C.), Neurological Clinical Research Institute, Massachusetts General Hospital, Boston; Department of Neurology (L.H.v.d.B., M.A.v.E., J.H.V., B.S.d.V.), Brain Center Rudolf Magnus, University Medical Center Utrecht, University Utrecht, the Netherlands; Department of Neurology (M.H.), Columbia University, New York, NY; Department of Neurology (R.H. Baloh), Cedars-Sinai Medical Center, Los Angeles, CA; Department of Neurology (R.H. Brown, N.W., D.M.-Y., M.A.O., C.D.), University of Massachusetts, Worcester; Department of Neurology (J.D.R.), Johns Hopkins University, Baltimore, MD; and Biogen Inc. (A.M., T.F.), Boston, MA
| | - Matthew Harms
- From the Department of Neurology (A.J.C., T.H., C.D., J.J.-B., A.M., S.B., A.S., T.J.E., C.C., T.M.M.), Washington University School of Medicine, St. Louis, MO; Department of Neurology (N.A., H.Y., A.V.S., M.C.), Neurological Clinical Research Institute, Massachusetts General Hospital, Boston; Department of Neurology (L.H.v.d.B., M.A.v.E., J.H.V., B.S.d.V.), Brain Center Rudolf Magnus, University Medical Center Utrecht, University Utrecht, the Netherlands; Department of Neurology (M.H.), Columbia University, New York, NY; Department of Neurology (R.H. Baloh), Cedars-Sinai Medical Center, Los Angeles, CA; Department of Neurology (R.H. Brown, N.W., D.M.-Y., M.A.O., C.D.), University of Massachusetts, Worcester; Department of Neurology (J.D.R.), Johns Hopkins University, Baltimore, MD; and Biogen Inc. (A.M., T.F.), Boston, MA
| | - Robert H Baloh
- From the Department of Neurology (A.J.C., T.H., C.D., J.J.-B., A.M., S.B., A.S., T.J.E., C.C., T.M.M.), Washington University School of Medicine, St. Louis, MO; Department of Neurology (N.A., H.Y., A.V.S., M.C.), Neurological Clinical Research Institute, Massachusetts General Hospital, Boston; Department of Neurology (L.H.v.d.B., M.A.v.E., J.H.V., B.S.d.V.), Brain Center Rudolf Magnus, University Medical Center Utrecht, University Utrecht, the Netherlands; Department of Neurology (M.H.), Columbia University, New York, NY; Department of Neurology (R.H. Baloh), Cedars-Sinai Medical Center, Los Angeles, CA; Department of Neurology (R.H. Brown, N.W., D.M.-Y., M.A.O., C.D.), University of Massachusetts, Worcester; Department of Neurology (J.D.R.), Johns Hopkins University, Baltimore, MD; and Biogen Inc. (A.M., T.F.), Boston, MA
| | - Robert H Brown
- From the Department of Neurology (A.J.C., T.H., C.D., J.J.-B., A.M., S.B., A.S., T.J.E., C.C., T.M.M.), Washington University School of Medicine, St. Louis, MO; Department of Neurology (N.A., H.Y., A.V.S., M.C.), Neurological Clinical Research Institute, Massachusetts General Hospital, Boston; Department of Neurology (L.H.v.d.B., M.A.v.E., J.H.V., B.S.d.V.), Brain Center Rudolf Magnus, University Medical Center Utrecht, University Utrecht, the Netherlands; Department of Neurology (M.H.), Columbia University, New York, NY; Department of Neurology (R.H. Baloh), Cedars-Sinai Medical Center, Los Angeles, CA; Department of Neurology (R.H. Brown, N.W., D.M.-Y., M.A.O., C.D.), University of Massachusetts, Worcester; Department of Neurology (J.D.R.), Johns Hopkins University, Baltimore, MD; and Biogen Inc. (A.M., T.F.), Boston, MA
| | - Michael A van Es
- From the Department of Neurology (A.J.C., T.H., C.D., J.J.-B., A.M., S.B., A.S., T.J.E., C.C., T.M.M.), Washington University School of Medicine, St. Louis, MO; Department of Neurology (N.A., H.Y., A.V.S., M.C.), Neurological Clinical Research Institute, Massachusetts General Hospital, Boston; Department of Neurology (L.H.v.d.B., M.A.v.E., J.H.V., B.S.d.V.), Brain Center Rudolf Magnus, University Medical Center Utrecht, University Utrecht, the Netherlands; Department of Neurology (M.H.), Columbia University, New York, NY; Department of Neurology (R.H. Baloh), Cedars-Sinai Medical Center, Los Angeles, CA; Department of Neurology (R.H. Brown, N.W., D.M.-Y., M.A.O., C.D.), University of Massachusetts, Worcester; Department of Neurology (J.D.R.), Johns Hopkins University, Baltimore, MD; and Biogen Inc. (A.M., T.F.), Boston, MA
| | - Jan H Veldink
- From the Department of Neurology (A.J.C., T.H., C.D., J.J.-B., A.M., S.B., A.S., T.J.E., C.C., T.M.M.), Washington University School of Medicine, St. Louis, MO; Department of Neurology (N.A., H.Y., A.V.S., M.C.), Neurological Clinical Research Institute, Massachusetts General Hospital, Boston; Department of Neurology (L.H.v.d.B., M.A.v.E., J.H.V., B.S.d.V.), Brain Center Rudolf Magnus, University Medical Center Utrecht, University Utrecht, the Netherlands; Department of Neurology (M.H.), Columbia University, New York, NY; Department of Neurology (R.H. Baloh), Cedars-Sinai Medical Center, Los Angeles, CA; Department of Neurology (R.H. Brown, N.W., D.M.-Y., M.A.O., C.D.), University of Massachusetts, Worcester; Department of Neurology (J.D.R.), Johns Hopkins University, Baltimore, MD; and Biogen Inc. (A.M., T.F.), Boston, MA
| | - Balint S de Vries
- From the Department of Neurology (A.J.C., T.H., C.D., J.J.-B., A.M., S.B., A.S., T.J.E., C.C., T.M.M.), Washington University School of Medicine, St. Louis, MO; Department of Neurology (N.A., H.Y., A.V.S., M.C.), Neurological Clinical Research Institute, Massachusetts General Hospital, Boston; Department of Neurology (L.H.v.d.B., M.A.v.E., J.H.V., B.S.d.V.), Brain Center Rudolf Magnus, University Medical Center Utrecht, University Utrecht, the Netherlands; Department of Neurology (M.H.), Columbia University, New York, NY; Department of Neurology (R.H. Baloh), Cedars-Sinai Medical Center, Los Angeles, CA; Department of Neurology (R.H. Brown, N.W., D.M.-Y., M.A.O., C.D.), University of Massachusetts, Worcester; Department of Neurology (J.D.R.), Johns Hopkins University, Baltimore, MD; and Biogen Inc. (A.M., T.F.), Boston, MA
| | - Jeffrey D Rothstein
- From the Department of Neurology (A.J.C., T.H., C.D., J.J.-B., A.M., S.B., A.S., T.J.E., C.C., T.M.M.), Washington University School of Medicine, St. Louis, MO; Department of Neurology (N.A., H.Y., A.V.S., M.C.), Neurological Clinical Research Institute, Massachusetts General Hospital, Boston; Department of Neurology (L.H.v.d.B., M.A.v.E., J.H.V., B.S.d.V.), Brain Center Rudolf Magnus, University Medical Center Utrecht, University Utrecht, the Netherlands; Department of Neurology (M.H.), Columbia University, New York, NY; Department of Neurology (R.H. Baloh), Cedars-Sinai Medical Center, Los Angeles, CA; Department of Neurology (R.H. Brown, N.W., D.M.-Y., M.A.O., C.D.), University of Massachusetts, Worcester; Department of Neurology (J.D.R.), Johns Hopkins University, Baltimore, MD; and Biogen Inc. (A.M., T.F.), Boston, MA
| | - Caroline Drain
- From the Department of Neurology (A.J.C., T.H., C.D., J.J.-B., A.M., S.B., A.S., T.J.E., C.C., T.M.M.), Washington University School of Medicine, St. Louis, MO; Department of Neurology (N.A., H.Y., A.V.S., M.C.), Neurological Clinical Research Institute, Massachusetts General Hospital, Boston; Department of Neurology (L.H.v.d.B., M.A.v.E., J.H.V., B.S.d.V.), Brain Center Rudolf Magnus, University Medical Center Utrecht, University Utrecht, the Netherlands; Department of Neurology (M.H.), Columbia University, New York, NY; Department of Neurology (R.H. Baloh), Cedars-Sinai Medical Center, Los Angeles, CA; Department of Neurology (R.H. Brown, N.W., D.M.-Y., M.A.O., C.D.), University of Massachusetts, Worcester; Department of Neurology (J.D.R.), Johns Hopkins University, Baltimore, MD; and Biogen Inc. (A.M., T.F.), Boston, MA
| | - Jennifer Jockel-Balsarotti
- From the Department of Neurology (A.J.C., T.H., C.D., J.J.-B., A.M., S.B., A.S., T.J.E., C.C., T.M.M.), Washington University School of Medicine, St. Louis, MO; Department of Neurology (N.A., H.Y., A.V.S., M.C.), Neurological Clinical Research Institute, Massachusetts General Hospital, Boston; Department of Neurology (L.H.v.d.B., M.A.v.E., J.H.V., B.S.d.V.), Brain Center Rudolf Magnus, University Medical Center Utrecht, University Utrecht, the Netherlands; Department of Neurology (M.H.), Columbia University, New York, NY; Department of Neurology (R.H. Baloh), Cedars-Sinai Medical Center, Los Angeles, CA; Department of Neurology (R.H. Brown, N.W., D.M.-Y., M.A.O., C.D.), University of Massachusetts, Worcester; Department of Neurology (J.D.R.), Johns Hopkins University, Baltimore, MD; and Biogen Inc. (A.M., T.F.), Boston, MA
| | - Amber Malcolm
- From the Department of Neurology (A.J.C., T.H., C.D., J.J.-B., A.M., S.B., A.S., T.J.E., C.C., T.M.M.), Washington University School of Medicine, St. Louis, MO; Department of Neurology (N.A., H.Y., A.V.S., M.C.), Neurological Clinical Research Institute, Massachusetts General Hospital, Boston; Department of Neurology (L.H.v.d.B., M.A.v.E., J.H.V., B.S.d.V.), Brain Center Rudolf Magnus, University Medical Center Utrecht, University Utrecht, the Netherlands; Department of Neurology (M.H.), Columbia University, New York, NY; Department of Neurology (R.H. Baloh), Cedars-Sinai Medical Center, Los Angeles, CA; Department of Neurology (R.H. Brown, N.W., D.M.-Y., M.A.O., C.D.), University of Massachusetts, Worcester; Department of Neurology (J.D.R.), Johns Hopkins University, Baltimore, MD; and Biogen Inc. (A.M., T.F.), Boston, MA
| | - Sonia Boodram
- From the Department of Neurology (A.J.C., T.H., C.D., J.J.-B., A.M., S.B., A.S., T.J.E., C.C., T.M.M.), Washington University School of Medicine, St. Louis, MO; Department of Neurology (N.A., H.Y., A.V.S., M.C.), Neurological Clinical Research Institute, Massachusetts General Hospital, Boston; Department of Neurology (L.H.v.d.B., M.A.v.E., J.H.V., B.S.d.V.), Brain Center Rudolf Magnus, University Medical Center Utrecht, University Utrecht, the Netherlands; Department of Neurology (M.H.), Columbia University, New York, NY; Department of Neurology (R.H. Baloh), Cedars-Sinai Medical Center, Los Angeles, CA; Department of Neurology (R.H. Brown, N.W., D.M.-Y., M.A.O., C.D.), University of Massachusetts, Worcester; Department of Neurology (J.D.R.), Johns Hopkins University, Baltimore, MD; and Biogen Inc. (A.M., T.F.), Boston, MA
| | - Amber Salter
- From the Department of Neurology (A.J.C., T.H., C.D., J.J.-B., A.M., S.B., A.S., T.J.E., C.C., T.M.M.), Washington University School of Medicine, St. Louis, MO; Department of Neurology (N.A., H.Y., A.V.S., M.C.), Neurological Clinical Research Institute, Massachusetts General Hospital, Boston; Department of Neurology (L.H.v.d.B., M.A.v.E., J.H.V., B.S.d.V.), Brain Center Rudolf Magnus, University Medical Center Utrecht, University Utrecht, the Netherlands; Department of Neurology (M.H.), Columbia University, New York, NY; Department of Neurology (R.H. Baloh), Cedars-Sinai Medical Center, Los Angeles, CA; Department of Neurology (R.H. Brown, N.W., D.M.-Y., M.A.O., C.D.), University of Massachusetts, Worcester; Department of Neurology (J.D.R.), Johns Hopkins University, Baltimore, MD; and Biogen Inc. (A.M., T.F.), Boston, MA
| | - Nicholas Wightman
- From the Department of Neurology (A.J.C., T.H., C.D., J.J.-B., A.M., S.B., A.S., T.J.E., C.C., T.M.M.), Washington University School of Medicine, St. Louis, MO; Department of Neurology (N.A., H.Y., A.V.S., M.C.), Neurological Clinical Research Institute, Massachusetts General Hospital, Boston; Department of Neurology (L.H.v.d.B., M.A.v.E., J.H.V., B.S.d.V.), Brain Center Rudolf Magnus, University Medical Center Utrecht, University Utrecht, the Netherlands; Department of Neurology (M.H.), Columbia University, New York, NY; Department of Neurology (R.H. Baloh), Cedars-Sinai Medical Center, Los Angeles, CA; Department of Neurology (R.H. Brown, N.W., D.M.-Y., M.A.O., C.D.), University of Massachusetts, Worcester; Department of Neurology (J.D.R.), Johns Hopkins University, Baltimore, MD; and Biogen Inc. (A.M., T.F.), Boston, MA
| | - Hong Yu
- From the Department of Neurology (A.J.C., T.H., C.D., J.J.-B., A.M., S.B., A.S., T.J.E., C.C., T.M.M.), Washington University School of Medicine, St. Louis, MO; Department of Neurology (N.A., H.Y., A.V.S., M.C.), Neurological Clinical Research Institute, Massachusetts General Hospital, Boston; Department of Neurology (L.H.v.d.B., M.A.v.E., J.H.V., B.S.d.V.), Brain Center Rudolf Magnus, University Medical Center Utrecht, University Utrecht, the Netherlands; Department of Neurology (M.H.), Columbia University, New York, NY; Department of Neurology (R.H. Baloh), Cedars-Sinai Medical Center, Los Angeles, CA; Department of Neurology (R.H. Brown, N.W., D.M.-Y., M.A.O., C.D.), University of Massachusetts, Worcester; Department of Neurology (J.D.R.), Johns Hopkins University, Baltimore, MD; and Biogen Inc. (A.M., T.F.), Boston, MA
| | - Alexander V Sherman
- From the Department of Neurology (A.J.C., T.H., C.D., J.J.-B., A.M., S.B., A.S., T.J.E., C.C., T.M.M.), Washington University School of Medicine, St. Louis, MO; Department of Neurology (N.A., H.Y., A.V.S., M.C.), Neurological Clinical Research Institute, Massachusetts General Hospital, Boston; Department of Neurology (L.H.v.d.B., M.A.v.E., J.H.V., B.S.d.V.), Brain Center Rudolf Magnus, University Medical Center Utrecht, University Utrecht, the Netherlands; Department of Neurology (M.H.), Columbia University, New York, NY; Department of Neurology (R.H. Baloh), Cedars-Sinai Medical Center, Los Angeles, CA; Department of Neurology (R.H. Brown, N.W., D.M.-Y., M.A.O., C.D.), University of Massachusetts, Worcester; Department of Neurology (J.D.R.), Johns Hopkins University, Baltimore, MD; and Biogen Inc. (A.M., T.F.), Boston, MA
| | - Thomas J Esparza
- From the Department of Neurology (A.J.C., T.H., C.D., J.J.-B., A.M., S.B., A.S., T.J.E., C.C., T.M.M.), Washington University School of Medicine, St. Louis, MO; Department of Neurology (N.A., H.Y., A.V.S., M.C.), Neurological Clinical Research Institute, Massachusetts General Hospital, Boston; Department of Neurology (L.H.v.d.B., M.A.v.E., J.H.V., B.S.d.V.), Brain Center Rudolf Magnus, University Medical Center Utrecht, University Utrecht, the Netherlands; Department of Neurology (M.H.), Columbia University, New York, NY; Department of Neurology (R.H. Baloh), Cedars-Sinai Medical Center, Los Angeles, CA; Department of Neurology (R.H. Brown, N.W., D.M.-Y., M.A.O., C.D.), University of Massachusetts, Worcester; Department of Neurology (J.D.R.), Johns Hopkins University, Baltimore, MD; and Biogen Inc. (A.M., T.F.), Boston, MA
| | - Diane McKenna-Yasek
- From the Department of Neurology (A.J.C., T.H., C.D., J.J.-B., A.M., S.B., A.S., T.J.E., C.C., T.M.M.), Washington University School of Medicine, St. Louis, MO; Department of Neurology (N.A., H.Y., A.V.S., M.C.), Neurological Clinical Research Institute, Massachusetts General Hospital, Boston; Department of Neurology (L.H.v.d.B., M.A.v.E., J.H.V., B.S.d.V.), Brain Center Rudolf Magnus, University Medical Center Utrecht, University Utrecht, the Netherlands; Department of Neurology (M.H.), Columbia University, New York, NY; Department of Neurology (R.H. Baloh), Cedars-Sinai Medical Center, Los Angeles, CA; Department of Neurology (R.H. Brown, N.W., D.M.-Y., M.A.O., C.D.), University of Massachusetts, Worcester; Department of Neurology (J.D.R.), Johns Hopkins University, Baltimore, MD; and Biogen Inc. (A.M., T.F.), Boston, MA
| | - Margaret A Owegi
- From the Department of Neurology (A.J.C., T.H., C.D., J.J.-B., A.M., S.B., A.S., T.J.E., C.C., T.M.M.), Washington University School of Medicine, St. Louis, MO; Department of Neurology (N.A., H.Y., A.V.S., M.C.), Neurological Clinical Research Institute, Massachusetts General Hospital, Boston; Department of Neurology (L.H.v.d.B., M.A.v.E., J.H.V., B.S.d.V.), Brain Center Rudolf Magnus, University Medical Center Utrecht, University Utrecht, the Netherlands; Department of Neurology (M.H.), Columbia University, New York, NY; Department of Neurology (R.H. Baloh), Cedars-Sinai Medical Center, Los Angeles, CA; Department of Neurology (R.H. Brown, N.W., D.M.-Y., M.A.O., C.D.), University of Massachusetts, Worcester; Department of Neurology (J.D.R.), Johns Hopkins University, Baltimore, MD; and Biogen Inc. (A.M., T.F.), Boston, MA
| | - Catherine Douthwright
- From the Department of Neurology (A.J.C., T.H., C.D., J.J.-B., A.M., S.B., A.S., T.J.E., C.C., T.M.M.), Washington University School of Medicine, St. Louis, MO; Department of Neurology (N.A., H.Y., A.V.S., M.C.), Neurological Clinical Research Institute, Massachusetts General Hospital, Boston; Department of Neurology (L.H.v.d.B., M.A.v.E., J.H.V., B.S.d.V.), Brain Center Rudolf Magnus, University Medical Center Utrecht, University Utrecht, the Netherlands; Department of Neurology (M.H.), Columbia University, New York, NY; Department of Neurology (R.H. Baloh), Cedars-Sinai Medical Center, Los Angeles, CA; Department of Neurology (R.H. Brown, N.W., D.M.-Y., M.A.O., C.D.), University of Massachusetts, Worcester; Department of Neurology (J.D.R.), Johns Hopkins University, Baltimore, MD; and Biogen Inc. (A.M., T.F.), Boston, MA
| | | | - Alexander McCampbell
- From the Department of Neurology (A.J.C., T.H., C.D., J.J.-B., A.M., S.B., A.S., T.J.E., C.C., T.M.M.), Washington University School of Medicine, St. Louis, MO; Department of Neurology (N.A., H.Y., A.V.S., M.C.), Neurological Clinical Research Institute, Massachusetts General Hospital, Boston; Department of Neurology (L.H.v.d.B., M.A.v.E., J.H.V., B.S.d.V.), Brain Center Rudolf Magnus, University Medical Center Utrecht, University Utrecht, the Netherlands; Department of Neurology (M.H.), Columbia University, New York, NY; Department of Neurology (R.H. Baloh), Cedars-Sinai Medical Center, Los Angeles, CA; Department of Neurology (R.H. Brown, N.W., D.M.-Y., M.A.O., C.D.), University of Massachusetts, Worcester; Department of Neurology (J.D.R.), Johns Hopkins University, Baltimore, MD; and Biogen Inc. (A.M., T.F.), Boston, MA
| | - Toby Ferguson
- From the Department of Neurology (A.J.C., T.H., C.D., J.J.-B., A.M., S.B., A.S., T.J.E., C.C., T.M.M.), Washington University School of Medicine, St. Louis, MO; Department of Neurology (N.A., H.Y., A.V.S., M.C.), Neurological Clinical Research Institute, Massachusetts General Hospital, Boston; Department of Neurology (L.H.v.d.B., M.A.v.E., J.H.V., B.S.d.V.), Brain Center Rudolf Magnus, University Medical Center Utrecht, University Utrecht, the Netherlands; Department of Neurology (M.H.), Columbia University, New York, NY; Department of Neurology (R.H. Baloh), Cedars-Sinai Medical Center, Los Angeles, CA; Department of Neurology (R.H. Brown, N.W., D.M.-Y., M.A.O., C.D.), University of Massachusetts, Worcester; Department of Neurology (J.D.R.), Johns Hopkins University, Baltimore, MD; and Biogen Inc. (A.M., T.F.), Boston, MA
| | - Carlos Cruchaga
- From the Department of Neurology (A.J.C., T.H., C.D., J.J.-B., A.M., S.B., A.S., T.J.E., C.C., T.M.M.), Washington University School of Medicine, St. Louis, MO; Department of Neurology (N.A., H.Y., A.V.S., M.C.), Neurological Clinical Research Institute, Massachusetts General Hospital, Boston; Department of Neurology (L.H.v.d.B., M.A.v.E., J.H.V., B.S.d.V.), Brain Center Rudolf Magnus, University Medical Center Utrecht, University Utrecht, the Netherlands; Department of Neurology (M.H.), Columbia University, New York, NY; Department of Neurology (R.H. Baloh), Cedars-Sinai Medical Center, Los Angeles, CA; Department of Neurology (R.H. Brown, N.W., D.M.-Y., M.A.O., C.D.), University of Massachusetts, Worcester; Department of Neurology (J.D.R.), Johns Hopkins University, Baltimore, MD; and Biogen Inc. (A.M., T.F.), Boston, MA
| | - Merit Cudkowicz
- From the Department of Neurology (A.J.C., T.H., C.D., J.J.-B., A.M., S.B., A.S., T.J.E., C.C., T.M.M.), Washington University School of Medicine, St. Louis, MO; Department of Neurology (N.A., H.Y., A.V.S., M.C.), Neurological Clinical Research Institute, Massachusetts General Hospital, Boston; Department of Neurology (L.H.v.d.B., M.A.v.E., J.H.V., B.S.d.V.), Brain Center Rudolf Magnus, University Medical Center Utrecht, University Utrecht, the Netherlands; Department of Neurology (M.H.), Columbia University, New York, NY; Department of Neurology (R.H. Baloh), Cedars-Sinai Medical Center, Los Angeles, CA; Department of Neurology (R.H. Brown, N.W., D.M.-Y., M.A.O., C.D.), University of Massachusetts, Worcester; Department of Neurology (J.D.R.), Johns Hopkins University, Baltimore, MD; and Biogen Inc. (A.M., T.F.), Boston, MA
| | - Timothy M Miller
- From the Department of Neurology (A.J.C., T.H., C.D., J.J.-B., A.M., S.B., A.S., T.J.E., C.C., T.M.M.), Washington University School of Medicine, St. Louis, MO; Department of Neurology (N.A., H.Y., A.V.S., M.C.), Neurological Clinical Research Institute, Massachusetts General Hospital, Boston; Department of Neurology (L.H.v.d.B., M.A.v.E., J.H.V., B.S.d.V.), Brain Center Rudolf Magnus, University Medical Center Utrecht, University Utrecht, the Netherlands; Department of Neurology (M.H.), Columbia University, New York, NY; Department of Neurology (R.H. Baloh), Cedars-Sinai Medical Center, Los Angeles, CA; Department of Neurology (R.H. Brown, N.W., D.M.-Y., M.A.O., C.D.), University of Massachusetts, Worcester; Department of Neurology (J.D.R.), Johns Hopkins University, Baltimore, MD; and Biogen Inc. (A.M., T.F.), Boston, MA.
| |
Collapse
|
47
|
Rooney J, Murray D, Campion A, Moloney H, Tattersall R, Doherty M, Hammond M, Heverin M, McLaughlin R, Hardiman O. The C9orf72 expansion is associated with accelerated respiratory function decline in a large Amyotrophic Lateral Sclerosis cohort. HRB Open Res 2019; 2:23. [PMID: 32296747 PMCID: PMC7140774 DOI: 10.12688/hrbopenres.12940.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2019] [Indexed: 12/11/2022] Open
Abstract
Introduction: The C9orf72 hexanucleotide repeat expansion is causal in amyotrophic lateral sclerosis (ALS) and has a negative effect on prognosis. The C9orf72 repeat expansion has been associated with an accelerated deterioration of respiratory function and survival in a cohort of 372 Portuguese patients. Methods: Cases presenting to the Irish ALS clinic with both longitudinal occluded sniff nasal inspiratory pressure (SNIP) and C9orf72 testing were including in the study. Clinical variables and survival characteristics of these patients were collected. Joint longitudinal and time to event models were constructed to explore the longitudinal characteristics of the cohort by C9orf72 status. Results: In total, 630 cases were included, of which 58 (9.2%) carried the C9orf72 repeat expansion. Plots of the longitudinal trend after joint modelling revealed that those carrying the expansion had worse respiratory function throughout the course of their disease than those without. The ALS Functional Rating Scale-revised (ALSFRS-R) respiratory sub-score did not distinguish C9orf72 normal from expanded cases. Furthermore, modelling by site of onset and gender sub-groups revealed that this difference was greatest in male spinal onset cases. Joint models further indicated that occluded SNIP values were of prognostic importance. Conclusions: Our results confirm findings from Portugal that the C9orf72 repeat expansion is associated with accelerated respiratory function decline. Analysis via joint models indicate that respiratory function is of prognostic importance and may explain previous observations of poorer prognosis in male spinal onset patients carrying the C9orf72 expansion.
Collapse
Affiliation(s)
- James Rooney
- Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Deirdre Murray
- Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Beaumont Hospital, Dublin, Ireland
| | | | - Hannah Moloney
- Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | | - Mark Doherty
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Michaela Hammond
- Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Mark Heverin
- Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | | - Orla Hardiman
- Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Beaumont Hospital, Dublin, Ireland
| |
Collapse
|
48
|
Caswell C, McMillan CT, Xie SX, Van Deerlin VM, Suh E, Lee EB, Trojanowski JQ, Lee VMY, Irwin DJ, Grossman M, Massimo LM. Genetic predictors of survival in behavioral variant frontotemporal degeneration. Neurology 2019; 93:e1707-e1714. [PMID: 31537715 DOI: 10.1212/wnl.0000000000008387] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 05/29/2019] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE To determine autosomal dominant genetic predictors of survival in individuals with behavioral variant frontotemporal degeneration (bvFTD). METHODS A retrospective chart review of 174 cases with a clinical phenotype of bvFTD but no associated elementary neurologic features was performed, with diagnosis either autopsy-confirmed (n = 57) or supported by CSF evidence of non-Alzheimer pathology (n = 117). Genetic analysis of the 3 most common genes with pathogenic autosomal dominant mutations associated with frontotemporal degeneration was performed in all patients, which identified cases with C9orf72 expansion (n = 28), progranulin (GRN) mutation (n = 12), and microtubule-associated protein tau (MAPT) mutation (n = 10). Cox proportional hazards regressions were used to test for associations between survival and mutation status, sex, age at symptom onset, and education. RESULTS Across all patients with bvFTD, the presence of a disease-associated pathogenic mutation was associated with shortened survival (hazard ratio [HR] 2.164, 95% confidence interval [CI] 1.391, 3.368). In separate models, a GRN mutation (HR 2.423, 95% CI 1.237, 4.744), MAPT mutation (HR 8.056, 95% CI 2.938, 22.092), and C9orf72 expansion (HR 1.832, 95% CI 1.034, 3.244) were each individually associated with shorter survival relative to sporadic bvFTD. A mutation on the MAPT gene results in an earlier age at onset than a C9orf72 expansion or mutation on the GRN gene (p = 0.016). CONCLUSIONS Our findings suggest that autosomal dominantly inherited mutations, modulated by age at symptom onset, associate with shorter survival among patients with bvFTD. We suggest that clinical trials and clinical management should consider mutation status and age at onset when evaluating disease progression.
Collapse
Affiliation(s)
- Carrie Caswell
- From the Department of Biostatistics, Epidemiology, and Informatics (C.C., S.X.X.), Department of Neurology (C.T.M., D.J.I., M.G., L.M.M.), Penn Frontotemporal Degeneration Center (C.T.M., D.J.I., M.G., L.M.M.), Translational Neuropathology Research Laboratory (E.B.L.), Department of Pathology and Laboratory Medicine (V.M.V.D., E.B.L., J.Q.T., V.M.-Y.L.), and Center for Neurodegenerative Disease Research (V.M.V.D., E.S., E.B.L., J.Q.T., V.M.-Y.L.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Corey T McMillan
- From the Department of Biostatistics, Epidemiology, and Informatics (C.C., S.X.X.), Department of Neurology (C.T.M., D.J.I., M.G., L.M.M.), Penn Frontotemporal Degeneration Center (C.T.M., D.J.I., M.G., L.M.M.), Translational Neuropathology Research Laboratory (E.B.L.), Department of Pathology and Laboratory Medicine (V.M.V.D., E.B.L., J.Q.T., V.M.-Y.L.), and Center for Neurodegenerative Disease Research (V.M.V.D., E.S., E.B.L., J.Q.T., V.M.-Y.L.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Sharon X Xie
- From the Department of Biostatistics, Epidemiology, and Informatics (C.C., S.X.X.), Department of Neurology (C.T.M., D.J.I., M.G., L.M.M.), Penn Frontotemporal Degeneration Center (C.T.M., D.J.I., M.G., L.M.M.), Translational Neuropathology Research Laboratory (E.B.L.), Department of Pathology and Laboratory Medicine (V.M.V.D., E.B.L., J.Q.T., V.M.-Y.L.), and Center for Neurodegenerative Disease Research (V.M.V.D., E.S., E.B.L., J.Q.T., V.M.-Y.L.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Vivianna M Van Deerlin
- From the Department of Biostatistics, Epidemiology, and Informatics (C.C., S.X.X.), Department of Neurology (C.T.M., D.J.I., M.G., L.M.M.), Penn Frontotemporal Degeneration Center (C.T.M., D.J.I., M.G., L.M.M.), Translational Neuropathology Research Laboratory (E.B.L.), Department of Pathology and Laboratory Medicine (V.M.V.D., E.B.L., J.Q.T., V.M.-Y.L.), and Center for Neurodegenerative Disease Research (V.M.V.D., E.S., E.B.L., J.Q.T., V.M.-Y.L.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - EunRan Suh
- From the Department of Biostatistics, Epidemiology, and Informatics (C.C., S.X.X.), Department of Neurology (C.T.M., D.J.I., M.G., L.M.M.), Penn Frontotemporal Degeneration Center (C.T.M., D.J.I., M.G., L.M.M.), Translational Neuropathology Research Laboratory (E.B.L.), Department of Pathology and Laboratory Medicine (V.M.V.D., E.B.L., J.Q.T., V.M.-Y.L.), and Center for Neurodegenerative Disease Research (V.M.V.D., E.S., E.B.L., J.Q.T., V.M.-Y.L.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Edward B Lee
- From the Department of Biostatistics, Epidemiology, and Informatics (C.C., S.X.X.), Department of Neurology (C.T.M., D.J.I., M.G., L.M.M.), Penn Frontotemporal Degeneration Center (C.T.M., D.J.I., M.G., L.M.M.), Translational Neuropathology Research Laboratory (E.B.L.), Department of Pathology and Laboratory Medicine (V.M.V.D., E.B.L., J.Q.T., V.M.-Y.L.), and Center for Neurodegenerative Disease Research (V.M.V.D., E.S., E.B.L., J.Q.T., V.M.-Y.L.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - John Q Trojanowski
- From the Department of Biostatistics, Epidemiology, and Informatics (C.C., S.X.X.), Department of Neurology (C.T.M., D.J.I., M.G., L.M.M.), Penn Frontotemporal Degeneration Center (C.T.M., D.J.I., M.G., L.M.M.), Translational Neuropathology Research Laboratory (E.B.L.), Department of Pathology and Laboratory Medicine (V.M.V.D., E.B.L., J.Q.T., V.M.-Y.L.), and Center for Neurodegenerative Disease Research (V.M.V.D., E.S., E.B.L., J.Q.T., V.M.-Y.L.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Virginia M-Y Lee
- From the Department of Biostatistics, Epidemiology, and Informatics (C.C., S.X.X.), Department of Neurology (C.T.M., D.J.I., M.G., L.M.M.), Penn Frontotemporal Degeneration Center (C.T.M., D.J.I., M.G., L.M.M.), Translational Neuropathology Research Laboratory (E.B.L.), Department of Pathology and Laboratory Medicine (V.M.V.D., E.B.L., J.Q.T., V.M.-Y.L.), and Center for Neurodegenerative Disease Research (V.M.V.D., E.S., E.B.L., J.Q.T., V.M.-Y.L.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - David J Irwin
- From the Department of Biostatistics, Epidemiology, and Informatics (C.C., S.X.X.), Department of Neurology (C.T.M., D.J.I., M.G., L.M.M.), Penn Frontotemporal Degeneration Center (C.T.M., D.J.I., M.G., L.M.M.), Translational Neuropathology Research Laboratory (E.B.L.), Department of Pathology and Laboratory Medicine (V.M.V.D., E.B.L., J.Q.T., V.M.-Y.L.), and Center for Neurodegenerative Disease Research (V.M.V.D., E.S., E.B.L., J.Q.T., V.M.-Y.L.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Murray Grossman
- From the Department of Biostatistics, Epidemiology, and Informatics (C.C., S.X.X.), Department of Neurology (C.T.M., D.J.I., M.G., L.M.M.), Penn Frontotemporal Degeneration Center (C.T.M., D.J.I., M.G., L.M.M.), Translational Neuropathology Research Laboratory (E.B.L.), Department of Pathology and Laboratory Medicine (V.M.V.D., E.B.L., J.Q.T., V.M.-Y.L.), and Center for Neurodegenerative Disease Research (V.M.V.D., E.S., E.B.L., J.Q.T., V.M.-Y.L.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Lauren M Massimo
- From the Department of Biostatistics, Epidemiology, and Informatics (C.C., S.X.X.), Department of Neurology (C.T.M., D.J.I., M.G., L.M.M.), Penn Frontotemporal Degeneration Center (C.T.M., D.J.I., M.G., L.M.M.), Translational Neuropathology Research Laboratory (E.B.L.), Department of Pathology and Laboratory Medicine (V.M.V.D., E.B.L., J.Q.T., V.M.-Y.L.), and Center for Neurodegenerative Disease Research (V.M.V.D., E.S., E.B.L., J.Q.T., V.M.-Y.L.), Perelman School of Medicine, University of Pennsylvania, Philadelphia.
| |
Collapse
|
49
|
Abstract
The discovery that repeat expansions in the C9orf72 gene are a frequent cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) has revolutionized our understanding of these diseases. Substantial headway has been made in characterizing C9orf72-mediated disease and unravelling its underlying aetiopathogenesis. Three main disease mechanisms have been proposed: loss of function of the C9orf72 protein and toxic gain of function from C9orf72 repeat RNA or from dipeptide repeat proteins produced by repeat-associated non-ATG translation. Several downstream processes across a range of cellular functions have also been implicated. In this article, we review the pathological and mechanistic features of C9orf72-associated FTD and ALS (collectively termed C9FTD/ALS), the model systems used to study these conditions, and the probable initiators of downstream disease mechanisms. We suggest that a combination of upstream mechanisms involving both loss and gain of function and downstream cellular pathways involving both cell-autonomous and non-cell-autonomous effects contributes to disease progression.
Collapse
Affiliation(s)
- Rubika Balendra
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.,Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, UCL, London, UK
| | - Adrian M Isaacs
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK. .,UK Dementia Research Institute at UCL, UCL Institute of Neurology, London, UK.
| |
Collapse
|
50
|
Abstract
PURPOSE OF REVIEW This article summarizes the clinical and anatomic features of the three named variants of primary progressive aphasia (PPA): semantic variant PPA, nonfluent/agrammatic variant PPA, and logopenic variant PPA. Three stroke aphasia syndromes that resemble the PPA variants (Broca aphasia, Wernicke aphasia, and conduction aphasia) are also presented. RECENT FINDINGS Semantic variant PPA and Wernicke aphasia are characterized by fluent speech with naming and comprehension difficulty; these syndromes are associated with disease in different portions of the left temporal lobe. Patients with nonfluent/agrammatic variant PPA or Broca aphasia have nonfluent speech with grammatical difficulty; these syndromes are associated with disease centered in the left inferior frontal lobe. Patients with logopenic variant PPA or conduction aphasia have difficulty with repetition and word finding in conversational speech; these syndromes are associated with disease in the left inferior parietal lobe. While PPA and stroke aphasias resemble one another, this article also presents their distinguishing features. SUMMARY Primary progressive and stroke aphasia syndromes interrupt the left perisylvian language network, resulting in identifiable aphasic syndromes.
Collapse
|