1
|
Weinhold ER, Vaughan CL, O'Hara M, Bloemen EM, Bakemen D, Ducharme S, Pressman PS. Clinical considerations regarding suspected "BvFTD-by-proxy": a case series. Neurocase 2024:1-7. [PMID: 39462825 DOI: 10.1080/13554794.2024.2419177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 10/15/2024] [Indexed: 10/29/2024]
Abstract
A diagnosis of behavioral variant frontotemporal dementia (bvFTD) often relies on informant reports of significant behavioral changes. "BvFTD-by-proxy" describes situations of neuropsychiatric changes reported solely by an informant under circumstances that may raise questions regarding their objectivity. We present three cases of bvFTD-like symptoms reported by spouses, where progression was unclear, testing showed mild but stable executive dysfunction, and neuroimaging was unremarkable. The subjective nature of bvFTD criteria leaves patients vulnerable to misleading informant reports, especially amid relational discord, and may threaten patient autonomy. Recognizing and managing this situation is critical but time-consuming, often requiring coordinated care across multiple providers.
Collapse
Affiliation(s)
- Erin R Weinhold
- Departments of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Departments of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Christina L Vaughan
- Departments of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Departments of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Mary O'Hara
- Departments of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Elizabeth M Bloemen
- Departments of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Delia Bakemen
- Departments of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Simon Ducharme
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Peter S Pressman
- Departments of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
2
|
Hoshina Y, Moser C, Wright MA, Sunderman E, Livsey CT, Spoth E, Clardy SL, Cliatt Brown CJ. C9orf72 Gene-Associated Frontotemporal Dementia Mimicking Autoimmune Pathology. Neurologist 2024:00127893-990000000-00153. [PMID: 39382125 DOI: 10.1097/nrl.0000000000000587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
INTRODUCTION The C9orf72 mutation can manifest in diverse clinical ways, including rapid cognitive decline, parkinsonism, or late-life neuropsychiatric symptoms, sometimes mimicking autoimmune encephalitis. CASE REPORT A 64-year-old female presented to the autoimmune neurology clinic with rapidly progressive dementia (RPD) associated with episodes of headache, confusion, auditory hallucinations, and abnormal electroencephalogram. She was treated empirically at an outside hospital for possible autoimmune encephalitis with intravenous methylprednisolone, but there was no improvement, and rapid cognitive decline continued. Family history was notable for RPD with akinetic mutism in her sister, sudden severe depression followed by parkinsonism with progressive dementia in her father in his 60s, and late-life gradually progressive dementia in her mother. Additional testing revealed a low titer positive contactin-associated protein-like 2 (CASPR2) immunoglobulin G (IgG) in the serum and elevated CSF 14-3-3 protein. CSF CASPR2 IgG and real-time quaking-induced conversion for Creutzfeldt-Jakob disease were negative. Brain MRI showed normal parenchymal volume. Genetic testing was conducted, which identified a heterozygous pathogenic hexanucleotide tandem repeat expansion in the C9orf72 gene. CONCLUSION This case underscores the phenotypic variability of C9orf72 mutation and the importance of a detailed family history exploring young or atypical deaths and neuropsychiatric symptoms or behavioral changes. Genetic etiologies are crucial to consider in those with a family history concerning autosomal dominant inheritance patterns of early-onset dementia, parkinsonism, or late-onset psychiatric disease. Emphasis is placed on considering alternative etiologies early, particularly when there is no response to first-line immunomodulation for suspected autoimmune dementia.
Collapse
Affiliation(s)
| | | | | | | | - Charles T Livsey
- Department of Neurology, Intermountain Medical Center, Murray, UT
| | | | - Stacey L Clardy
- Department of Neurology, University of Utah
- Department of Neurology, George E. Wahlen Veterans Affairs Medical Center, Salt Lake City
| | - Christine J Cliatt Brown
- Department of Neurology, University of Utah
- Department of Neurology, George E. Wahlen Veterans Affairs Medical Center, Salt Lake City
| |
Collapse
|
3
|
Spencer BE, Xie SX, Elman L, Quinn CC, Amado D, Baer M, Lee EB, Van Deerlin VM, Dratch L, Massimo L, Irwin DJ, McMillan CT. C9orf72 repeat expansions modify risk for secondary motor and cognitive-behavioral symptoms in behavioral-variant frontotemporal degeneration and amyotrophic lateral sclerosis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.30.24306638. [PMID: 38746326 PMCID: PMC11092697 DOI: 10.1101/2024.04.30.24306638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
In behavioral-variant frontotemporal degeneration (bvFTD) and amyotrophic lateral sclerosis (ALS), secondary motor or cognitive-behavioral symptoms, respectively, are associated with shorter survival. However, factors influencing secondary symptom development remain largely unexplored. We performed a retrospective evaluation of the entire disease course of individuals with ALS (n=172) and bvFTD (n=69). Only individuals who had neuropathological confirmation of TDP-43 proteinopathy at autopsy or a C9orf72 hexanucleotide repeat expansion were included for analysis. We examined the odds and hazard of secondary symptom development and assessed whether each was modified by the presence of a C9orf72 expansion or initial clinical syndrome. Binary logistic regression and Cox proportional hazard analyses revealed increased odds (OR=4.25 [95% CI 1.97-9.14], p<0.001) and an increased hazard (HR= 4.77 [95% CI 2.33-9.79], p<0.001) for developing secondary symptoms in those with a C9orf72 expansion compared to those without. Initial clinical syndrome (bvFTD or ALS), age at symptom onset, and sex were not associated with development of secondary symptoms. These data highlight the need for clinician vigilance to detect the onset of secondary motor and cognitive-behavioral symptoms in patients carrying a C9orf72 expansion, regardless of initial clinical syndrome. C9orf72 clinical care can be enhanced through coordination between cognitive and neuromuscular clinics.
Collapse
|
4
|
Selig M, Lee G, Lebowitz B, Franceschi D, Absar N. A Case of Personality and Behavioral Changes with Frontotemporal and Cerebellar Atrophy on MRI with Corresponding Hypometabolism on FDG-PET. Case Rep Psychiatry 2024; 2024:2863662. [PMID: 39188506 PMCID: PMC11347030 DOI: 10.1155/2024/2863662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 07/17/2024] [Accepted: 07/26/2024] [Indexed: 08/28/2024] Open
Abstract
Background The differential diagnosis of a patient with cognitive, behavioral, and motor symptoms is broad. There is much overlap between neurocognitive disorders due to frontotemporal dementia and other subcortical dementia. A less known diagnosis, cerebellar cognitive affective syndrome (CCAS), should also be considered. Case History. A 29-year-old female presented with ataxia and left-sided weakness. CSF showed oligoclonal bands, and MRI showed multiple white matter lesions with some atrophy. She was diagnosed with multiple sclerosis (MS). At age 35, she developed frontal lobe symptoms and executive dysfunction; she was diagnosed with MS with bipolar disorder. Neuropsychological evaluation at that time showed significant deficits in multiple cognitive domains. Subsequent MRI showed progressive frontotemporal atrophy, and FDG-PET uncovered hypometabolism in the frontotemporal lobes and cerebellum. At age 38, her behavior worsened with aggression, and she was started on olanzapine. She responded well with decreased agitation and improved motivation and attention. Compared with previous scans, most recent MRI and FDG-PET showed interval increase in cerebellar atrophy with increase in hypometabolism in the cerebellum, respectively. Conclusion Based on cerebellar, affective, and subcortical cognitive examination findings, our diagnosis is probable CCAS. The cerebellum should be considered as a possible etiology of frontal subcortical cognitive impairment.
Collapse
Affiliation(s)
- Megan Selig
- Department of NeuropsychiatryStony Brook University Renaissance School of Medicine, Stony Brook, NY, USA
| | - Gloria Lee
- Department of NeuropsychiatryStony Brook University Renaissance School of Medicine, Stony Brook, NY, USA
| | - Brian Lebowitz
- Department of NeuropsychiatryStony Brook University Renaissance School of Medicine, Stony Brook, NY, USA
| | - Dinko Franceschi
- Department of NeuropsychiatryStony Brook University Renaissance School of Medicine, Stony Brook, NY, USA
| | - Nicole Absar
- Neurocognitive ClinicMass General Brigham—Martha's Vineyard Hospital, Martha's Vineyard, MA, USA
| |
Collapse
|
5
|
Goutman SA, Goyal NA, Payne K, Paisán‐Ruiz C, Kupelian V, Kang ML, Mitchell AA, Fecteau TE. ALS Identified: two-year findings from a sponsored ALS genetic testing program. Ann Clin Transl Neurol 2024; 11:2201-2211. [PMID: 39044379 PMCID: PMC11330217 DOI: 10.1002/acn3.52140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 06/21/2024] [Accepted: 06/22/2024] [Indexed: 07/25/2024] Open
Abstract
OBJECTIVE To report initial results from the Amyotrophic Lateral Sclerosis (ALS) Identified genetic testing (GT) program on characteristics of individuals tested and frequency of reported disease-causing variants. METHODS ALS Identified used the Invitae Amyotrophic Lateral Sclerosis panel (Invitae, San Francisco, CA, USA) to assay 22 ALS-associated genes. Sponsored by Biogen (Cambridge, MA, USA), the program was launched in June 2021 and was available at no charge to individuals ≥18 years in the United States and Puerto Rico with an ALS diagnosis or a known family history of ALS. Deidentified data were available to Biogen. RESULTS As of 26 October 2023, 998 healthcare professionals ordered the panel at 681 unique care sites. Of 8054 individuals examined, 7483 (92.9%) were reported to have a clinical diagnosis of ALS, while 571 (7.1%) were asymptomatic relatives. Of the individuals with a clinical ALS diagnosis, 57.7% were male (n = 4319) and 42.3% female (n = 3164). Mean (SD) age at diagnosis is 62 (13) years. Out of the 7483 clinically diagnosed individuals, 1810 (24.2%) showed genetic variations in ALS-associated genes. Among these, 865 individuals (47.8%) carried pathogenic variants, and 44 (2.4%) had likely pathogenic variants, totaling 12.1% of the clinically diagnosed population. INTERPRETATION Since 2021 there has been robust uptake and sustained use of the ALS Identified program, one of the largest samples of people with ALS to date across the United States, demonstrating the interest and need for genetic ALS testing.
Collapse
|
6
|
R K Roy A, Noohi F, Morris NA, Ljubenkov P, Heuer H, Fong J, Hall M, Lario Lago A, Rankin KP, Miller BL, Boxer AL, Rosen HJ, Seeley WW, Perry DC, Yokoyama JS, Lee SE, Sturm VE. Basal parasympathetic deficits in C9orf72 hexanucleotide repeat expansion carriers relate to smaller frontoinsula and thalamus volume and lower empathy. Neuroimage Clin 2024; 43:103649. [PMID: 39098187 PMCID: PMC11342757 DOI: 10.1016/j.nicl.2024.103649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 07/16/2024] [Accepted: 07/29/2024] [Indexed: 08/06/2024]
Abstract
Diminished basal parasympathetic nervous system activity is a feature of frontotemporal dementia that relates to left frontoinsula dysfunction and empathy impairment. Individuals with a pathogenic expansion of the hexanucleotide repeat in chromosome 9 open reading frame 72 (C9orf72), the most common genetic cause of frontotemporal dementia and amyotrophic lateral sclerosis, provide a unique opportunity to examine whether parasympathetic activity is disrupted in genetic forms of frontotemporal dementia and to investigate when parasympathetic deficits manifest in the pathophysiological cascade. We measured baseline respiratory sinus arrhythmia, a parasympathetic measure of heart rate variability, over two minutes in a sample of 102 participants that included 19 asymptomatic expansion carriers (C9+ asymp), 14 expansion carriers with mild cognitive impairment (C9+ MCI), 16 symptomatic expansion carriers with frontotemporal dementia (C9+ FTD), and 53 expansion-negative healthy controls (C9- HC) who also underwent structural magnetic resonance imaging. In follow-up analyses, we compared baseline respiratory sinus arrhythmia in the C9+ FTD group with an independent age-, sex-, and clinical severity-matched group of 26 people with sporadic behavioral variant frontotemporal dementia. The Frontotemporal Lobar Degeneration-modified Clinical Dementia Rating-Sum of Boxes score was used to quantify behavioral symptom severity, and informant ratings on the Interpersonal Reactivity Index provided measures of participants' current emotional (empathic concern) and cognitive (perspective-taking) empathy. Results indicated that the C9+ FTD group had lower baseline respiratory sinus arrhythmia than the C9+ MCI, C9+ asymp, and C9- HC groups, a deficit that was comparable to that of sporadic behavioral variant frontotemporal dementia. Linear regression analyses indicated that lower baseline respiratory sinus arrhythmia was associated with worse behavioral symptom severity and lower empathic concern and perspective-taking across the C9orf72 expansion carrier clinical spectrum. Whole-brain voxel-based morphometry analyses in participants with C9orf72 pathogenic expansions found that lower baseline respiratory sinus arrhythmia correlated with smaller gray matter volume in the left frontoinsula and bilateral thalamus, key structures that support parasympathetic function, and in the bilateral parietal lobes, occipital lobes, and cerebellum, regions that are also vulnerable in individuals with C9orf72 expansions. This study provides novel evidence that basal parasympathetic functioning is diminished in FTD due to C9orf72 expansions and suggests that baseline respiratory sinus arrhythmia may be a potential non-invasive biomarker that is sensitive to behavioral symptoms in the early stages of disease.
Collapse
Affiliation(s)
- Ashlin R K Roy
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| | - Fate Noohi
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| | - Nathaniel A Morris
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| | - Peter Ljubenkov
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| | - Hilary Heuer
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| | - Jamie Fong
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| | - Matthew Hall
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| | | | - Katherine P Rankin
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| | - Bruce L Miller
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| | - Adam L Boxer
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| | - Howard J Rosen
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| | - William W Seeley
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| | - David C Perry
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| | - Jennifer S Yokoyama
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| | - Suzee E Lee
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| | - Virginia E Sturm
- Department of Neurology, University of California, San Francisco, CA 94158, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA 94143, USA.
| |
Collapse
|
7
|
Mousele C, Holden D, Gnanapavan S. Neurofilaments in neurologic disease. Adv Clin Chem 2024; 123:65-128. [PMID: 39181624 DOI: 10.1016/bs.acc.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Neurofilaments (NFs), major cytoskeletal constituents of neurons, have emerged as universal biomarkers of neuronal injury. Neuroaxonal damage underlies permanent disability in various neurological conditions. It is crucial to accurately quantify and longitudinally monitor this damage to evaluate disease progression, evaluate treatment effectiveness, contribute to novel treatment development, and offer prognostic insights. Neurofilaments show promise for this purpose, as their levels increase with neuroaxonal damage in both cerebrospinal fluid and blood, independent of specific causal pathways. New assays with high sensitivity allow reliable measurement of neurofilaments in body fluids and open avenues to investigate their role in neurological disorders. This book chapter will delve into the evolving landscape of neurofilaments, starting with their structure and cellular functions within neurons. It will then provide a comprehensive overview of their broad clinical value as biomarkers in diseases affecting the central or peripheral nervous system.
Collapse
|
8
|
Pasternak M, Mirza SS, Luciw N, Mutsaerts HJMM, Petr J, Thomas D, Cash D, Bocchetta M, Tartaglia MC, Mitchell SB, Black SE, Freedman M, Tang‐Wai D, Rogaeva E, Russell LL, Bouzigues A, van Swieten JC, Jiskoot LC, Seelaar H, Laforce R, Tiraboschi P, Borroni B, Galimberti D, Rowe JB, Graff C, Finger E, Sorbi S, de Mendonça A, Butler C, Gerhard A, Sanchez‐Valle R, Moreno F, Synofzik M, Vandenberghe R, Ducharme S, Levin J, Otto M, Santana I, Strafella AP, MacIntosh BJ, Rohrer JD, Masellis M. Longitudinal cerebral perfusion in presymptomatic genetic frontotemporal dementia: GENFI results. Alzheimers Dement 2024; 20:3525-3542. [PMID: 38623902 PMCID: PMC11095434 DOI: 10.1002/alz.13750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/16/2024] [Accepted: 01/21/2024] [Indexed: 04/17/2024]
Abstract
INTRODUCTION Effective longitudinal biomarkers that track disease progression are needed to characterize the presymptomatic phase of genetic frontotemporal dementia (FTD). We investigate the utility of cerebral perfusion as one such biomarker in presymptomatic FTD mutation carriers. METHODS We investigated longitudinal profiles of cerebral perfusion using arterial spin labeling magnetic resonance imaging in 42 C9orf72, 70 GRN, and 31 MAPT presymptomatic carriers and 158 non-carrier controls. Linear mixed effects models assessed perfusion up to 5 years after baseline assessment. RESULTS Perfusion decline was evident in all three presymptomatic groups in global gray matter. Each group also featured its own regional pattern of hypoperfusion over time, with the left thalamus common to all groups. Frontal lobe regions featured lower perfusion in those who symptomatically converted versus asymptomatic carriers past their expected age of disease onset. DISCUSSION Cerebral perfusion is a potential biomarker for assessing genetic FTD and its genetic subgroups prior to symptom onset. HIGHLIGHTS Gray matter perfusion declines in at-risk genetic frontotemporal dementia (FTD). Regional perfusion decline differs between at-risk genetic FTD subgroups . Hypoperfusion in the left thalamus is common across all presymptomatic groups. Converters exhibit greater right frontal hypoperfusion than non-converters past their expected conversion date. Cerebral hypoperfusion is a potential early biomarker of genetic FTD.
Collapse
|
9
|
Chatterjee A, Hirsch-Reinshagen V, Scott I, Cashman N, Hsiung GYR. A Systematic Review of the Genetics and Pathology of Psychosis in Frontotemporal Dementia. Can J Neurol Sci 2024; 51:369-378. [PMID: 37385628 DOI: 10.1017/cjn.2023.248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/01/2023]
Abstract
OBJECTIVES Frontotemporal dementia (FTD) patients frequently present with psychosis, which complicates diagnosis and management. In this study, we aim to examine the relationship between psychosis and the most common genetic mutations predisposing to FTD, and in the different pathological subtypes of FTD. DESIGN We conducted a systematic review, searching the literature up to December 2022, and reviewed 50 articles that met our inclusion criteria. From the reviewed articles, we extracted and summarized data regarding the frequency of psychosis and patient characteristics in each major genetic and pathological subtype of FTD. RESULTS Among FTD patients with confirmed genetic mutations or pathological diagnosss, the frequency of psychosis was 24.2%. Among the genetic mutation carriers, C9orf72 mutation carriers had the highest frequency of psychosis (31.4%), whereas GRN (15.0%) and MAPT (9.2%) mutation carriers had lower frequencies of psychosis. MAPT mutation carriers notably developed psychosis at a younger age compared to other genetic groups. The most common psychotic symptoms were delusions among C9orf72 carriers and visual hallucinations among GRN mutation carriers. Among the pathological subtypes, 30% of patients with FUS pathology, 25.3% of patients with TDP-43 pathology, and 16.4% of patients with tau pathology developed psychosis. In the TDP-43 group, subtype B pathology was the most common subtype reported in association with psychosis. CONCLUSION Our systematic review suggests a high frequency of psychosis in specific subgroups of FTD patients. Further studies are required to understand the structural and biological underpinnings of psychosis in FTD.
Collapse
Affiliation(s)
- Atri Chatterjee
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | | | - Imogene Scott
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Neil Cashman
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Ging-Yuek Robin Hsiung
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
10
|
Cagnin A, Pigato G, Pettenuzzo I, Zorzi G, Roiter B, Anglani MG, Bussè C, Mozzetta S, Gabelli C, Campi C, Cecchin D. Data-driven analysis of regional brain metabolism in behavioral frontotemporal dementia and late-onset primary psychiatric diseases with frontal lobe syndrome: A PET/MRI study. Neurobiol Aging 2024; 137:47-54. [PMID: 38422798 DOI: 10.1016/j.neurobiolaging.2024.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 01/10/2024] [Accepted: 01/30/2024] [Indexed: 03/02/2024]
Abstract
Late-onset primary psychiatric disease (PPD) and behavioral frontotemporal dementia (bvFTD) present with a similar frontal lobe syndrome. We compare brain glucose metabolism in bvFTD and late-onset PPD and investigate the metabolic correlates of cognitive and behavioral disturbances through FDG-PET/MRI. We studied 37 bvFTD and 20 late-onset PPD with a mean clinical follow-up of three years. At baseline evaluation, metabolism of the dorsolateral, ventrolateral, orbitofrontal regions and caudate could classify the patients with a diagnostic accuracy of 91% (95% CI: 0.81-0.98%). 45% of PPD showed low-grade hypometabolism in the anterior cingulate and/or parietal regions. Frontal lobe metabolism was normal in 32% of genetic bvFTD and bvFTD with motor neuron signs. Hypometabolism of the frontal and caudate regions could help in distinguishing bvFTD from PPD, except in cases with motor neuron signs and/or genetic bvFTD for which brain metabolism may be less informative.
Collapse
Affiliation(s)
- Annachiara Cagnin
- Neurology Unit, Department of Neuroscience (DNS) University of Padua, Padua, Italy; Padua Neuroscience Center, University of Padua, Padua, Italy.
| | - Giorgio Pigato
- Psychiatry Unit, Department of Neuroscience (DNS), University of Padua, Padua, Italy
| | - Ilaria Pettenuzzo
- Neurology Unit, Department of Neuroscience (DNS) University of Padua, Padua, Italy
| | - Giovanni Zorzi
- Neurology Unit, Department of Neuroscience (DNS) University of Padua, Padua, Italy; Padua Neuroscience Center, University of Padua, Padua, Italy; CRIC, Azienda Ospedale-Università of Padua, Italy
| | - Beatrice Roiter
- Psychiatry Unit, Department of Neuroscience (DNS), University of Padua, Padua, Italy
| | | | - Cinzia Bussè
- Neurology Unit, Department of Neuroscience (DNS) University of Padua, Padua, Italy
| | - Stefano Mozzetta
- Neurology Unit, Department of Neuroscience (DNS) University of Padua, Padua, Italy
| | | | - Cristina Campi
- Nuclear Medicine Unit, Department of Medicine (DIMED), University of Padua, Padua, Italy; Department of Mathematics, University of Genoa and IRCCS Policlinico San Martino Hospital, Genoa
| | - Diego Cecchin
- Padua Neuroscience Center, University of Padua, Padua, Italy; Nuclear Medicine Unit, Department of Medicine (DIMED), University of Padua, Padua, Italy
| |
Collapse
|
11
|
Devenney EM, Tse NY, O’Callaghan C, Kumfor F, Ahmed RM, Caga J, Hazelton JL, Carrick J, Halliday GM, Piguet O, Kiernan MC, Hodges JR. An attentional and working memory theory of hallucination vulnerability in frontotemporal dementia. Brain Commun 2024; 6:fcae123. [PMID: 38725706 PMCID: PMC11081077 DOI: 10.1093/braincomms/fcae123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 01/30/2024] [Accepted: 04/18/2024] [Indexed: 05/12/2024] Open
Abstract
The rate and prevalence of hallucinations in behavioural variant frontotemporal dementia is well established. The mechanisms for underlying vulnerability however are the least well described in FTD compared with other neuropsychiatric conditions, despite the presence of these features significantly complicating the diagnostic process. As such, this present study aimed to provide a detailed characterization of the neural, cognitive and behavioural profile associated with a predisposition to hallucinatory experiences in behavioural variant frontotemporal dementia. In total, 153 patients with behavioural variant frontotemporal dementia were recruited sequentially for this study. A group of patients with well characterized hallucinations and good-quality volumetric MRI scans (n = 23) were genetically and demographically matched to a group without hallucinations (n = 23) and a healthy control cohort (n = 23). All patients were assessed at their initial visit by means of a detailed clinical interview, a comprehensive battery of neuropsychological tests and MRI. Data were analysed according to three levels: (i) the relationship between neural structures, cognition, behaviour and hallucinations in behavioural variant frontotemporal dementia; (ii) the impact of the C9orf72 expansion; and (iii) hallucination subtype on expression of hallucinations. Basic and complex attentional (including divided attention and working memory) and visual function measures differed between groups (all P < 0.001) with hallucinators demonstrating poorer performance, along with evidence of structural changes centred on the prefrontal cortex, caudate and cerebellum (corrected for False Discovery Rate at P < 0.05 with a cluster threshold of 100 contiguous voxels). Attentional processes were also implicated in C9orf72 carriers with hallucinations with structural changes selectively involving the thalamus. Patients with visual hallucinations in isolation showed a similar pattern with emphasis on cerebellar atrophy. Our findings provided novel insights that attentional and visual function subsystems and related distributed brain structures are implicated in the generation of hallucinations in behavioural variant frontotemporal dementia, that dissociate across C9orf72, sporadic behavioural variant frontotemporal dementia and for the visual subtype of hallucinations. This loading on attentional and working memory measures is in line with current mechanistic models of hallucinations that frequently suggest a failure of integration of cognitive and perceptual processes. We therefore propose a novel cognitive and neural model for hallucination predisposition in behavioural variant frontotemporal dementia that aligns with a transdiagnostic model for hallucinations across neurodegeneration and psychiatry.
Collapse
Affiliation(s)
- Emma M Devenney
- Brain & Mind Centre, The University of Sydney, Sydney 2050, Australia
- Neurology Department, Western Sydney Local Health District, Sydney 2145, Australia
| | - Nga Yan Tse
- Brain & Mind Centre, The University of Sydney, Sydney 2050, Australia
- Systems Lab, Department of Psychiatry, The University of Melbourne, Parkville 3052, Australia
| | - Claire O’Callaghan
- Brain & Mind Centre, The University of Sydney, Sydney 2050, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney 2050, Australia
| | - Fiona Kumfor
- Brain & Mind Centre, The University of Sydney, Sydney 2050, Australia
- School of Psychology, The University of Sydney, Sydney 2050, Australia
| | - Rebekah M Ahmed
- Brain & Mind Centre, The University of Sydney, Sydney 2050, Australia
- Memory and Cognition Clinic, Institute of Clinical Neurosciences, Royal Prince Alfred Hospital, Sydney 2050, Australia
| | - Jashelle Caga
- Brain & Mind Centre, The University of Sydney, Sydney 2050, Australia
| | - Jessica L Hazelton
- School of Psychology, The University of Sydney, Sydney 2050, Australia
- Memory and Cognition Clinic, Institute of Clinical Neurosciences, Royal Prince Alfred Hospital, Sydney 2050, Australia
- Cognitive Neuroscience Center (CNC), Universidad de San Andrés, Buenos Aires B1644BID, Argentina
- Latin American Brain Health Institute (Brain Lat), Universidad Adolfo Ibáñez, Santiago 7941169, Chile
| | - James Carrick
- Brain & Mind Centre, The University of Sydney, Sydney 2050, Australia
| | - Glenda M Halliday
- Brain & Mind Centre, The University of Sydney, Sydney 2050, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney 2050, Australia
| | - Olivier Piguet
- Brain & Mind Centre, The University of Sydney, Sydney 2050, Australia
- School of Psychology, The University of Sydney, Sydney 2050, Australia
| | - Matthew C Kiernan
- Neuroscience Research Australia, Randwick 2031, Australia
- Faculty of Medicine and Health, University of New South Wales 2031, Australia
- Neurology Department, South Eastern Sydney Local Health District, NSW 2031, Australia
| | - John R Hodges
- Brain & Mind Centre, The University of Sydney, Sydney 2050, Australia
| |
Collapse
|
12
|
Apostolopoulos A, Kawamoto N, Chow SYA, Tsuiji H, Ikeuchi Y, Shichino Y, Iwasaki S. dCas13-mediated translational repression for accurate gene silencing in mammalian cells. Nat Commun 2024; 15:2205. [PMID: 38467613 PMCID: PMC10928199 DOI: 10.1038/s41467-024-46412-7] [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: 05/19/2023] [Accepted: 02/27/2024] [Indexed: 03/13/2024] Open
Abstract
Current gene silencing tools based on RNA interference (RNAi) or, more recently, clustered regularly interspaced short palindromic repeats (CRISPR)‒Cas13 systems have critical drawbacks, such as off-target effects (RNAi) or collateral mRNA cleavage (CRISPR‒Cas13). Thus, a more specific method of gene knockdown is needed. Here, we develop CRISPRδ, an approach for translational silencing, harnessing catalytically inactive Cas13 proteins (dCas13). Owing to its tight association with mRNA, dCas13 serves as a physical roadblock for scanning ribosomes during translation initiation and does not affect mRNA stability. Guide RNAs covering the start codon lead to the highest efficacy regardless of the translation initiation mechanism: cap-dependent, internal ribosome entry site (IRES)-dependent, or repeat-associated non-AUG (RAN) translation. Strikingly, genome-wide ribosome profiling reveals the ultrahigh gene silencing specificity of CRISPRδ. Moreover, the fusion of a translational repressor to dCas13 further improves the performance. Our method provides a framework for translational repression-based gene silencing in eukaryotes.
Collapse
Grants
- JP20H05784 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- JP21H05278 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- JP21H05734 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- JP23H04268 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- JP20H05786 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- JP23H02415 MEXT | Japan Society for the Promotion of Science (JSPS)
- JP20K07016 MEXT | Japan Society for the Promotion of Science (JSPS)
- JP23K05648 MEXT | Japan Society for the Promotion of Science (JSPS)
- JP21K15023 MEXT | Japan Society for the Promotion of Science (JSPS)
- JP23KJ2175 MEXT | Japan Society for the Promotion of Science (JSPS)
- JP20gm1410001 Japan Agency for Medical Research and Development (AMED)
- JP20gm1410001 Japan Agency for Medical Research and Development (AMED)
- JP23gm6910005h0001 Japan Agency for Medical Research and Development (AMED)
- JP23gm6910005 Japan Agency for Medical Research and Development (AMED)
- JP20gm1410001 Japan Agency for Medical Research and Development (AMED)
- Pioneering Projects MEXT | RIKEN
- Pioneering Projects MEXT | RIKEN
- Exploratory Research Center on Life and Living Systems (ExCELLS), 23EX601
Collapse
Affiliation(s)
- Antonios Apostolopoulos
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, 277-8561, Japan
- RNA Systems Biochemistry Laboratory, RIKEN Cluster for Pioneering Research, Wako, Saitama, 351-0198, Japan
| | - Naohiro Kawamoto
- RNA Systems Biochemistry Laboratory, RIKEN Cluster for Pioneering Research, Wako, Saitama, 351-0198, Japan
| | - Siu Yu A Chow
- Institute of Industrial Science, The University of Tokyo, Meguro-ku, Tokyo, 153-8505, Japan
| | - Hitomi Tsuiji
- Education and Research Division of Pharmacy, School of Pharmacy, Aichi Gakuin University, Nagoya, Aichi, 464-8650, Japan
| | - Yoshiho Ikeuchi
- Institute of Industrial Science, The University of Tokyo, Meguro-ku, Tokyo, 153-8505, Japan
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
- Institute for AI and Beyond, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Yuichi Shichino
- RNA Systems Biochemistry Laboratory, RIKEN Cluster for Pioneering Research, Wako, Saitama, 351-0198, Japan.
| | - Shintaro Iwasaki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, 277-8561, Japan.
- RNA Systems Biochemistry Laboratory, RIKEN Cluster for Pioneering Research, Wako, Saitama, 351-0198, Japan.
| |
Collapse
|
13
|
Chen Y, Spina S, Callahan P, Grinberg LT, Seeley WW, Rosen HJ, Kramer JH, Miller BL, Rankin KP. Pathology-specific patterns of cerebellar atrophy in neurodegenerative disorders. Alzheimers Dement 2024; 20:1771-1783. [PMID: 38109286 PMCID: PMC10984510 DOI: 10.1002/alz.13551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 12/20/2023]
Abstract
INTRODUCTION Associations of cerebellar atrophy with specific neuropathologies in Alzheimer's disease and related dementias (ADRD) have not been systematically analyzed. This study examined cerebellar gray matter volume across major pathological subtypes of ADRD. METHODS Cerebellar gray matter volume was examined using voxel-based morphometry in 309 autopsy-proven ADRD cases and 80 healthy controls. ADRD subtypes included AD, mixed Lewy body disease and AD (LBD-AD), and frontotemporal lobar degeneration (FTLD). Clinical function was assessed using the Clinical Dementia Rating (CDR) scale. RESULTS Distinct patterns of cerebellar atrophy were observed in all ADRD subtypes. Significant cerebellar gray matter changes appeared in the early stages of most subtypes and the very early stages of AD, LBD-AD, FTLD-TDP type A, and progressive supranuclear palsy. Cortical atrophy positively predicted cerebellar atrophy across all subtypes. DISCUSSION Our findings establish pathology-specific profiles of cerebellar atrophy in ADRD and propose cerebellar neuroimaging as a non-invasive biomarker for differential diagnosis and disease monitoring. HIGHLIGHTS Cerebellar atrophy was examined in 309 patients with autopsy-proven neurodegeneration. Distinct patterns of cerebellar atrophy are found in all pathological subtypes of Alzheimer's disease and related dementias (ADRD). Cerebellar atrophy is seen in early-stage (Clinical Dementia Rating [CDR] ≤1) AD, Lewy body dementia (LBD), frontotemporal lobar degeneration with tau-positive inclusion (FTLD-tau), and FTLD-transactive response DNA binding protein (FTLD-TDP). Cortical atrophy positively predicts cerebellar atrophy across all neuropathologies.
Collapse
Affiliation(s)
- Yu Chen
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Salvatore Spina
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Patrick Callahan
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Lea T. Grinberg
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of PathologyUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - William W. Seeley
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of PathologyUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Howard J. Rosen
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Joel H. Kramer
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Bruce L. Miller
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Katherine P. Rankin
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| |
Collapse
|
14
|
Bhatt N, Puangmalai N, Sengupta U, Jerez C, Kidd M, Gandhi S, Kayed R. C9orf72-associated dipeptide protein repeats form A11-positive oligomers in amyotrophic lateral sclerosis and frontotemporal dementia. J Biol Chem 2024; 300:105628. [PMID: 38295729 PMCID: PMC10844744 DOI: 10.1016/j.jbc.2024.105628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/19/2023] [Accepted: 12/24/2023] [Indexed: 02/12/2024] Open
Abstract
Hexanucleotide repeat expansion in C9orf72 is one of the most common causes of amyotrophic lateral sclerosis and frontotemporal dementia. The hexanucleotide expansion, formed by GGGGCC (G4C2) repeats, leads to the production of five dipeptide protein repeats (DPRs) via repeat-associated non-AUG translation. Among the five dipeptide repeats, Gly-Arg, Pro-Arg, and Gly-Ala form neuronal inclusions that contain aggregates of the peptides. Several studies have attempted to model DPR-associated toxicity using various repeat lengths, which suggests a unique conformation that is cytotoxic and is independent of the repeat length. However, the structural characteristics of DPR aggregates have yet to be determined. Increasing evidence suggests that soluble species, such as oligomers, are the main cause of toxicity in proteinopathies, such as Alzheimer's and Parkinson's disease. To investigate the ability of DPRs to aggregate and form toxic oligomers, we adopted a reductionist approach using small dipeptide repeats of 3, 6, and 12. This study shows that DPRs, particularly glycine-arginine and proline-arginine, form oligomers that exhibit distinct dye-binding properties and morphologies. Importantly, we also identified toxic DPR oligomers in amyotrophic lateral sclerosis and frontotemporal dementia postmortem brains that are morphologically similar to those generated recombinantly. This study demonstrates that, similar to soluble oligomers formed by various amyloid proteins, DPR oligomers are toxic, independent of their repeat length.
Collapse
Affiliation(s)
- Nemil Bhatt
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch, Galveston, Texas, USA; Department of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Nicha Puangmalai
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch, Galveston, Texas, USA; Department of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Urmi Sengupta
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch, Galveston, Texas, USA; Department of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Cynthia Jerez
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch, Galveston, Texas, USA; Department of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Madison Kidd
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch, Galveston, Texas, USA; Department of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Shailee Gandhi
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch, Galveston, Texas, USA; Department of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch, Galveston, Texas, USA; Department of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas, USA.
| |
Collapse
|
15
|
Zhou Z, Kim J, Huang AY, Nolan M, Park J, Doan R, Shin T, Miller MB, Chhouk B, Morillo K, Yeh RC, Kenny C, Neil JE, Lee CZ, Ohkubo T, Ravits J, Ansorge O, Ostrow LW, Lagier-Tourenne C, Lee EA, Walsh CA. Somatic Mosaicism in Amyotrophic Lateral Sclerosis and Frontotemporal Dementia Reveals Widespread Degeneration from Focal Mutations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.30.569436. [PMID: 38077003 PMCID: PMC10705414 DOI: 10.1101/2023.11.30.569436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
Although mutations in dozens of genes have been implicated in familial forms of amyotrophic lateral sclerosis (fALS) and frontotemporal degeneration (fFTD), most cases of these conditions are sporadic (sALS and sFTD), with no family history, and their etiology remains obscure. We tested the hypothesis that somatic mosaic mutations, present in some but not all cells, might contribute in these cases, by performing ultra-deep, targeted sequencing of 88 genes associated with neurodegenerative diseases in postmortem brain and spinal cord samples from 404 individuals with sALS or sFTD and 144 controls. Known pathogenic germline mutations were found in 20.6% of ALS, and 26.5% of FTD cases. Predicted pathogenic somatic mutations in ALS/FTD genes were observed in 2.7% of sALS and sFTD cases that did not carry known pathogenic or novel germline mutations. Somatic mutations showed low variant allele fraction (typically <2%) and were often restricted to the region of initial discovery, preventing detection through genetic screening in peripheral tissues. Damaging somatic mutations were preferentially enriched in primary motor cortex of sALS and prefrontal cortex of sFTD, mirroring regions most severely affected in each disease. Somatic mutation analysis of bulk RNA-seq data from brain and spinal cord from an additional 143 sALS cases and 23 controls confirmed an overall enrichment of somatic mutations in sALS. Two adult sALS cases were identified bearing pathogenic somatic mutations in DYNC1H1 and LMNA, two genes associated with pediatric motor neuron degeneration. Our study suggests that somatic mutations in fALS/fFTD genes, and in genes associated with more severe diseases in the germline state, contribute to sALS and sFTD, and that mosaic mutations in a small fraction of cells in focal regions of the nervous system can ultimately result in widespread degeneration.
Collapse
Affiliation(s)
- Zinan Zhou
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA
- Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Junho Kim
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA
- Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Biological Sciences, Sungkyunkwan University, Suwon, South Korea
| | - August Yue Huang
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA
- Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Matthew Nolan
- Department of Neurology, The Sean M. Healey and AMG Center for ALS at Mass General, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Junseok Park
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA
- Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Ryan Doan
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Taehwan Shin
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA
- Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Michael B. Miller
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Brian Chhouk
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA
- Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Katherine Morillo
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA
- Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Rebecca C. Yeh
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA
- Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Connor Kenny
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA
- Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Jennifer E. Neil
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA
- Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA, USA
| | - Chao-Zong Lee
- Department of Neurology, The Sean M. Healey and AMG Center for ALS at Mass General, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Takuya Ohkubo
- Department of Neurology, Yokohama City Minato Red Cross Hospital, Yokohama, Kanagawa, Japan
- Department of Neurosciences, School of Medicine, University of California at San Diego, La Jolla, CA, USA
| | - John Ravits
- Department of Neurosciences, School of Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Olaf Ansorge
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, Oxfordshire, UK
| | - Lyle W. Ostrow
- Department of Neurology, Lewis Katz School of Medicine at Temple University, Philadelphia, USA
| | - Clotilde Lagier-Tourenne
- Department of Neurology, The Sean M. Healey and AMG Center for ALS at Mass General, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Eunjung Alice Lee
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA
- Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Christopher A. Walsh
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA
- Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA, USA
| |
Collapse
|
16
|
Tan RH, McCann H, Shepherd CE, Pinkerton M, Mazumder S, Devenney EM, Adler GL, Rowe DB, Kril J, Halliday GM, Kiernan MC. Heterogeneity of cortical pTDP-43 inclusion morphologies in amyotrophic lateral sclerosis. Acta Neuropathol Commun 2023; 11:180. [PMID: 37957721 PMCID: PMC10642010 DOI: 10.1186/s40478-023-01670-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 10/10/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND Despite the presence of significant cortical pTDP-43 inclusions of heterogeneous morphologies in patients diagnosed with amyotrophic lateral sclerosis (ALS), pathological subclassification is routinely performed in the minority of patients with concomitant frontotemporal dementia (FTD). OBJECTIVE In order to improve current understanding of the presence and relevance of pathological pTDP-43 subtypes in ALS, the present study examined the pattern of cortical pTDP-43 aggregates in 61 ALS cases without FTD. RESULTS Based on the presence, morphology and composition of pTDP-43 pathology, three distinct ALS-TDP subtypes were delineated: (1) A predominant pattern of pTDP-43 granulofilamentous neuronal inclusions (GFNIs) and grains that were immuno-negative for p62 was identified in 18% of cases designated ALS-TDP type E; (2) neuronal cytoplasmic inclusions (NCIs) that were immuno-positive for both pTDP-43 and p62 were observed in 67% of cases assigned ALS-TDP type B; and (3) scarce cortical pTDP-43 and p62 aggregates were identified in 15% of cases coined ALS-TDP type SC (scarce cortical). Quantitative analyses revealed a significantly greater burden of pTDP-43 GFNI and grains in ALS-TDP type E. Principal component analysis demonstrated significant relationships between GFNIs, grains and ALS-TDP subtypes to support the distinction of subtypes E and B. No significant difference in age at death or disease duration was found between ALS-TDP subgroups to suggest that these subtypes represent earlier or later stages of the same disease process. Instead, a significantly higher ALS-TDP stage, indicating greater topographical spread of pTDP-43, was identified in ALS-TDP type E. Alzheimer's disease neuropathological change (ABC score ≥ intermediate) and Lewy body disease (Braak stage ≥ IV) was more prevalent in the ALS-TDP type SC cohort, which also demonstrated a significantly lower overall cognitive score. CONCLUSION In summary, the present study demonstrates that ALS-TDP does not represent a single homogenous neuropathology. We propose the subclassification of ALS-TDP into three distinct subtypes using standard immuno-stains for pTDP-43 and p62 in the motor cortex, which is routinely sampled and evaluated for diagnostic neuropathological characterisation of ALS. We propose that future studies specify both clinicopathological group and pTDP-43 subtype to advance current understanding of the pathogenesis of clinical phenotypes in pTDP-43 proteinopathies, which will have significant relevance to the development of targeted therapies for this heterogeneous disorder.
Collapse
Affiliation(s)
- Rachel H Tan
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia.
- Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Camperdown, NSW, Australia.
| | - Heather McCann
- Neuroscience Research Australia, Randwick, NSW, Australia
| | | | - Monica Pinkerton
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Camperdown, NSW, Australia
| | - Srestha Mazumder
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
| | - Emma M Devenney
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
| | - Gabrielle L Adler
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Camperdown, NSW, Australia
| | - Dominic B Rowe
- Macquarie University Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Jillian Kril
- Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Camperdown, NSW, Australia
- Dementia Research Centre, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Glenda M Halliday
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Camperdown, NSW, Australia
| | - Matthew C Kiernan
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
- Institute of Clinical Neurosciences, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| |
Collapse
|
17
|
Vuic B, Milos T, Tudor L, Nikolac Perkovic M, Konjevod M, Nedic Erjavec G, Farkas V, Uzun S, Mimica N, Svob Strac D. Pharmacogenomics of Dementia: Personalizing the Treatment of Cognitive and Neuropsychiatric Symptoms. Genes (Basel) 2023; 14:2048. [PMID: 38002991 PMCID: PMC10671071 DOI: 10.3390/genes14112048] [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: 10/10/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
Dementia is a syndrome of global and progressive deterioration of cognitive skills, especially memory, learning, abstract thinking, and orientation, usually affecting the elderly. The most common forms are Alzheimer's disease, vascular dementia, and other (frontotemporal, Lewy body disease) dementias. The etiology of these multifactorial disorders involves complex interactions of various environmental and (epi)genetic factors and requires multiple forms of pharmacological intervention, including anti-dementia drugs for cognitive impairment, antidepressants, antipsychotics, anxiolytics and sedatives for behavioral and psychological symptoms of dementia, and other drugs for comorbid disorders. The pharmacotherapy of dementia patients has been characterized by a significant interindividual variability in drug response and the development of adverse drug effects. The therapeutic response to currently available drugs is partially effective in only some individuals, with side effects, drug interactions, intolerance, and non-compliance occurring in the majority of dementia patients. Therefore, understanding the genetic basis of a patient's response to pharmacotherapy might help clinicians select the most effective treatment for dementia while minimizing the likelihood of adverse reactions and drug interactions. Recent advances in pharmacogenomics may contribute to the individualization and optimization of dementia pharmacotherapy by increasing its efficacy and safety via a prediction of clinical outcomes. Thus, it can significantly improve the quality of life in dementia patients.
Collapse
Affiliation(s)
- Barbara Vuic
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| | - Tina Milos
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| | - Lucija Tudor
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| | - Matea Nikolac Perkovic
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| | - Marcela Konjevod
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| | - Gordana Nedic Erjavec
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| | - Vladimir Farkas
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| | - Suzana Uzun
- Department for Biological Psychiatry and Psychogeriatry, University Hospital Vrapce, 10000 Zagreb, Croatia; (S.U.); (N.M.)
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Ninoslav Mimica
- Department for Biological Psychiatry and Psychogeriatry, University Hospital Vrapce, 10000 Zagreb, Croatia; (S.U.); (N.M.)
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Dubravka Svob Strac
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| |
Collapse
|
18
|
Kortazar-Zubizarreta I, Manero-Azua A, Afonso-Agüera J, Perez de Nanclares G. C9ORF72 Gene GGGGCC Hexanucleotide Expansion: A High Clinical Variability from Amyotrophic Lateral Sclerosis to Frontotemporal Dementia. J Pers Med 2023; 13:1396. [PMID: 37763163 PMCID: PMC10532825 DOI: 10.3390/jpm13091396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 09/15/2023] [Accepted: 09/16/2023] [Indexed: 09/29/2023] Open
Abstract
The expanded GGGGCC hexanucleotide repeat (HRE) in the non-coding region of the C9ORF72 gene (C9ORF72-HRE) is the most common genetic cause of familial forms of amyotrophic lateral sclerosis (ALS), FTD, and concurrent ALS and FTD (ALS-FTD), in addition to contributing to the sporadic forms of these diseases. Both syndromes overlap not only genetically, but also sharing similar clinical and neuropathological findings, being considered as a spectrum. In this paper we describe the clinical-genetic findings in a Basque family with different manifestations within the spectrum, our difficulties in reaching the diagnosis, and a narrative review, carried out as a consequence, of the main features associated with C9ORF72-HRE. Family members underwent a detailed clinical assessment, neurological examination, and genetic analysis by repeat-primed PCR. We studied 10 relatives of a symptomatic carrier of the C9ORF72-HRE expansion. Two of them presented the expansion in the pathological range, one of them was symptomatic whereas the other one remained asymptomatic at 72 years. Given the great intrafamilial clinical variability of C9ORF72-HRE, the characterization of patients and family members with particular clinical and genetic subgroups within ALS and FTD becomes a bottleneck for medication development, in particular for genetically focused medicines for ALS and FTD.
Collapse
Affiliation(s)
- Izaro Kortazar-Zubizarreta
- Department of Neurology, Bioaraba Health Research Institute, Araba University Hospital-Txagorritxu, 01009 Vitoria-Gasteiz, Spain
| | - Africa Manero-Azua
- Molecular (Epi) Genetics Laboratory, Bioaraba Health Research Institute, Araba University Hospital, 01009 Vitoria-Gasteiz, Spain; (A.M.-A.); (G.P.d.N.)
| | - Juan Afonso-Agüera
- Department of Neurology, Central University Hospital of Asturias, 33006 Oviedo, Spain;
| | - Guiomar Perez de Nanclares
- Molecular (Epi) Genetics Laboratory, Bioaraba Health Research Institute, Araba University Hospital, 01009 Vitoria-Gasteiz, Spain; (A.M.-A.); (G.P.d.N.)
| |
Collapse
|
19
|
Chatzidimitriou E, Ioannidis P, Aretouli E, Papaliagkas V, Moraitou D. Correlates of Functional Impairment in Patients with the Behavioral Variant of Frontotemporal Dementia: A PRISMA-Compliant Systematic Review. Int J Mol Sci 2023; 24:13810. [PMID: 37762113 PMCID: PMC10531075 DOI: 10.3390/ijms241813810] [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: 08/08/2023] [Revised: 09/04/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
The behavioral variant of frontotemporal dementia (bvFTD) has a devastating effect on multiple domains of daily living. The purpose of this PRISMA-compliant systematic review is to summarize the most important factors associated with functional impairment in this clinical group by critically analyzing the existing literature spanning the period from 2000 to 2023. To be included in the review, a study had to investigate any kind of correlates of functional status in bvFTD patients, using a previously validated instrument of functional assessment. Out of 40 articles assessed for eligibility, 18 met the inclusion criteria. The anatomical pattern of cerebral atrophy at baseline appeared to be the strongest predictor of the rate of functional decline over time, with the frontal-dominant anatomical subtype being associated with a faster rate of functional impairment. Additionally, executive dysfunction as well as apathy appeared to contribute significantly to functional disability in bvFTD patients. A comparative examination of bvFTD in relation to other clinical subtypes of FTD and other types of dementia in general suggests that it is the predominant atrophy of the frontal lobes along with the subsequent unique combination of cognitive and neuropsychiatric manifestations that account for the pronounced functional limitations observed in these individuals, even from the early stages of the disease.
Collapse
Affiliation(s)
- Electra Chatzidimitriou
- Laboratory of Psychology, Department of Cognition, Brain and Behavior, School of Psychology, Faculty of Philosophy, Aristotle University of Thessaloniki (AUTh), 54124 Thessaloniki, Greece;
- Laboratory of Neurodegenerative Diseases, Center of Interdisciplinary Research and Innovation (CIRI-AUTH), Balcan Center, Buildings A & B, 57001 Thessaloniki, Greece
| | - Panagiotis Ioannidis
- B’ Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki (AUTH), 54124 Thessaloniki, Greece
| | - Eleni Aretouli
- Department of Psychology, School of Social Sciences, University of Ioannina, 45500 Ioannina, Greece
| | - Vasileios Papaliagkas
- Department of Biomedical Sciences, School of Health Sciences, International Hellenic University, Alexandrion University Campus, 57400 Thessaloniki, Greece
| | - Despina Moraitou
- Laboratory of Psychology, Department of Cognition, Brain and Behavior, School of Psychology, Faculty of Philosophy, Aristotle University of Thessaloniki (AUTh), 54124 Thessaloniki, Greece;
- Laboratory of Neurodegenerative Diseases, Center of Interdisciplinary Research and Innovation (CIRI-AUTH), Balcan Center, Buildings A & B, 57001 Thessaloniki, Greece
| |
Collapse
|
20
|
Gittings LM, Alsop EB, Antone J, Singer M, Whitsett TG, Sattler R, Van Keuren-Jensen K. Cryptic exon detection and transcriptomic changes revealed in single-nuclei RNA sequencing of C9ORF72 patients spanning the ALS-FTD spectrum. Acta Neuropathol 2023; 146:433-450. [PMID: 37466726 PMCID: PMC10412668 DOI: 10.1007/s00401-023-02599-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/09/2023] [Accepted: 06/12/2023] [Indexed: 07/20/2023]
Abstract
The C9ORF72-linked diseases amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are characterized by the nuclear depletion and cytoplasmic accumulation of TAR DNA-binding protein 43 (TDP-43). Recent studies have shown that the loss of TDP-43 function leads to the inclusion of cryptic exons (CE) in several RNA transcript targets of TDP-43. Here, we show for the first time the detection of CEs in a single-nuclei RNA sequencing (snRNA-seq) dataset obtained from frontal and occipital cortices of C9ORF72 patients that phenotypically span the ALS-FTD disease spectrum. We assessed each cellular cluster for detection of recently described TDP-43-induced CEs. Transcripts containing CEs in the genes STMN2 and KALRN were detected in the frontal cortex of all C9ORF72 disease groups with the highest frequency in excitatory neurons in the C9ORF72-FTD group. Within the excitatory neurons, the cluster with the highest proportion of cells containing a CE had transcriptomic similarities to von Economo neurons, which are known to be vulnerable to TDP-43 pathology and selectively lost in C9ORF72-FTD. Differential gene expression and pathway analysis of CE-containing neurons revealed multiple dysregulated metabolic processes. Our findings reveal novel insights into the transcriptomic changes of neurons vulnerable to TDP-43 pathology.
Collapse
Affiliation(s)
- Lauren M Gittings
- Department of Translational Neuroscience, Barrow Neurological Institute, 350 W Thomas Road, Phoenix, AZ, 85013, USA
| | - Eric B Alsop
- Neurogenomics Division, Translational Genomics Research Institute, part of City of Hope, Phoenix, AZ, USA
| | - Jerry Antone
- Neurogenomics Division, Translational Genomics Research Institute, part of City of Hope, Phoenix, AZ, USA
| | - Mo Singer
- Department of Translational Neuroscience, Barrow Neurological Institute, 350 W Thomas Road, Phoenix, AZ, 85013, USA
| | - Timothy G Whitsett
- Neurogenomics Division, Translational Genomics Research Institute, part of City of Hope, Phoenix, AZ, USA
| | - Rita Sattler
- Department of Translational Neuroscience, Barrow Neurological Institute, 350 W Thomas Road, Phoenix, AZ, 85013, USA.
| | - Kendall Van Keuren-Jensen
- Neurogenomics Division, Translational Genomics Research Institute, part of City of Hope, Phoenix, AZ, USA.
| |
Collapse
|
21
|
Antonioni A, Raho EM, Lopriore P, Pace AP, Latino RR, Assogna M, Mancuso M, Gragnaniello D, Granieri E, Pugliatti M, Di Lorenzo F, Koch G. Frontotemporal Dementia, Where Do We Stand? A Narrative Review. Int J Mol Sci 2023; 24:11732. [PMID: 37511491 PMCID: PMC10380352 DOI: 10.3390/ijms241411732] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Frontotemporal dementia (FTD) is a neurodegenerative disease of growing interest, since it accounts for up to 10% of middle-age-onset dementias and entails a social, economic, and emotional burden for the patients and caregivers. It is characterised by a (at least initially) selective degeneration of the frontal and/or temporal lobe, generally leading to behavioural alterations, speech disorders, and psychiatric symptoms. Despite the recent advances, given its extreme heterogeneity, an overview that can bring together all the data currently available is still lacking. Here, we aim to provide a state of the art on the pathogenesis of this disease, starting with established findings and integrating them with more recent ones. In particular, advances in the genetics field will be examined, assessing them in relation to both the clinical manifestations and histopathological findings, as well as considering the link with other diseases, such as amyotrophic lateral sclerosis (ALS). Furthermore, the current diagnostic criteria will be explored, including neuroimaging methods, nuclear medicine investigations, and biomarkers on biological fluids. Of note, the promising information provided by neurophysiological investigations, i.e., electroencephalography and non-invasive brain stimulation techniques, concerning the alterations in brain networks and neurotransmitter systems will be reviewed. Finally, current and experimental therapies will be considered.
Collapse
Affiliation(s)
- Annibale Antonioni
- Unit of Clinical Neurology, Neurosciences and Rehabilitation Department, University of Ferrara, 44121 Ferrara, Italy
- Doctoral Program in Translational Neurosciences and Neurotechnologies, University of Ferrara, 44121 Ferrara, Italy
| | - Emanuela Maria Raho
- Unit of Clinical Neurology, Neurosciences and Rehabilitation Department, University of Ferrara, 44121 Ferrara, Italy
| | - Piervito Lopriore
- Neurological Institute, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Antonia Pia Pace
- Institute of Radiology, Department of Medicine, University of Udine, University Hospital S. Maria della Misericordia, Azienda Sanitaria-Universitaria Friuli Centrale, 33100 Udine, Italy
| | - Raffaela Rita Latino
- Complex Structure of Neurology, Emergency Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Martina Assogna
- Centro Demenze, Policlinico Tor Vergata, University of Rome 'Tor Vergata', 00133 Rome, Italy
- Non Invasive Brain Stimulation Unit, Istituto di Ricovero e Cura a Carattere Scientifico Santa Lucia, 00179 Rome, Italy
| | - Michelangelo Mancuso
- Neurological Institute, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Daniela Gragnaniello
- Nuerology Unit, Neurosciences and Rehabilitation Department, Ferrara University Hospital, 44124 Ferrara, Italy
| | - Enrico Granieri
- Unit of Clinical Neurology, Neurosciences and Rehabilitation Department, University of Ferrara, 44121 Ferrara, Italy
| | - Maura Pugliatti
- Unit of Clinical Neurology, Neurosciences and Rehabilitation Department, University of Ferrara, 44121 Ferrara, Italy
| | - Francesco Di Lorenzo
- Non Invasive Brain Stimulation Unit, Istituto di Ricovero e Cura a Carattere Scientifico Santa Lucia, 00179 Rome, Italy
| | - Giacomo Koch
- Non Invasive Brain Stimulation Unit, Istituto di Ricovero e Cura a Carattere Scientifico Santa Lucia, 00179 Rome, Italy
- Iit@Unife Center for Translational Neurophysiology, Istituto Italiano di Tecnologia, 44121 Ferrara, Italy
- Section of Human Physiology, Neurosciences and Rehabilitation Department, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
22
|
Martinelli I, Zucchi E, Simonini C, Gianferrari G, Zamboni G, Pinti M, Mandrioli J. The landscape of cognitive impairment in superoxide dismutase 1-amyotrophic lateral sclerosis. Neural Regen Res 2023; 18:1427-1433. [PMID: 36571338 PMCID: PMC10075107 DOI: 10.4103/1673-5374.361535] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Although mutations in the superoxide dismutase 1 gene account for only a minority of total amyotrophic lateral sclerosis cases, the discovery of this gene has been crucial for amyotrophic lateral sclerosis research. Since the identification of superoxide dismutase 1 in 1993, the field of amyotrophic lateral sclerosis genetics has considerably widened, improving our understanding of the diverse pathogenic basis of amyotrophic lateral sclerosis. In this review, we focus on cognitive impairment in superoxide dismutase 1-amyotrophic lateral sclerosis patients. Literature has mostly reported that cognition remains intact in superoxide dismutase 1-amyotrophic lateral sclerosis patients, but recent reports highlight frontal lobe function frailty in patients carrying different superoxide dismutase 1-amyotrophic lateral sclerosis mutations. We thoroughly reviewed all the various mutations reported in the literature to contribute to a comprehensive database of superoxide dismutase 1-amyotrophic lateral sclerosis genotype-phenotype correlation. Such a resource could ultimately improve our mechanistic understanding of amyotrophic lateral sclerosis, enabling a more robust assessment of how the amyotrophic lateral sclerosis phenotype responds to different variants across genes, which is important for the therapeutic strategy targeting genetic mutations. Cognition in superoxide dismutase 1-amyotrophic lateral sclerosis deserves further longitudinal research since this peculiar frailty in patients with similar mutations can be conditioned by external factors, including environment and other unidentified agents including modifier genes.
Collapse
Affiliation(s)
- Ilaria Martinelli
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia; Department of Neurosciences, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Elisabetta Zucchi
- Department of Neurosciences, Azienda Ospedaliero-Universitaria di Modena; Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Cecilia Simonini
- Department of Neurosciences, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Giulia Gianferrari
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Giovanna Zamboni
- Department of Neurosciences, Azienda Ospedaliero-Universitaria di Modena; Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Marcello Pinti
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Jessica Mandrioli
- Department of Neurosciences, Azienda Ospedaliero-Universitaria di Modena; Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
23
|
Cannon AE, Zürrer WE, Zejlon C, Kulcsar Z, Lewandowski S, Piehl F, Granberg T, Ineichen BV. Neuroimaging findings in preclinical amyotrophic lateral sclerosis models-How well do they mimic the clinical phenotype? A systematic review. Front Vet Sci 2023; 10:1135282. [PMID: 37205225 PMCID: PMC10185801 DOI: 10.3389/fvets.2023.1135282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 04/10/2023] [Indexed: 05/21/2023] Open
Abstract
Background and objectives Animal models for motor neuron diseases (MND) such as amyotrophic lateral sclerosis (ALS) are commonly used in preclinical research. However, it is insufficiently understood how much findings from these model systems can be translated to humans. Thus, we aimed at systematically assessing the translational value of MND animal models to probe their external validity with regards to magnetic resonance imaging (MRI) features. Methods In a comprehensive literature search in PubMed and Embase, we retrieved 201 unique publications of which 34 were deemed eligible for qualitative synthesis including risk of bias assessment. Results ALS animal models can indeed present with human ALS neuroimaging features: Similar to the human paradigm, (regional) brain and spinal cord atrophy as well as signal changes in motor systems are commonly observed in ALS animal models. Blood-brain barrier breakdown seems to be more specific to ALS models, at least in the imaging domain. It is noteworthy that the G93A-SOD1 model, mimicking a rare clinical genotype, was the most frequently used ALS proxy. Conclusions Our systematic review provides high-grade evidence that preclinical ALS models indeed show imaging features highly reminiscent of human ALS assigning them a high external validity in this domain. This opposes the high attrition of drugs during bench-to-bedside translation and thus raises concerns that phenotypic reproducibility does not necessarily render an animal model appropriate for drug development. These findings emphasize a careful application of these model systems for ALS therapy development thereby benefiting refinement of animal experiments. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/, identifier: CRD42022373146.
Collapse
Affiliation(s)
| | | | - Charlotte Zejlon
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Zsolt Kulcsar
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | | | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Center of Neurology, Academic Specialist Center, Stockholm Health Services, Stockholm, Sweden
| | - Tobias Granberg
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Benjamin Victor Ineichen
- Center for Reproducible Science, University of Zurich, Zurich, Switzerland
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
24
|
Samra K, Macdougall A, Peakman G, Bouzigues A, Bocchetta M, Cash DM, Greaves CV, Convery RS, van Swieten JC, Jiskoot LC, Seelaar H, Moreno F, Sánchez-Valle R, Laforce R, Graff C, Masellis M, Tartaglia MC, Rowe JB, Borroni B, Finger E, Synofzik M, Galimberti D, Vandenberghe R, de Mendonca A, Butler CR, Gerhard A, Ducharme S, Le Ber I, Tiraboschi P, Santana I, Pasquier F, Levin J, Otto M, Sorbi S, Rohrer JD, Russell LL. Neuropsychiatric symptoms in genetic frontotemporal dementia: developing a new module for Clinical Rating Scales. J Neurol Neurosurg Psychiatry 2023; 94:357-368. [PMID: 36627201 PMCID: PMC10176351 DOI: 10.1136/jnnp-2022-330152] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 12/05/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND Current clinical rating scales in frontotemporal dementia (FTD) often do not incorporate neuropsychiatric features and may therefore inadequately measure disease stage. METHODS 832 participants from the Genetic FTD Initiative (GENFI) were recruited: 522 mutation carriers and 310 mutation-negative controls. The standardised GENFI clinical questionnaire assessed the frequency and severity of 14 neuropsychiatric symptoms: visual, auditory, and tactile hallucinations, delusions, depression, anxiety, irritability/lability, agitation/aggression, euphoria/elation, aberrant motor behaviour, hypersexuality, hyperreligiosity, impaired sleep, and altered sense of humour. A principal component analysis (PCA) was performed to identify key groupings of neuropsychiatric and behavioural items in order to create a new neuropsychiatric module that could be used as an addition to the Clinical Dementia Rating (CDR) plus National Alzheimer's Coordinating Center Behaviour and Language Domains (NACC FTLD) rating scale. RESULTS Overall, 46.4% of mutation carriers had neuropsychiatric symptoms (51.6% C9orf72, 40.8% GRN, 46.6% MAPT) compared with 24.5% of controls. Anxiety and depression were the most common in all genetic groups but fluctuated longitudinally and loaded separately in the PCA. Hallucinations and delusions loaded together, with the remaining neuropsychiatric symptoms loading with the core behavioural features of FTD. These results suggest using a single 'psychosis' neuropsychiatric module consisting of hallucinations and delusions. Adding this to the CDR plus NACC FTLD, called the CDR plus NACC FTLD-N, leads to a number of participants being scored more severely, including those who were previously considered asymptomatic now being scored as prodromal. CONCLUSIONS Neuropsychiatric symptoms occur in mutation carriers at all disease stages across all three genetic groups. However, only psychosis features provided additional staging benefit to the CDR plus NACC FTLD. Inclusion of these features brings us closer to optimising the rating scale for use in trials.
Collapse
Affiliation(s)
- Kiran Samra
- Dementia Reseach Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Amy Macdougall
- London School of Hygiene & Tropical Medicine, London, UK
| | - Georgia Peakman
- Dementia Reseach Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Arabella Bouzigues
- Dementia Reseach Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Martina Bocchetta
- Dementia Reseach Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - David M Cash
- Dementia Reseach Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- Centre for Medical Image Computing, University College London, London, UK
| | - Caroline V Greaves
- Dementia Reseach Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Rhian S Convery
- Dementia Reseach Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | | | | | - Harro Seelaar
- Neurology, Erasmus MC Alzheimer Centre, Rotterdam, The Netherlands
| | - Fermin Moreno
- Cognitive Disorders Unit, Department of Neurology, Donostia University Hospital Gipuzkoa Building, San Sebastian, Spain
| | - Raquel Sánchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Robert Laforce
- Interdisciplinary Memory Clinic, Department of Neurological Sciences, Laval University, Quebec, Quebec, Canada
| | - Caroline Graff
- Center for Alzheimer Research, Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Unit for Hereditary Dementias, Theme Aging, Karolinska University Hospital, Solna, Sweden
| | - Mario Masellis
- Neurology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Maria Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, Ontario, Canada
| | - James B Rowe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Barbara Borroni
- Centre for Ageing Brain and Neurodegenerative Disorders, Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Elizabeth Finger
- Clinical Neurological Sciences, University of Western Ontario, London, Ontario, Canada
| | - Matthis Synofzik
- Dept. of Neurodegenerative Diseases, Eberhard Karls University Tubingen Hertie Institute for Clinical Brain Research, Tubingen, Germany
| | - Daniela Galimberti
- Department of Neurological Sciences, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium
- Neurology Service, KU Leuven University Hospitals Leuven, Leuven, Belgium
| | | | - Christopher R Butler
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Alexander Gerhard
- Division of Neuroscience and Experimental Psychology, The University of Manchester, Manchester, UK
- Departments of Geriatric Medicine and Nuclear Medicine, University of Duisburg-Essen, Duisburg, Germany
| | - Simon Ducharme
- McConnell Brain Imaging Centre, Department of Neurology & Neurosurgery, Montreal Neurological Institute and Hospital, Montreal, Québec, Canada
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, Québec, Canada
| | - Isabelle Le Ber
- Inserm U1127, CNRS UMR 7225, FrontLab - Reference Centre for Rare or Early Dementias, IM2A, Département de Neurologie, Hôpital Universitaire Pitié Salpêtrière, Sorbonne Université, Paris Brain Institute - Institut du Cerveau - ICM, Paris, France
- National Reference Center On Rare Dementias, Groupe Hospitalier La Pitié Salpêtrière-Charles Foix, Paris, France
| | - Pietro Tiraboschi
- Division of Neurology V and Neuropathology, Foundation IRCCS Carlo Besta Neurological Institute, Milano, Italy
| | - Isabel Santana
- Neurology, Hospital and University Centre of Coimbra, Coimbra, Portugal
- Centre for Neuroscience and Cell Biology (CNC).IBILI, University of Coimbra, Coimbra, Portugal
| | - Florence Pasquier
- Inserm U1171, University of Lille, Lille, France
- Memory Clinic, Neurology, CHU Lille, Lille, France
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE), DZNE, Bonn, Germany
- Department of Neurology, Ludwig Maximilians University Munich, Munchen, Germany
| | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Sandro Sorbi
- Neurosciences Drugs and Child Health, University of Florence, Firenze, Italy
- IRCCS Firenze, Fondazione Don Carlo Gnocchi Onlus, Firenze, Italy
| | - Jonathan D Rohrer
- Dementia Reseach Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Lucy L Russell
- Dementia Reseach Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| |
Collapse
|
25
|
Santillo AF, Leuzy A, Honer M, Landqvist Waldö M, Tideman P, Harper L, Ohlsson T, Moes S, Giannini L, Jögi J, Groot C, Ossenkoppele R, Strandberg O, van Swieten J, Smith R, Hansson O. [ 18F]RO948 tau positron emission tomography in genetic and sporadic frontotemporal dementia syndromes. Eur J Nucl Med Mol Imaging 2023; 50:1371-1383. [PMID: 36513817 PMCID: PMC10027632 DOI: 10.1007/s00259-022-06065-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022]
Abstract
PURPOSE To examine [18F]RO948 retention in FTD, sampling the underlying protein pathology heterogeneity. METHODS A total of 61 individuals with FTD (n = 35), matched cases of AD (n = 13) and Aβ-negative cognitively unimpaired individuals (n = 13) underwent [18F]RO948PET and MRI. FTD included 21 behavioral variant FTD (bvFTD) cases, 11 symptomatic C9orf72 mutation carriers, one patient with non-genetic bvFTD-ALS, one individual with bvFTD due to a GRN mutation, and one due to a MAPT mutation (R406W). Tracer retention was examined using a region-of-interest and voxel-wise approaches. Two individuals (bvFTD due to C9orf72) underwent postmortem neuropathological examination. Tracer binding was additionally assessed in vitro using [3H]RO948 autoradiography in six separate cases. RESULTS [18F]RO948 retention across ROIs was clearly lower than in AD and comparable to that in Aβ-negative cognitively unimpaired individuals. Only minor loci of tracer retention were seen in bvFTD; these did not overlap with the observed cortical atrophy in the cases, the expected pattern of atrophy, nor the expected or verified protein pathology distribution. Autoradiography analyses showed no specific [3H]RO948 binding. The R406W MAPT mutation carriers were clear exceptions with AD-like retention levels and specific in-vitro binding. CONCLUSION [18F]RO948 uptake is not significantly increased in the majority of FTD patients, with a clear exception being specific MAPT mutations.
Collapse
Affiliation(s)
- Alexander F Santillo
- Department of Clinical Sciences, Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund/Malmö, Sweden.
- Memory Clinic, Skåne University Hospital, SE-20502, Malmö, Sweden.
| | - Antoine Leuzy
- Department of Clinical Sciences, Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund/Malmö, Sweden
| | - Michael Honer
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| | - Maria Landqvist Waldö
- Clinical Sciences Helsingborg, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Pontus Tideman
- Department of Clinical Sciences, Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund/Malmö, Sweden
| | - Luke Harper
- Department of Clinical Sciences, Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund/Malmö, Sweden
| | - Tomas Ohlsson
- Radiation Physics, Skane University Hospital, Scania, Sweden
| | - Svenja Moes
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| | - Lucia Giannini
- Alzheimer Center, Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jonas Jögi
- Clinical Physiology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Colin Groot
- Department of Clinical Sciences, Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund/Malmö, Sweden
| | - Rik Ossenkoppele
- Department of Clinical Sciences, Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund/Malmö, Sweden
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Olof Strandberg
- Department of Clinical Sciences, Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund/Malmö, Sweden
| | - John van Swieten
- Alzheimer Center, Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ruben Smith
- Department of Clinical Sciences, Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund/Malmö, Sweden
- Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Oskar Hansson
- Department of Clinical Sciences, Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund/Malmö, Sweden
- Memory Clinic, Skåne University Hospital, SE-20502, Malmö, Sweden
| |
Collapse
|
26
|
Premi E, Pengo M, Mattioli I, Cantoni V, Dukart J, Gasparotti R, Buratti E, Padovani A, Bocchetta M, Todd EG, Bouzigues A, Cash DM, Convery RS, Russell LL, Foster P, Thomas DL, van Swieten JC, Jiskoot LC, Seelaar H, Galimberti D, Sanchez-Valle R, Laforce R, Moreno F, Synofzik M, Graff C, Masellis M, Tartaglia MC, Rowe JB, Tsvetanov KA, Vandenberghe R, Finger E, Tiraboschi P, de Mendonça A, Santana I, Butler CR, Ducharme S, Gerhard A, Levin J, Otto M, Sorbi S, Le Ber I, Pasquier F, Rohrer JD, Borroni B. Early neurotransmitters changes in prodromal frontotemporal dementia: A GENFI study. Neurobiol Dis 2023; 179:106068. [PMID: 36898614 DOI: 10.1016/j.nbd.2023.106068] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/01/2023] [Accepted: 03/04/2023] [Indexed: 03/12/2023] Open
Abstract
BACKGROUND Neurotransmitters deficits in Frontotemporal Dementia (FTD) are still poorly understood. Better knowledge of neurotransmitters impairment, especially in prodromal disease stages, might tailor symptomatic treatment approaches. METHODS In the present study, we applied JuSpace toolbox, which allowed for cross-modal correlation of Magnetic Resonance Imaging (MRI)-based measures with nuclear imaging derived estimates covering various neurotransmitter systems including dopaminergic, serotonergic, noradrenergic, GABAergic and glutamatergic neurotransmission. We included 392 mutation carriers (157 GRN, 164 C9orf72, 71 MAPT), together with 276 non-carrier cognitively healthy controls (HC). We tested if the spatial patterns of grey matter volume (GMV) alterations in mutation carriers (relative to HC) are correlated with specific neurotransmitter systems in prodromal (CDR® plus NACC FTLD = 0.5) and in symptomatic (CDR® plus NACC FTLD≥1) FTD. RESULTS In prodromal stages of C9orf72 disease, voxel-based brain changes were significantly associated with spatial distribution of dopamine and acetylcholine pathways; in prodromal MAPT disease with dopamine and serotonin pathways, while in prodromal GRN disease no significant findings were reported (p < 0.05, Family Wise Error corrected). In symptomatic FTD, a widespread involvement of dopamine, serotonin, glutamate and acetylcholine pathways across all genetic subtypes was found. Social cognition scores, loss of empathy and poor response to emotional cues were found to correlate with the strength of GMV colocalization of dopamine and serotonin pathways (all p < 0.01). CONCLUSIONS This study, indirectly assessing neurotransmitter deficits in monogenic FTD, provides novel insight into disease mechanisms and might suggest potential therapeutic targets to counteract disease-related symptoms.
Collapse
Affiliation(s)
- Enrico Premi
- Neurology, Department of Neurological and Vision Sciences, ASST Spedali Civili, Brescia, Italy
| | - Marta Pengo
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Irene Mattioli
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Valentina Cantoni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Juergen Dukart
- Institute of Neuroscience and Medicine, Brain & Behaviour (INM-7), Research CentreJülich, Jülich, Germany; Institute of Systems Neuroscience, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Roberto Gasparotti
- Neuroradiology Unit, Department of Medical and Surgical Specialties, University of Brescia, Brescia, Italy
| | | | - Alessandro Padovani
- Neurology, Department of Neurological and Vision Sciences, ASST Spedali Civili, Brescia, Italy; Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Martina Bocchetta
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom; Centre for Cognitive and Clinical Neuroscience, Division of Psychology, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, London, United Kingdom
| | - Emily G Todd
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Arabella Bouzigues
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - David M Cash
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom; Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - Rhian S Convery
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Lucy L Russell
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Phoebe Foster
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - David L Thomas
- Neuroradiological Academic Unit, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - John C van Swieten
- Department of Neurology and Alzheimer center, Erasmus Medical Center Rotterdam, the Netherlands
| | - Lize C Jiskoot
- Department of Neurology and Alzheimer center, Erasmus Medical Center Rotterdam, the Netherlands
| | - Harro Seelaar
- Department of Neurology and Alzheimer center, Erasmus Medical Center Rotterdam, the Netherlands
| | - Daniela Galimberti
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy; Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Raquel Sanchez-Valle
- Neurology Department, Hospital Clinic, Institut d'Investigacions Biomèdiques, Barcelona, Spain
| | - Robert Laforce
- Clinique Interdisciplinaire de Mémoire, Département des Sciences Neurologiques, CHU de Québec, Faculté de Médecine, Université Laval, Québec, Canada
| | - Fermin Moreno
- Hospital Universitario Donostia, San Sebastian, Spain
| | - Matthis Synofzik
- Division Translational Genomics of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research (HIH), University of Tübingen, Tübingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Caroline Graff
- Karolinska Institutet, Department NVS, Division of Neurogeriatrics, Stockholm, Sweden; Unit for Hereditray Dementia, Theme Aging, Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Mario Masellis
- Campbell Cognitive Neurology Research Unit, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Maria Carmela Tartaglia
- Toronto Western Hospital, Tanz Centre for Research in Neurodegenerative Disease, Toronto, ON, Canada
| | - James B Rowe
- Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust and Medical Research Council Cognition and brain Sciences Unit, University of Cambridge, Cambridge, United Kingdom
| | - Kamen A Tsvetanov
- Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust and Medical Research Council Cognition and brain Sciences Unit, University of Cambridge, Cambridge, United Kingdom
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Elizabeth Finger
- Department of Clinical Neurological Sciences, University of Western Ontario, London, ON, Canada
| | - Pietro Tiraboschi
- Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Neurologico Carlo Besta, Milan, Italy
| | | | - Isabel Santana
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Portugal
| | - Chris R Butler
- Department of Clinical Neurology, University of Oxford, Oxford, United Kingdom
| | - Simon Ducharme
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Alexander Gerhard
- Division of Neuroscience and Experimental Psychology, Wolfson Molecular Imaging Centre, University of Manchester, Manchester, United Kingdom; Departments of Geriatric Medicine and Nuclear Medicine, University of Duisburg-Essen, Germany
| | - Johannes Levin
- Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster of System Neurology, Munich, Germany
| | - Markus Otto
- Department of Neurology, University Hospital Halle, Halle, Germany
| | - Sandro Sorbi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy; IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Isabelle Le Ber
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau - ICM, Inserm U1127, CNRS UMR 7225, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Centre de référence des démences rares ou précoces, IM2A, Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Reference Network for Rare Neurological Diseases (ERN-RND)
| | - Florence Pasquier
- University of Lille, France; Inserm 1172, Lille, France; CHU, CNR-MAJ, Labex Distalz, LiCEND Lille, France
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Barbara Borroni
- Neurology, Department of Neurological and Vision Sciences, ASST Spedali Civili, Brescia, Italy; Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.
| |
Collapse
|
27
|
Jiskoot LC, Russell LL, Peakman G, Convery RS, Greaves CV, Bocchetta M, Poos JM, Seelaar H, Giannini LAA, van Swieten JC, van Minkelen R, Pijnenburg YAL, Rowe JB, Borroni B, Galimberti D, Masellis M, Tartaglia C, Finger E, Butler CR, Graff C, Laforce R, Sanchez-Valle R, de Mendonça A, Moreno F, Synofzik M, Vandenberghe R, Ducharme S, le Ber I, Levin J, Otto M, Pasquier F, Santana I, Cash DM, Thomas D, Rohrer JD. The Benson Complex Figure Test detects deficits in visuoconstruction and visual memory in symptomatic familial frontotemporal dementia: A GENFI study. J Neurol Sci 2023; 446:120590. [PMID: 36812822 DOI: 10.1016/j.jns.2023.120590] [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: 10/26/2022] [Revised: 01/24/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023]
Abstract
OBJECTIVE Sensitive cognitive markers are still needed for frontotemporal dementia (FTD). The Benson Complex Figure Test (BCFT) is an interesting candidate test, as it assesses visuospatial, visual memory, and executive abilities, allowing the detection of multiple mechanisms of cognitive impairment. To investigate differences in BCFT Copy, Recall and Recognition in presymptomatic and symptomatic FTD mutation carriers, and to explore its cognitive and neuroimaging correlates. METHOD We included cross-sectional data from 332 presymptomatic and 136 symptomatic mutation carriers (GRN, MAPT or C9orf72 mutations), and 290 controls in the GENFI consortium. We examined gene-specific differences between mutation carriers (stratified by CDR® NACC-FTLD score) and controls using Quade's / Pearson Χ2 tests. We investigated associations with neuropsychological test scores and grey matter volume using partial correlations and multiple regression models respectively. RESULTS No significant differences were found between groups at CDR® NACC-FTLD 0-0.5. Symptomatic GRN and C9orf72 mutation carriers had lower Copy scores at CDR® NACC-FTLD ≥2. All three groups had lower Recall scores at CDR® NACC-FTLD ≥2, with MAPT mutation carriers starting at CDR® NACC-FTLD ≥1. All three groups had lower Recognition scores at CDR® NACC FTLD ≥2. Performance correlated with tests for visuoconstruction, memory, and executive function. Copy scores correlated with frontal-subcortical grey matter atrophy, while Recall scores correlated with temporal lobe atrophy. CONCLUSIONS In the symptomatic stage, the BCFT identifies differential mechanisms of cognitive impairment depending on the genetic mutation, corroborated by gene-specific cognitive and neuroimaging correlates. Our findings suggest that impaired performance on the BCFT occurs relatively late in the genetic FTD disease process. Therefore its potential as cognitive biomarker for upcoming clinical trials in presymptomatic to early-stage FTD is most likely limited.
Collapse
Affiliation(s)
- Lize C Jiskoot
- Department of Neurology, Erasmus Medical Center, Rotterdam, the Netherlands; Dementia Research Centre, University College London, London, UK.
| | - Lucy L Russell
- Dementia Research Centre, University College London, London, UK.
| | - Georgia Peakman
- Dementia Research Centre, University College London, London, UK.
| | - Rhian S Convery
- Dementia Research Centre, University College London, London, UK.
| | | | | | - Jackie M Poos
- Department of Neurology, Erasmus Medical Center, Rotterdam, the Netherlands.
| | - Harro Seelaar
- Department of Neurology, Erasmus Medical Center, Rotterdam, the Netherlands.
| | - Lucia A A Giannini
- Department of Neurology, Erasmus Medical Center, Rotterdam, the Netherlands.
| | - John C van Swieten
- Department of Neurology, Erasmus Medical Center, Rotterdam, the Netherlands.
| | - Rick van Minkelen
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, the Netherlands.
| | - Yolande A L Pijnenburg
- Alzheimer Center and Department of Neurology, Neuroscience Campus Amsterdam, Amsterdam, the Netherlands.
| | - James B Rowe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.
| | - Barbara Borroni
- Centre for Neurodegenerative Disorders, Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Daniela Galimberti
- University of Milan, Centro Dino Ferrari, Milan, Italy; Fondazione IRCCS Ca' Granda, Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy.
| | - Mario Masellis
- Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, University of Toronto, Toronto, Canada.
| | - Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada..
| | - Elizabeth Finger
- Department of Clinical Neurological Sciences, University of Western Ontario, London, Ontario, Canada.
| | - Chris R Butler
- Department of Clinical Neurology, University of Oxford, Oxford, UK.
| | - Caroline Graff
- Department of Geriatric Medicine, Karolinska University Hospital-Huddinge, Stockholm, Sweden..
| | - Robert Laforce
- Clinique Interdisciplinaire de Mémoire, Département des Sciences Neurologiques, Université Laval, Québec, Canada.
| | - Raquel Sanchez-Valle
- Alzheimer's disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Barcelona, Spain.
| | | | - Fermin Moreno
- Cognitive Disorders Unit, Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa, Spain
| | - Matthis Synofzik
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Tübingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium.
| | - Simon Ducharme
- Department of Psychiatry, McGill University Health Centre, McGill University, Montreal, Québec, Canada.
| | - Isabelle le Ber
- Paris Brain Institute - Institut du Cerveau - Hôpital Pitié-Salpêtrière, Sorbonne Université, Paris, France; Centre de référence des démences rares ou précoces, IM2A, Département de Neurologie, Hôpital Pitié-Salpêtrière, Paris, France; Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians-University, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany.
| | - Florence Pasquier
- University of Lille, Lille, France; Inserm, 1172 Lille, France; CHU, CNR-MAJ, Labex Distalz, LiCEND, Lille, France.
| | - Isabel Santana
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| | - David M Cash
- Dementia Research Centre, University College London, London, UK.
| | - David Thomas
- Dementia Research Centre, University College London, London, UK.
| | | | | |
Collapse
|
28
|
Samra K, MacDougall AM, Peakman G, Bouzigues A, Bocchetta M, Cash DM, Greaves CV, Convery RS, van Swieten JC, Jiskoot L, Seelaar H, Moreno F, Sanchez-Valle R, Laforce R, Graff C, Masellis M, Tartaglia C, Rowe JB, Borroni B, Finger E, Synofzik M, Galimberti D, Vandenberghe R, de Mendonça A, Butler CR, Gerhard A, Ducharme S, Le Ber I, Tiraboschi P, Santana I, Pasquier F, Levin J, Otto M, Sorbi S, Rohrer JD, Russell LL. Motor symptoms in genetic frontotemporal dementia: developing a new module for clinical rating scales. J Neurol 2023; 270:1466-1477. [PMID: 36385202 PMCID: PMC9971048 DOI: 10.1007/s00415-022-11442-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To investigate the optimal method of adding motor features to a clinical rating scale for frontotemporal dementia (FTD). METHODS Eight hundred and thirty-two participants from the international multicentre Genetic FTD Initiative (GENFI) study were recruited: 522 mutation carriers (with C9orf72, GRN and MAPT mutations) and 310 mutation-negative controls. A standardised clinical questionnaire was used to assess eight motor symptoms (dysarthria, dysphagia, tremor, slowness, weakness, gait disorder, falls and functional difficulties using hands). Frequency and severity of each motor symptom was assessed, and a principal component analysis (PCA) was performed to identify how the different motor symptoms loaded together. Finally, addition of a motor component to the CDR® plus NACC FTLD was investigated (CDR® plus NACC FTLD-M). RESULTS 24.3% of mutation carriers had motor symptoms (31.7% C9orf72, 18.8% GRN, 19.3% MAPT) compared to 6.8% of controls. Slowness and gait disorder were the commonest in all genetic groups while tremor and falls were the least frequent. Symptom severity scores were similar to equivalent physical motor examination scores. PCA revealed that all motor symptoms loaded together so a single additional motor component was added to the CDR® plus NACC FTLD to form the CDR® plus NACC FTLD-M. Individual global scores were more severe with the CDR® plus NACC FTLD-M, and no patients with a clinically diagnosed motor disorder (ALS/FTD-ALS or parkinsonism) were classified anymore as asymptomatic (unlike the CDR® plus NACC FTLD alone). CONCLUSIONS Motor features are present in mutation carriers at all disease stages across all three genetic groups. Inclusion of motor symptoms in a rating scale that can be used in future clinical trials will not only ensure a more accurate severity measure is recorded but that a wider spectrum of FTD phenotypes can be included in the same trial.
Collapse
Affiliation(s)
- Kiran Samra
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK.
| | - Amy M MacDougall
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London, UK
| | - Georgia Peakman
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Arabella Bouzigues
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Martina Bocchetta
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - David M Cash
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Caroline V Greaves
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Rhian S Convery
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | | | - Lize Jiskoot
- Department of Neurology, Erasmus Medical Centre, Rotterdam, Netherlands
| | - Harro Seelaar
- Department of Neurology, Erasmus Medical Centre, Rotterdam, Netherlands
| | - Fermin Moreno
- Cognitive Disorders Unit, Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain
- Neuroscience Area, Biodonostia Health Research Institute, Gipuzkoa, San Sebastian, Spain
| | - Raquel Sanchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Barcelona, Spain
| | - Robert Laforce
- Clinique Interdisciplinaire de Mémoire, Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Quebec City, Quebec, Canada
| | - Caroline Graff
- Center for Alzheimer Research, Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Bioclinicum, Karolinska Institutet, Solna, Sweden
- Unit for Hereditary Dementias, Theme Aging, Karolinska University Hospital, Solna, Sweden
| | - Mario Masellis
- Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, University of Toronto, Toronto, Canada
| | - Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - James B Rowe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Barbara Borroni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Elizabeth Finger
- Department of Clinical Neurological Sciences, University of Western Ontario, London, ON, Canada
| | - Matthis Synofzik
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Tübingen, Germany
- Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Daniela Galimberti
- Fondazione Ca' Granda, IRCCS Ospedale Policlinico, Milan, Italy
- University of Milan, Centro Dino Ferrari, Milan, Italy
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium
- Neurology Service, University Hospitals Leuven, Leuven, Belgium
- Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Alexandre de Mendonça
- Laboratory of Neurosciences, Institute of Molecular Medicine, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Chris R Butler
- Nuffield Department of Clinical Neurosciences, Medical Sciences Division, University of Oxford, Oxford, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Alexander Gerhard
- Division of Neuroscience and Experimental Psychology, Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
- Departments of Geriatric Medicine and Nuclear Medicine, University of Duisburg-Essen, Essen, Germany
| | - Simon Ducharme
- Department of Psychiatry, McGill University Health Centre, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Isabelle Le Ber
- Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière, Sorbonne Université, Paris, France
- Centre de référence des démences rares ou précoces, IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière, Paris, France
- Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière, Paris, France
- Reference Network for Rare Neurological Diseases (ERN-RND), Paris, France
| | | | - Isabel Santana
- Neurology Service, Faculty of Medicine, University Hospital of Coimbra (HUC), University of Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Florence Pasquier
- Univ Lille, Lille, France
- Inserm 1172, Lille, France
- CHU, CNR-MAJ, Labex DistalzLiCEND Lille, Lille, France
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians Universität München, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Sandro Sorbi
- Department of Neurofarba, University of Florence, Florence, Italy
- IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Lucy L Russell
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| |
Collapse
|
29
|
McKenna MC, Lope J, Bede P, Tan EL. Thalamic pathology in frontotemporal dementia: Predilection for specific nuclei, phenotype-specific signatures, clinical correlates, and practical relevance. Brain Behav 2023; 13:e2881. [PMID: 36609810 PMCID: PMC9927864 DOI: 10.1002/brb3.2881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/17/2022] [Accepted: 12/18/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Frontotemporal dementia (FTD) phenotypes are classically associated with distinctive cortical atrophy patterns and regional hypometabolism. However, the spectrum of cognitive and behavioral manifestations in FTD arises from multisynaptic network dysfunction. The thalamus is a key hub of several corticobasal and corticocortical circuits. The main circuits relayed via the thalamic nuclei include the dorsolateral prefrontal circuit, the anterior cingulate circuit, and the orbitofrontal circuit. METHODS In this paper, we have reviewed evidence for thalamic pathology in FTD based on radiological and postmortem studies. Original research papers were systematically reviewed for preferential involvement of specific thalamic regions, for phenotype-associated thalamic disease burden patterns, characteristic longitudinal changes, and genotype-associated thalamic signatures. Moreover, evidence for presymptomatic thalamic pathology was also reviewed. Identified papers were systematically scrutinized for imaging methods, cohort sizes, clinical profiles, clinicoradiological associations, and main anatomical findings. The findings of individual research papers were amalgamated for consensus observations and their study designs further evaluated for stereotyped shortcomings. Based on the limitations of existing studies and conflicting reports in low-incidence FTD variants, we sought to outline future research directions and pressing research priorities. RESULTS FTD is associated with focal thalamic degeneration. Phenotype-specific thalamic traits mirror established cortical vulnerability patterns. Thalamic nuclei mediating behavioral and language functions are preferentially involved. Given the compelling evidence for considerable thalamic disease burden early in the course of most FTD subtypes, we also reflect on the practical relevance, diagnostic role, prognostic significance, and monitoring potential of thalamic metrics in FTD. CONCLUSIONS Cardinal manifestations of FTD phenotypes are likely to stem from thalamocortical circuitry dysfunction and are not exclusively driven by focal cortical changes.
Collapse
Affiliation(s)
- Mary Clare McKenna
- Computational Neuroimaging Group, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Department of Neurology, St James's Hospital, Dublin, Ireland
| | - Jasmin Lope
- Computational Neuroimaging Group, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Peter Bede
- Computational Neuroimaging Group, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Department of Neurology, St James's Hospital, Dublin, Ireland
| | - Ee Ling Tan
- Computational Neuroimaging Group, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
30
|
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
|
31
|
Mazumder S, Kiernan MC, Halliday GM, Timmins HC, Mahoney CJ. The contribution of brain banks to knowledge discovery in amyotrophic lateral sclerosis: A systematic review. Neuropathol Appl Neurobiol 2022; 48:e12845. [PMID: 35921237 PMCID: PMC9804699 DOI: 10.1111/nan.12845] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 06/17/2022] [Accepted: 07/23/2022] [Indexed: 01/09/2023]
Abstract
Over the past decade, considerable efforts have been made to accelerate pathophysiological understanding of fatal neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS) with brain banks at the forefront. In addition to exploratory disease mechanisms, brain banks have aided our understanding with regard to clinical diagnosis, genetics and cell biology. Across neurodegenerative disorders, the impact of brain tissue in ALS research has yet to be quantified. This review aims to outline (i) how postmortem tissues from brain banks have influenced our understanding of ALS over the last 15 years, (ii) correlate the location of dedicated brain banks with the geographical prevalence of ALS, (iii) identify the frequency of features reported from postmortem studies and (iv) propose common reporting standards for materials obtained from dedicated brain banks. A systematic review was conducted using PubMed and Web of Science databases using key words. From a total of 1439 articles, 73 articles were included in the final review, following PRISMA guidelines. Following a thematic analysis, articles were categorised into five themes; clinico-pathological (13), genetic (20), transactive response DNA binding protein 43 (TDP-43) pathology (12), non-TDP-43 neuronal pathology (nine) and extraneuronal pathology (19). Research primarily focused on the genetics of ALS, followed by protein pathology. About 63% of the brain banks were in the United States of America and United Kingdom. The location of brain banks overall aligned with the incidence of ALS worldwide with 88% of brain banks situated in Europe and North America. An overwhelming lack of consistency in reporting and replicability was observed, strengthening the need for a standardised reporting system. Overall, postmortem material from brain banks generated substantial new knowledge in areas of genetics and proteomics and supports their ongoing role as an important research tool.
Collapse
Affiliation(s)
- Srestha Mazumder
- ForeFront Clinic, Brain and Mind CentreUniversity of SydneySydneyNew South WalesAustralia
| | - Matthew C. Kiernan
- ForeFront Clinic, Brain and Mind CentreUniversity of SydneySydneyNew South WalesAustralia
| | - Glenda M. Halliday
- Frontier, Brain and Mind CentreUniversity of SydneySydneyNew South WalesAustralia
| | - Hannah C. Timmins
- ForeFront Clinic, Brain and Mind CentreUniversity of SydneySydneyNew South WalesAustralia
| | - Colin J. Mahoney
- ForeFront Clinic, Brain and Mind CentreUniversity of SydneySydneyNew South WalesAustralia
| |
Collapse
|
32
|
Chipika RH, Mulkerrin G, Pradat PF, Murad A, Ango F, Raoul C, Bede P. Cerebellar pathology in motor neuron disease: neuroplasticity and neurodegeneration. Neural Regen Res 2022; 17:2335-2341. [PMID: 35535867 PMCID: PMC9120698 DOI: 10.4103/1673-5374.336139] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Amyotrophic lateral sclerosis is a relentlessly progressive multi-system condition. The clinical picture is dominated by upper and lower motor neuron degeneration, but extra-motor pathology is increasingly recognized, including cerebellar pathology. Post-mortem and neuroimaging studies primarily focus on the characterization of supratentorial disease, despite emerging evidence of cerebellar degeneration in amyotrophic lateral sclerosis. Cardinal clinical features of amyotrophic lateral sclerosis, such as dysarthria, dysphagia, cognitive and behavioral deficits, saccade abnormalities, gait impairment, respiratory weakness and pseudobulbar affect are likely to be exacerbated by co-existing cerebellar pathology. This review summarizes in vivo and post mortem evidence for cerebellar degeneration in amyotrophic lateral sclerosis. Structural imaging studies consistently capture cerebellar grey matter volume reductions, diffusivity studies readily detect both intra-cerebellar and cerebellar peduncle white matter alterations and functional imaging studies commonly report increased functional connectivity with supratentorial regions. Increased functional connectivity is commonly interpreted as evidence of neuroplasticity representing compensatory processes despite the lack of post-mortem validation. There is a scarcity of post-mortem studies focusing on cerebellar alterations, but these detect pTDP-43 in cerebellar nuclei. Cerebellar pathology is an overlooked facet of neurodegeneration in amyotrophic lateral sclerosis despite its contribution to a multitude of clinical symptoms, widespread connectivity to spinal and supratentorial regions and putative role in compensating for the degeneration of primary motor regions.
Collapse
Affiliation(s)
- Rangariroyashe H Chipika
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Grainne Mulkerrin
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | | - Aizuri Murad
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Fabrice Ango
- The Neuroscience Institute of Montpellier (INM), INSERM, CNRS, Montpellier, France
| | - Cédric Raoul
- The Neuroscience Institute of Montpellier (INM), INSERM, CNRS, Montpellier, France
| | - Peter Bede
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Pitié-Salpêtrière University Hospital, Sorbonne University, Paris, France
| |
Collapse
|
33
|
Snowden JS. Changing perspectives on frontotemporal dementia: A review. J Neuropsychol 2022. [DOI: 10.1111/jnp.12297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/04/2022] [Accepted: 10/05/2022] [Indexed: 11/05/2022]
Affiliation(s)
- Julie S. Snowden
- Cerebral Function Unit, Manchester Centre for Neurosciences Salford Royal NHS Foundation Trust Salford UK
- Division of Neuroscience & Experimental Psychology School of Biological Sciences, University of Manchester Manchester UK
| |
Collapse
|
34
|
Sawyer RP, Stone HK, Salim H, Lu X, Weirauch MT, Kottyan L. Frontotemporal degeneration genetic risk loci and transcription regulation as a possible mechanistic link to disease risk. Medicine (Baltimore) 2022; 101:e31078. [PMID: 36253972 PMCID: PMC9575772 DOI: 10.1097/md.0000000000031078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
The etiology of Frontotemporal Degeneration (FTD) is not well understood. Genetic studies have established common genetic variants (GVs) that are associated with increased FTD risk. We review previous genome wide association studies (GWAS) of FTD and nominate specific transcriptional regulators as potential key players in the etiology of this disease. A list of GVs associated with FTD was compiled from published GWAS. The regulatory element locus intersection (RELI) tool was used to calculate the enrichment of the overlap between disease risk GVs and the genomic coordinates of data from a collection of >10,000 chromatin immunoprecipitation (ChIP-seq) experiments. After linkage disequilibrium expansion of the previously reported tag associated GVs, we identified 914 GV at 47 independent risk loci. Using the RELI algorithm, we identified several transcriptional regulators with enriched binding at FTD risk loci (0.05 < corrected P value <1.18 × 10-27), including Tripartite motif-containing 28 (TRIM28) and Chromodomain-Helicase DNA-binding 1 (CHD1) which have previously observed roles in FTD. FTD is a complex disease, and immune dysregulation has been previously implicated as a potential underlying cause. This assessment of established FTD risk loci and analysis of possible function implicates transcriptional dysregulation, and specifically particular transcriptional regulators with known roles in the immune response as important in the genetic etiology of FTD.
Collapse
Affiliation(s)
- Russell P. Sawyer
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH, USA
- *Correspondence: Russell P. Sawyer, Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH, 45219, USA (e-mail: )
| | - Hillarey K. Stone
- Division of Nephrology and Hypertension, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Hanan Salim
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Xiaoming Lu
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Matthew T. Weirauch
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Leah Kottyan
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
35
|
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
|
36
|
Kaliszewska A, Allison J, Col TT, Shaw C, Arias N. Elucidating the Role of Cerebellar Synaptic Dysfunction in C9orf72-ALS/FTD - a Systematic Review and Meta-Analysis. CEREBELLUM (LONDON, ENGLAND) 2022; 21:681-714. [PMID: 34491551 PMCID: PMC9325807 DOI: 10.1007/s12311-021-01320-0] [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] [Subscribe] [Scholar Register] [Accepted: 08/16/2021] [Indexed: 12/28/2022]
Abstract
A hexanucleotide repeat expansion in the C9orf72 gene is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) with synaptic dysfunction identified as an early pathological hallmark. Although TDP-43 pathology and overt neurodegeneration are largely absent from the cerebellum, the pathological hallmarks of RNA foci and dipeptide repeat protein (DPR) inclusions are most abundant. Here, we present a systematic literature search in the databases of PubMed, Scopus, Embase, Web of Science and Science Direct up until March 5, 2021, which yielded 19,515 publications. Following the exclusion criteria, 72 articles were included having referred to C9orf72, synapses and the cerebellum. Meta-analyses were conducted on studies which reported experimental and control groups with means and standard deviations extracted from figures using the online tool PlotDigitizer. This revealed dendritic defects (P = 0.03), reduced C9orf72 in human patients (P = 0.005) and DPR-related neuronal loss (P = 0.0006) but no neuromuscular junction abnormalities (P = 0.29) or cerebellar neuronal loss (P = 0.23). Our results suggest that dendritic arborisation defects, synaptic gene dysregulation and altered synaptic neurotransmission may drive cerebellar synaptic dysfunction in C9-ALS/FTD. In this review, we discuss how the chronological appearance of the different pathological hallmarks alters synaptic integrity which may have profound implications for disease progression. We conclude that a reduction in C9orf72 protein levels combined with the accumulation of RNA foci and DPRs act synergistically to drive C9 synaptopathy in the cerebellum of C9-ALS/FTD patients.
Collapse
Affiliation(s)
- Aleksandra Kaliszewska
- UK Dementia Research Institute At King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic & Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, 5 Cutcombe road, Camberwell, SE59RX, London, UK
| | - Joseph Allison
- UK Dementia Research Institute At King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic & Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, 5 Cutcombe road, Camberwell, SE59RX, London, UK
| | - Tarik-Tarkan Col
- UK Dementia Research Institute At King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic & Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, 5 Cutcombe road, Camberwell, SE59RX, London, UK
| | - Christopher Shaw
- UK Dementia Research Institute At King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic & Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, 5 Cutcombe road, Camberwell, SE59RX, London, UK
- Centre for Brain Research, University of Auckland, 85 Grafton Road, Auckland, 1023, New Zealand
| | - Natalia Arias
- UK Dementia Research Institute At King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic & Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, 5 Cutcombe road, Camberwell, SE59RX, London, UK.
- INEUROPA, Instituto de Neurociencias del Principado de Asturias, Plaza Feijoo s/n, 33003, Oviedo, Spain.
| |
Collapse
|
37
|
Liu Y, Andreucci A, Iwamoto N, Yin Y, Yang H, Liu F, Bulychev A, Hu XS, Lin X, Lamore S, Patil S, Mohapatra S, Purcell-Estabrook E, Taborn K, Dale E, Vargeese C. Preclinical evaluation of WVE-004, aninvestigational stereopure oligonucleotide forthe treatment of C9orf72-associated ALS or FTD. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 28:558-570. [PMID: 35592494 PMCID: PMC9092894 DOI: 10.1016/j.omtn.2022.04.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 04/15/2022] [Indexed: 12/12/2022]
Abstract
A large hexanucleotide (G4C2) repeat expansion in the first intronic region of C9orf72 is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Several mechanisms have been proposed to explain how the repeat expansion drives disease, and we hypothesize that a variant-selective approach, in which transcripts affected by the repeat expansion are preferentially decreased, has the potential to address most of them. We report a stereopure antisense oligonucleotide, WVE-004, that executes this variant-selective mechanism of action. WVE-004 dose-dependently and selectively reduces repeat-containing transcripts in patient-derived motor neurons carrying a C9orf72-repeat expansion, as well as in the spinal cord and cortex of C9 BAC transgenic mice. In mice, selective transcript knockdown was accompanied by substantial decreases in dipeptide-repeat proteins, which are pathological biomarkers associated with the repeat expansion, and by preservation of healthy C9orf72 protein expression. These in vivo effects were durable, persisting for at least 6 months. These data support the advancement of WVE-004 as an investigational stereopure antisense oligonucleotide targeting C9orf72 for the treatment of C9orf72-associated ALS or FTD.
Collapse
Affiliation(s)
- Yuanjing Liu
- Wave Life Sciences, 733 Concord Avenue, Cambridge, MA 02138, USA
| | - Amy Andreucci
- Wave Life Sciences, 733 Concord Avenue, Cambridge, MA 02138, USA
| | - Naoki Iwamoto
- Wave Life Sciences, 733 Concord Avenue, Cambridge, MA 02138, USA
| | - Yuan Yin
- Wave Life Sciences, 733 Concord Avenue, Cambridge, MA 02138, USA
| | - Hailin Yang
- Wave Life Sciences, 733 Concord Avenue, Cambridge, MA 02138, USA
| | - Fangjun Liu
- Wave Life Sciences, 733 Concord Avenue, Cambridge, MA 02138, USA
| | - Alexey Bulychev
- Wave Life Sciences, 733 Concord Avenue, Cambridge, MA 02138, USA
| | - Xiao Shelley Hu
- Wave Life Sciences, 733 Concord Avenue, Cambridge, MA 02138, USA
| | - Xuena Lin
- Wave Life Sciences, 733 Concord Avenue, Cambridge, MA 02138, USA
| | - Sarah Lamore
- Wave Life Sciences, 733 Concord Avenue, Cambridge, MA 02138, USA
| | - Saurabh Patil
- Wave Life Sciences, 733 Concord Avenue, Cambridge, MA 02138, USA
| | | | | | - Kristin Taborn
- Wave Life Sciences, 733 Concord Avenue, Cambridge, MA 02138, USA
| | - Elena Dale
- Wave Life Sciences, 733 Concord Avenue, Cambridge, MA 02138, USA
| | - Chandra Vargeese
- Wave Life Sciences, 733 Concord Avenue, Cambridge, MA 02138, USA
| |
Collapse
|
38
|
Smukowski SN, Maioli H, Latimer CS, Bird TD, Jayadev S, Valdmanis PN. Progress in Amyotrophic Lateral Sclerosis Gene Discovery: Reflecting on Classic Approaches and Leveraging Emerging Technologies. Neurol Genet 2022; 8:e669. [PMID: 35620141 PMCID: PMC9128037 DOI: 10.1212/nxg.0000000000000669] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 03/14/2022] [Indexed: 11/22/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is the most prominent motor neuron disease in humans. Its etiology consists of progressive motor neuron degeneration resulting in a rapid decline in motor function starting in the limbs or bulbar muscles and eventually fatally impairing central organs most typically resulting in loss of respiration. Pathogenic variants in 4 main genes, SOD1, TARDBP, FUS, and C9orf72, have been well characterized as causative for more than a decade now. However, these only account for a small fraction of all ALS cases. In this review, we highlight many additional variants that appear to be causative or confer increased risk for ALS, and we reflect on the technologies that have led to these discoveries. Next, we call attention to new challenges and opportunities for ALS and suggest next steps to increase our understanding of ALS genetics. Finally, we conclude with a synopsis of gene therapy paradigms and how increased understanding of ALS genetics can lead us to developing effective treatments. Ultimately, a consolidated update of the field can provide a launching point for researchers and clinicians to improve our search for ALS-related genes, defining pathogenic mechanisms, form diagnostics, and develop therapies.
Collapse
Affiliation(s)
- Samuel N Smukowski
- Division of Medical Genetics (S.N.S., T.D.B., S.J., P.N.V.), University of Washington School of Medicine; Department of Laboratory Medicine and Pathology (H.M., C.S.L.), University of Washington; Northwest Mental Illness Research (C.M.), Education and Clinical Centers, VA Puget Sound Health Care System; Department of Neurology (T.D.B., S.J.), University of Washington; and Geriatric Research Education and Clinical Center (T.B.), VA Puget Sound Health Care System, Seattle, WA
| | - Heather Maioli
- Division of Medical Genetics (S.N.S., T.D.B., S.J., P.N.V.), University of Washington School of Medicine; Department of Laboratory Medicine and Pathology (H.M., C.S.L.), University of Washington; Northwest Mental Illness Research (C.M.), Education and Clinical Centers, VA Puget Sound Health Care System; Department of Neurology (T.D.B., S.J.), University of Washington; and Geriatric Research Education and Clinical Center (T.B.), VA Puget Sound Health Care System, Seattle, WA
| | - Caitlin S Latimer
- Division of Medical Genetics (S.N.S., T.D.B., S.J., P.N.V.), University of Washington School of Medicine; Department of Laboratory Medicine and Pathology (H.M., C.S.L.), University of Washington; Northwest Mental Illness Research (C.M.), Education and Clinical Centers, VA Puget Sound Health Care System; Department of Neurology (T.D.B., S.J.), University of Washington; and Geriatric Research Education and Clinical Center (T.B.), VA Puget Sound Health Care System, Seattle, WA
| | - Thomas D Bird
- Division of Medical Genetics (S.N.S., T.D.B., S.J., P.N.V.), University of Washington School of Medicine; Department of Laboratory Medicine and Pathology (H.M., C.S.L.), University of Washington; Northwest Mental Illness Research (C.M.), Education and Clinical Centers, VA Puget Sound Health Care System; Department of Neurology (T.D.B., S.J.), University of Washington; and Geriatric Research Education and Clinical Center (T.B.), VA Puget Sound Health Care System, Seattle, WA
| | - Suman Jayadev
- Division of Medical Genetics (S.N.S., T.D.B., S.J., P.N.V.), University of Washington School of Medicine; Department of Laboratory Medicine and Pathology (H.M., C.S.L.), University of Washington; Northwest Mental Illness Research (C.M.), Education and Clinical Centers, VA Puget Sound Health Care System; Department of Neurology (T.D.B., S.J.), University of Washington; and Geriatric Research Education and Clinical Center (T.B.), VA Puget Sound Health Care System, Seattle, WA
| | - Paul N Valdmanis
- Division of Medical Genetics (S.N.S., T.D.B., S.J., P.N.V.), University of Washington School of Medicine; Department of Laboratory Medicine and Pathology (H.M., C.S.L.), University of Washington; Northwest Mental Illness Research (C.M.), Education and Clinical Centers, VA Puget Sound Health Care System; Department of Neurology (T.D.B., S.J.), University of Washington; and Geriatric Research Education and Clinical Center (T.B.), VA Puget Sound Health Care System, Seattle, WA
| |
Collapse
|
39
|
How can we define the presymptomatic C9orf72 disease in 2022? An overview on the current definitions of preclinical and prodromal phases. Rev Neurol (Paris) 2022; 178:426-436. [PMID: 35525633 DOI: 10.1016/j.neurol.2022.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/08/2022] [Indexed: 11/24/2022]
Abstract
Repeat expansions in C9orf72 gene are the main genetic cause of frontotemporal dementia, amyotrophic lateral sclerosis and related phenotypes. With the advent of disease-modifying treatments, the presymptomatic disease phase is getting increasing interest as an ideal time window in which innovant therapeutic approaches could be administered. Recommendations issued from international study groups distinguish between a preclinical disease stage, during which lesions accumulate in absence of any symptoms or signs, and a prodromal stage, marked by the appearance the first subtle cognitive, behavioral, psychiatric and motor signs, before the full-blown disease. This paper summarizes the current definitions and criteria for these stages, in particular focusing on how fluid-based, neuroimaging and cognitive biomarkers can be useful to monitor disease trajectory across the presymptomatic phase, as well as to detect the earliest signs of clinical conversion. Continuous advances in the knowledge of C9orf72 pathophysiology, and the integration of biomarkers in the clinical evaluation of mutation carriers will allow a better diagnostic definition of C9orf72 disease spectrum from the earliest stages, with relevant impact on the possibility of disease prevention.
Collapse
|
40
|
Nelson A, Russell LL, Peakman G, Convery RS, Bouzigues A, Greaves CV, Bocchetta M, Cash DM, van Swieten JC, Jiskoot L, Moreno F, Sanchez-Valle R, Laforce R, Graff C, Masellis M, Tartaglia MC, Rowe JB, Borroni B, Finger E, Synofzik M, Galimberti D, Vandenberghe R, de Mendonça A, Butler CR, Gerhard A, Ducharme S, Le Ber I, Santana I, Pasquier F, Levin J, Otto M, Sorbi S, Rohrer JD, Almeida MR, Anderl‐Straub S, Andersson C, Antonell A, Archetti S, Arighi A, Balasa M, Barandiaran M, Bargalló N, Bartha R, Bender B, Benussi A, Bertoux M, Bertrand A, Bessi V, Black S, Bocchetta M, Borrego‐Ecija S, Bras J, Brice A, Bruffaerts R, Camuzat A, Cañada M, Cantoni V, Caroppo P, Cash D, Castelo‐Branco M, Colliot O, Cope T, Deramecourt V, Arriba M, Di Fede G, Díez A, Duro D, Fenoglio C, Ferrari C, Ferreira CB, Fox N, Freedman M, Fumagalli G, Funkiewiez A, Gabilondo A, Gasparotti R, Gauthier S, Gazzina S, Giaccone G, Gorostidi A, Greaves C, Guerreiro R, Heller C, Hoegen T, Indakoetxea B, Jelic V, Karnath H, Keren R, Kuchcinski G, Langheinrich T, Lebouvier T, Leitão MJ, Lladó A, Lombardi G, Loosli S, Maruta C, Mead S, Meeter L, Miltenberger G, Minkelen R, Mitchell S, Moore K, Nacmias B, Nelson A, Öijerstedt L, Olives J, Ourselin S, Padovani A, Panman J, Papma JM, Pijnenburg Y, Polito C, Premi E, Prioni S, Prix C, Rademakers R, Redaelli V, Rinaldi D, Rittman T, Rogaeva E, Rollin A, Rosa‐Neto P, Rossi G, Rossor M, Santiago B, Saracino D, Sayah S, Scarpini E, Schönecker S, Seelaar H, Semler E, Shafei R, Shoesmith C, Swift I, Tábuas‐Pereira M, Tainta M, Taipa R, Tang‐Wai D, Thomas DL, Thompson P, Thonberg H, Timberlake C, Tiraboschi P, Todd E, Van Damme P, Vandenbulcke M, Veldsman M, Verdelho A, Villanua J, Warren J, Wilke C, Wlasich E, Zetterberg H, Zulaica M. The CBI-R detects early behavioural impairment in genetic frontotemporal dementia. Ann Clin Transl Neurol 2022; 9:644-658. [PMID: 35950369 PMCID: PMC9082390 DOI: 10.1002/acn3.51544] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Behavioural dysfunction is a key feature of genetic frontotemporal dementia (FTD) but validated clinical scales measuring behaviour are lacking at present. METHODS We assessed behaviour using the revised version of the Cambridge Behavioural Inventory (CBI-R) in 733 participants from the Genetic FTD Initiative study: 466 mutation carriers (195 C9orf72, 76 MAPT, 195 GRN) and 267 non-mutation carriers (controls). All mutation carriers were stratified according to their global CDR plus NACC FTLD score into three groups: asymptomatic (CDR = 0), prodromal (CDR = 0.5) and symptomatic (CDR = 1+). Mixed-effects models adjusted for age, education, sex and family clustering were used to compare between the groups. Neuroanatomical correlates of the individual domains were assessed within each genetic group. RESULTS CBI-R total scores were significantly higher in all CDR 1+ mutation carrier groups compared with controls [C9orf72 mean 70.5 (standard deviation 27.8), GRN 56.2 (33.5), MAPT 62.1 (36.9)] as well as their respective CDR 0.5 groups [C9orf72 13.5 (14.4), GRN 13.3 (13.5), MAPT 9.4 (10.4)] and CDR 0 groups [C9orf72 6.0 (7.9), GRN 3.6 (6.0), MAPT 8.5 (13.3)]. The C9orf72 and GRN 0.5 groups scored significantly higher than the controls. The greatest impairment was seen in the Motivation domain for the C9orf72 and GRN symptomatic groups, whilst in the symptomatic MAPTgroup, the highest-scoring domains were Stereotypic and Motor Behaviours and Memory and Orientation. Neural correlates of each CBI-R domain largely overlapped across the different mutation carrier groups. CONCLUSIONS The CBI-R detects early behavioural change in genetic FTD, suggesting that it could be a useful measure within future clinical trials.
Collapse
Affiliation(s)
- Annabel Nelson
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK.,Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London, UK
| | - Lucy L Russell
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Georgia Peakman
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Rhian S Convery
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Arabella Bouzigues
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Caroline V Greaves
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Martina Bocchetta
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - David M Cash
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | | | - Lize Jiskoot
- Department of Neurology, Erasmus Medical Centre, Rotterdam, Netherlands
| | - Fermin Moreno
- Cognitive Disorders Unit, Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain.,Neuroscience Area, Biodonostia Health Research Institute, San Sebastian, Gipuzkoa, Spain
| | - Raquel Sanchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Barcelona, Spain
| | - Robert Laforce
- Clinique Interdisciplinaire de Mémoire, Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Québec, Canada
| | - Caroline Graff
- Center for Alzheimer Research, Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Bioclinicum, Karolinska Institutet, Solna, Sweden.,Unit for Hereditary Dementias, Theme Aging, Karolinska University Hospital, Solna, Sweden
| | - Mario Masellis
- Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, University of Toronto, Toronto, Canada
| | - Maria Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - James B Rowe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Barbara Borroni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Elizabeth Finger
- Department of Clinical Neurological Sciences, University of Western Ontario, London, Ontario, Canada
| | - Matthis Synofzik
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Tübingen, Germany.,Center for Neurodegenerative Diseases (DZNE, Tübingen, Germany
| | - Daniela Galimberti
- Fondazione Ca' Granda, IRCCS Ospedale Policlinico, Milan, Italy.,University of Milan, Centro Dino Ferrari, Milan, Italy
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium.,Neurology Service, University Hospitals Leuven, Leuven, Belgium.,Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | | | - Chris R Butler
- Nuffield Department of Clinical Neurosciences, Medical Sciences Division, University of Oxford, Oxford, UK.,Department of Brain Sciences, Imperial College London, London, UK
| | - Alexander Gerhard
- Division of Neuroscience and Experimental Psychology, Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK.,Departments of Geriatric Medicine and Nuclear Medicine, University of Duisburg-Essen, Duisburg, Germany
| | - Simon Ducharme
- Department of Psychiatry, McGill University Health Centre, McGill University, Montreal, QC, Canada.,McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Isabelle Le Ber
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau - ICM, Inserm U1127, CNRS UMR 7225, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France.,Département de Neurologie, Centre de référence des démences rares ou précoces, IM2A, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France.,Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France.,Reference Network for Rare Neurological Diseases (ERN-RND), European Union
| | - Isabel Santana
- University Hospital of Coimbra (HUC), Neurology Service, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Florence Pasquier
- Univ Lille, Lille, France.,Inserm 1172, Lille, France.,CHU, CNR-MAJ, Labex Distalz, LiCEND Lille, Lille, France
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians Universität München, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Sandro Sorbi
- Department of Neurofarba, University of Florence, Florence, Italy.,IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Pandya S, Maia PD, Freeze B, Menke RAL, Talbot K, Turner MR, Raj A. Modeling seeding and neuroanatomic spread of pathology in amyotrophic lateral sclerosis. Neuroimage 2022; 251:118968. [PMID: 35143975 PMCID: PMC10729776 DOI: 10.1016/j.neuroimage.2022.118968] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 01/26/2022] [Accepted: 02/02/2022] [Indexed: 12/12/2022] Open
Abstract
The neurodegenerative disorder amyotrophic lateral sclerosis (ALS) is characterized by the progressive loss of upper and lower motor neurons, with pathological involvement of cerebral motor and extra-motor areas in a clinicopathological spectrum with frontotemporal dementia (FTD). A key unresolved issue is how the non-random distribution of pathology in ALS reflects differential network vulnerability, including molecular factors such as regional gene expression, or preferential spread of pathology via anatomical connections. A system of histopathological staging of ALS based on the regional burden of TDP-43 pathology observed in postmortem brains has been supported to some extent by analysis of distribution of in vivo structural MRI changes. In this paper, computational modeling using a Network Diffusion Model (NDM) was used to investigate whether a process of focal pathological 'seeding' followed by structural network-based spread recapitulated postmortem histopathological staging and, secondly, whether this had any correlation to the pattern of expression of a panel of genes implicated in ALS across the healthy brain. Regionally parcellated T1-weighted MRI data from ALS patients (baseline n=79) was studied in relation to a healthy control structural connectome and a database of associated regional cerebral gene expression. The NDM provided strong support for a structural network-based basis for regional pathological spread in ALS, but no simple relationship to the spatial distribution of ALS-related genes in the healthy brain. Interestingly, OPTN gene was identified as a significant but a weaker non-NDM contributor within the network-gene interaction model (LASSO). Intriguingly, the critical seed regions for spread within the model were not within the primary motor cortex but basal ganglia, thalamus and insula, where NDM recapitulated aspects of the postmortem histopathological staging system. Within the ALS-FTD clinicopathological spectrum, non-primary motor structures may be among the earliest sites of cerebral pathology.
Collapse
Affiliation(s)
- Sneha Pandya
- Department of Radiology, Weill Cornell Medicine, 1300 York Avenue, New York, NY, United States.
| | - Pedro D Maia
- Department of Mathematics, University of Texas at Arlington, TX, United States
| | - Benjamin Freeze
- Scripps Health/MD Anderson Cancer Center, Department of Radiology, CA, United States
| | - Ricarda A L Menke
- Wellcome Centre for Integrative Neuroimaging, University of Oxford, West Wing Level 6, Oxford OX2 7PZ, United Kingdom
| | - Kevin Talbot
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Martin R Turner
- Wellcome Centre for Integrative Neuroimaging, University of Oxford, West Wing Level 6, Oxford OX2 7PZ, United Kingdom; Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom.
| | - Ashish Raj
- Department of Radiology, Weill Cornell Medicine, 1300 York Avenue, New York, NY, United States; Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94121, United States.
| |
Collapse
|
42
|
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
|
43
|
Benson BC, Shaw PJ, Azzouz M, Highley JR, Hautbergue GM. Proteinopathies as Hallmarks of Impaired Gene Expression, Proteostasis and Mitochondrial Function in Amyotrophic Lateral Sclerosis. Front Neurosci 2022; 15:783624. [PMID: 35002606 PMCID: PMC8733206 DOI: 10.3389/fnins.2021.783624] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 11/26/2021] [Indexed: 01/15/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal adult-onset neurodegenerative disease characterized by progressive degeneration of upper and lower motor neurons. As with the majority of neurodegenerative diseases, the pathological hallmarks of ALS involve proteinopathies which lead to the formation of various polyubiquitylated protein aggregates in neurons and glia. ALS is a highly heterogeneous disease, with both familial and sporadic forms arising from the convergence of multiple disease mechanisms, many of which remain elusive. There has been considerable research effort invested into exploring these disease mechanisms and in recent years dysregulation of RNA metabolism and mitochondrial function have emerged as of crucial importance to the onset and development of ALS proteinopathies. Widespread alterations of the RNA metabolism and post-translational processing of proteins lead to the disruption of multiple biological pathways. Abnormal mitochondrial structure, impaired ATP production, dysregulation of energy metabolism and calcium homeostasis as well as apoptosis have been implicated in the neurodegenerative process. Dysfunctional mitochondria further accumulate in ALS motor neurons and reflect a wider failure of cellular quality control systems, including mitophagy and other autophagic processes. Here, we review the evidence for RNA and mitochondrial dysfunction as some of the earliest critical pathophysiological events leading to the development of ALS proteinopathies, explore their relative pathological contributions and their points of convergence with other key disease mechanisms. This review will focus primarily on mutations in genes causing four major types of ALS (C9ORF72, SOD1, TARDBP/TDP-43, and FUS) and in protein homeostasis genes (SQSTM1, OPTN, VCP, and UBQLN2) as well as sporadic forms of the disease. Finally, we will look to the future of ALS research and how an improved understanding of central mechanisms underpinning proteinopathies might inform research directions and have implications for the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Bridget C Benson
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kingdom
| | - Pamela J Shaw
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kingdom.,Neuroscience Institute, University of Sheffield, Sheffield, United Kingdom
| | - Mimoun Azzouz
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kingdom.,Neuroscience Institute, University of Sheffield, Sheffield, United Kingdom.,Healthy Lifespan Institute (HELSI), University of Sheffield, Sheffield, United Kingdom
| | - J Robin Highley
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kingdom.,Neuroscience Institute, University of Sheffield, Sheffield, United Kingdom.,Healthy Lifespan Institute (HELSI), University of Sheffield, Sheffield, United Kingdom
| | - Guillaume M Hautbergue
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kingdom.,Neuroscience Institute, University of Sheffield, Sheffield, United Kingdom.,Healthy Lifespan Institute (HELSI), University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
44
|
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]
|
45
|
Gossink F, Dols A, Stek ML, Scheltens P, Nijmeijer B, Cohn Hokke P, Dijkstra A, Van Ruissen F, Aalfs C, Pijnenburg YAL. Early life involvement in C9orf72 repeat expansion carriers. J Neurol Neurosurg Psychiatry 2022; 93:93-100. [PMID: 33906932 DOI: 10.1136/jnnp-2020-325994] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 03/12/2021] [Accepted: 04/12/2021] [Indexed: 12/20/2022]
Abstract
OBJECTIVES The chromosome 9 open reading frame 72 gene (C9orf72) hexanucleotide repeat expansion (C9orf72RE) is the most common genetic cause of behavioural variant frontotemporal dementia (bvFTD). Since the onset of the C9orf72RE-associated disease is sometimes hard to define, we hypothesise that C9orf72RE may cause a lifelong neuropsychiatric vulnerability. The first aim of our study was to explore lifelong behavioural and personality characteristics in C9orf72RE. Second, we aimed to describe distinctive characteristics of C9orf72RE during disease course. METHODS Out of 183 patients from the Amsterdam Dementia Cohort that underwent genetic testing between 2011 and 2018, 20 C9orf72RE bvFTD patients and 23 C9orf72RE negative bvFTD patients were included. Patients and their relatives were interviewed extensively to chart their biography. Data analysis was performed through a mixed-methods approach including qualitative and quantitative analyses. RESULTS Education, type of professional career and number of intimate partners were not different between carriers and non-carriers. Carriers were more often described by their relatives as having 'fixed behavioural patterns in daily life' and with limited empathy already years before onset of bvFTD symptoms. In carriers, disease course was more often characterised by excessive buying and obsessive physical exercise than in non-carriers. CONCLUSION This is the first study thoroughly exploring biographies of bvFTD patients with C9orf72RE, revealing that subtle personality traits may be present early in life. Our study suggests that C9orf72RE exerts a lifelong neuropsychiatric vulnerability. This may strengthen hypotheses of links between neurodevelopmental and neurodegenerative diseases. Moreover, the presence of a distinct C9orf72RE -associated syndrome within the FTD spectrum opens doors for investigation of vulnerable neuronal networks.
Collapse
Affiliation(s)
- Flora Gossink
- Alzheimer Center, Department of Neurology, Location VU University Medical Center, Amsterdam University Medical Centres, Amsterdam, The Netherlands
| | - Annemiek Dols
- Department of Old Age psychiatry, GGZ inGeest Amsterdam locatie De Nieuwe Valerius, Amsterdam, The Netherlands
| | - Max L Stek
- Department of Old Age psychiatry, GGZ inGeest Amsterdam locatie De Nieuwe Valerius, Amsterdam, The Netherlands
| | - Philip Scheltens
- Alzheimer Center, Department of Neurology, Location VU University Medical Center, Amsterdam University Medical Centres, Amsterdam, The Netherlands
| | - Bas Nijmeijer
- Clinical Genetics, Amsterdam UMC Locatie VUmc, Amsterdam, The Netherlands
| | | | - Anke Dijkstra
- Amsterdam UMC Locatie VUmc, Amsterdam, The Netherlands
| | - Fred Van Ruissen
- Department of Clinical Genetics, Amsterdam UMC Location AMC, Amsterdam, The Netherlands
| | | | - Yolande A L Pijnenburg
- Alzheimer Center, Department of Neurology, Location VU University Medical Center, Amsterdam University Medical Centres, Amsterdam, The Netherlands
| |
Collapse
|
46
|
Genetic architecture of motor neuron diseases. J Neurol Sci 2021; 434:120099. [PMID: 34965490 DOI: 10.1016/j.jns.2021.120099] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/26/2021] [Accepted: 12/14/2021] [Indexed: 12/18/2022]
Abstract
Motor neuron diseases (MNDs) are rare and frequently fatal neurological disorders in which motor neurons within the brainstem and spinal cord regions slowly die. MNDs are primarily caused by genetic mutations, and > 100 different mutant genes in humans have been discovered thus far. Given the fact that many more MND-related genes have yet to be discovered, the growing body of genetic evidence has offered new insights into the diverse cellular and molecular mechanisms involved in the aetiology and pathogenesis of MNDs. This search may aid in the selection of potential candidate genes for future investigation and, eventually, may open the door to novel interventions to slow down disease progression. In this review paper, we have summarized detailed existing research findings of different MNDs, such as amyotrophic lateral sclerosis (ALS), spinal muscular atrophy (SMA), spinal bulbar muscle atrophy (SBMA) and hereditary spastic paraplegia (HSP) in relation to their complex genetic architecture.
Collapse
|
47
|
Geraudie A, Battista P, García AM, Allen IE, Miller ZA, Gorno-Tempini ML, Montembeault M. Speech and language impairments in behavioral variant frontotemporal dementia: A systematic review. Neurosci Biobehav Rev 2021; 131:1076-1095. [PMID: 34673112 DOI: 10.1016/j.neubiorev.2021.10.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 10/12/2021] [Accepted: 10/14/2021] [Indexed: 01/11/2023]
Abstract
Although behavioral variant frontotemporal dementia (bvFTD) is classically defined by behavioral and socio-emotional changes, impairments often extend to other cognitive functions. These include early speech and language deficits related to the disease's core neural disruptions. Yet, their scope and clinical relevance remains poorly understood. This systematic review characterizes such disturbances in bvFTD, considering clinically, neuroanatomically, genetically, and neuropathologically defined subgroups. We included 181 experimental studies, with at least 5 bvFTD patients diagnosed using accepted criteria, comparing speech and language outcomes between bvFTD patients and healthy controls or between bvFTD subgroups. Results reveal extensive and heterogeneous deficits across cohorts, with (a) consistent lexico-semantic, reading & writing, and prosodic impairments; (b) inconsistent deficits in motor speech and grammar; and (c) relative preservation of phonological skills. Also, preliminary findings suggest that the severity of speech and language deficits might be associated with global cognitive impairment, predominantly temporal or fronto-temporal atrophy and MAPT mutations (vs C9orf72). Although under-recognized, these impairments contribute to patient characterization and phenotyping, while potentially informing diagnosis and management.
Collapse
Affiliation(s)
- Amandine Geraudie
- Memory and Aging Center, Department of Neurology, University of California San Francisco, CA, USA; Neurology Department, Toulouse University Hospital, Toulouse, France
| | - Petronilla Battista
- Memory and Aging Center, Department of Neurology, University of California San Francisco, CA, USA; Global Brain Health Institute, University of California, San Francisco, USA; Istituti Clinici Scientifici Maugeri IRCCS, Institute of Bari, Via Generale Nicola Bellomo, Bari, Italy
| | - Adolfo M García
- Global Brain Health Institute, University of California, San Francisco, USA; Universidad De San Andrés, Buenos Aires, Argentina; National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina; Departamento de Lingüística y Literatura, Facultad de Humanidades, Universidad de Santiago de Chile, Santiago, Chile
| | - Isabel E Allen
- Global Brain Health Institute, University of California, San Francisco, USA; Department of Epidemiology & Biostatistics, University of California San Francisco, CA, USA
| | - Zachary A Miller
- Memory and Aging Center, Department of Neurology, University of California San Francisco, CA, USA
| | - Maria Luisa Gorno-Tempini
- Memory and Aging Center, Department of Neurology, University of California San Francisco, CA, USA; Global Brain Health Institute, University of California, San Francisco, USA
| | - Maxime Montembeault
- Memory and Aging Center, Department of Neurology, University of California San Francisco, CA, USA.
| |
Collapse
|
48
|
Querin G, Grazia Biferi M, Pradat PF. Biomarkers for C9orf7-ALS in Symptomatic and Pre-symptomatic Patients: State-of-the-art in the New Era of Clinical Trials. J Neuromuscul Dis 2021; 9:25-37. [PMID: 34864683 PMCID: PMC8842771 DOI: 10.3233/jnd-210754] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The development of new possible treatments for C9orf72-related ALS and the possibility of early identification of subjects genetically at risk of developing the disease is creating a critical need for biomarkers to track neurodegeneration that could be used as outcome measures in clinical trials. Current candidate biomarkers in C9orf72-ALS include neuropsychology tests, imaging, electrophysiology as well as different circulating biomarkers. Neuropsychology tests show early executive and verbal function involvement both in symptomatic and asymptomatic mutation carriers. At brain MRI, C9orf72-ALS patients present diffuse white and grey matter degeneration, which are already identified up to 20 years before symptom onset and that seem to be slowly progressive over time, while regions of altered connectivity at fMRI and of hypometabolism at [18F]FDG PET have been described as well. At the same time, spinal cord MRI has also shown progressive decrease of FA in the cortico-spinal tract over time. On the side of wet biomarkers, neurofilament proteins are increased both in the CSF and serum just before symptom onset and tend to slowly increase over time, while poly(GP) protein can be detected in the CSF and probably used as target engagement marker in clinical trials.
Collapse
Affiliation(s)
- Giorgia Querin
- Institut de Myologie, I-Motion Adult ClinicalTrials Platform, Hôpital Pitié-Salpêtrière, Paris, France.,APHP, Centre de référence desmaladies neuromusculaires Nord/Est/Ile de France, HôpitalPitié-Salpêtrière, Paris, France
| | - Maria Grazia Biferi
- Sorbonne Université, Inserm UMRS974, Centre of Research in Myology (CRM), Institut de Myologie, GH PitiéSalpêtrière, Paris, France
| | - Pierre-Francois Pradat
- APHP, Département de Neurologie, Centre Référent SLA, Hôpital Pitié-Salpêtrière, Paris, France.,Laboratoire d'Imagerie Biomédicale, CNRS, INSERM, Sorbonne Université, Paris, France.,Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute Ulster University, C-TRIC, Altnagelvin Hospital, Londonderry, United Kingdom
| |
Collapse
|
49
|
Ahmed RM, Bocchetta M, Todd EG, Tse NY, Devenney EM, Tu S, Caga J, Hodges JR, Halliday GM, Irish M, Kiernan MC, Piguet O, Rohrer JD. Tackling clinical heterogeneity across the amyotrophic lateral sclerosis-frontotemporal dementia spectrum using a transdiagnostic approach. Brain Commun 2021; 3:fcab257. [PMID: 34805999 PMCID: PMC8599039 DOI: 10.1093/braincomms/fcab257] [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] [Received: 01/11/2021] [Revised: 07/31/2021] [Accepted: 08/18/2021] [Indexed: 11/28/2022] Open
Abstract
The disease syndromes of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) display considerable clinical, genetic and pathological overlap, yet mounting evidence indicates substantial differences in progression and survival. To date, there has been limited examination of how profiles of brain atrophy might differ between clinical phenotypes. Here, we address this longstanding gap in the literature by assessing cortical and subcortical grey and white matter volumes on structural MRI in a large cohort of 209 participants. Cognitive and behavioural changes were assessed using the Addenbrooke’s Cognitive Examination and the Cambridge Behavioural Inventory. Relative to 58 controls, behavioural variant FTD (n = 58) and ALS–FTD (n = 41) patients displayed extensive atrophy of frontoinsular, cingulate, temporal and motor cortices, with marked subcortical atrophy targeting the hippocampus, amygdala, thalamus and striatum, with atrophy further extended to the brainstem, pons and cerebellum in the latter group. At the other end of the spectrum, pure-ALS patients (n = 52) displayed considerable frontoparietal atrophy, including right insular and motor cortices and pons and brainstem regions. Subcortical regions included the bilateral pallidum and putamen, but to a lesser degree than in the ALS–FTD and behavioural variant FTD groups. Across the spectrum the most affected region in all three groups was the insula, and specifically the anterior part (76–90% lower than controls). Direct comparison of the patient groups revealed disproportionate temporal atrophy and widespread subcortical involvement in ALS–FTD relative to pure-ALS. In contrast, pure-ALS displayed significantly greater parietal atrophy. Both behavioural variant FTD and ALS–FTD were characterized by volume decrease in the frontal lobes relative to pure-ALS. The motor cortex and insula emerged as differentiating structures between clinical syndromes, with bilateral motor cortex atrophy more pronounced in ALS–FTD compared with pure-ALS, and greater left motor cortex and insula atrophy relative to behavioural variant FTD. Taking a transdiagnostic approach, we found significant associations between abnormal behaviour and volume loss in a predominantly frontoinsular network involving the amygdala, striatum and thalamus. Our findings demonstrate the presence of distinct atrophy profiles across the ALS–FTD spectrum, with key structures including the motor cortex and insula. Notably, our results point to subcortical involvement in the origin of behavioural disturbances, potentially accounting for the marked phenotypic variability typically observed across the spectrum.
Collapse
Affiliation(s)
- Rebekah M Ahmed
- Memory and Cognition Clinic, Institute of Clinical Neurosciences, Royal Prince Alfred Hospital, Sydney 2050, Australia.,Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia
| | - Martina Bocchetta
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London WC1E, UK
| | - Emily G Todd
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London WC1E, UK
| | - Nga Yan Tse
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia
| | - Emma M Devenney
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia
| | - Sicong Tu
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia
| | - Jashelle Caga
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia
| | - John R Hodges
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia.,School of Psychology and Brain and Mind Centre, The University of Sydney, Sydney 2050, Australia
| | - Glenda M Halliday
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia
| | - Muireann Irish
- School of Psychology and Brain and Mind Centre, The University of Sydney, Sydney 2050, Australia
| | - Matthew C Kiernan
- Memory and Cognition Clinic, Institute of Clinical Neurosciences, Royal Prince Alfred Hospital, Sydney 2050, Australia.,Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia
| | - Olivier Piguet
- School of Psychology and Brain and Mind Centre, The University of Sydney, Sydney 2050, Australia
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London WC1E, UK
| |
Collapse
|
50
|
Saracino D, Géraudie A, Remes AM, Ferrieux S, Noguès-Lassiaille M, Bottani S, Cipriano L, Houot M, Funkiewiez A, Camuzat A, Rinaldi D, Teichmann M, Pariente J, Couratier P, Boutoleau-Bretonnière C, Auriacombe S, Etcharry-Bouyx F, Levy R, Migliaccio R, Solje E, Le Ber I. Primary progressive aphasias associated with C9orf72 expansions: Another side of the story. Cortex 2021; 145:145-159. [PMID: 34717271 DOI: 10.1016/j.cortex.2021.09.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 08/04/2021] [Accepted: 09/15/2021] [Indexed: 12/13/2022]
Abstract
C9orf72 repeat expansions are rarely associated with primary progressive aphasias (PPA). In-depth characterization of the linguistic deficits, and the underlying patterns of grey-matter atrophy in PPA associated with the C9orf72 expansions (PPA-C9orf72) are currently lacking. In this study, we comprehensively analyzed a unique series of 16 patients affected by PPA-C9orf72. Eleven patients were issued from two independent French and Finnish cohorts, and five were identified by means of literature review. Voxel-based morphometry (VBM) studies were performed on three of them. This study depicts the spectrum of C9orf72-related aphasic phenotypes, and illustrates their linguistic presentation. The non-fluent/agrammatic variant was the most frequent phenotype in our series (9/16 patients, 56%), with apraxia of speech being the main defining feature. Left frontal lobe atrophy was present in these subjects, peaking in inferior frontal gyrus. Three patients (19%) showed the semantic variant, with progression of atrophy in temporo-polar regions, later involving orbitofrontal cortex. Anterior temporal lobe dysfunction was also particularly relevant in two patients (12.5%) with mixed forms of PPA. Lastly, two patients (12.5%) had unclassifiable PPA with predominating word-finding difficulties. No PPA-C9orf72 patients in our series fulfilled the criteria of the logopenic variant. Importantly, this study underlines the role of C9orf72 mutation in the disruption of the most anterior parts of the language network, including prefrontal and temporo-polar areas. It provides guidelines for C9orf72 testing in PPA patients, with important clinical impact as gene-specific therapies are upcoming.
Collapse
Affiliation(s)
- Dario Saracino
- Sorbonne Université, Paris Brain Institute - Institut Du Cerveau - ICM, Inserm U1127, CNRS UMR 7225, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Reference Centre for Rare or Early-Onset Dementias, IM2A, Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Aramis Project Team, Inria Research Center of Paris, Paris, France
| | - Amandine Géraudie
- Department of Neurology, Toulouse University Hospital, Toulouse, France; ToNIC, Toulouse NeuroImaging Centre, Inserm, UPS, University of Toulouse, Toulouse, France
| | - Anne M Remes
- Research Unit of Clinical Neuroscience, Neurology, University of Oulu, Oulu, Finland; MRC Oulu, Oulu University Hospital, Oulu, Finland
| | - Sophie Ferrieux
- Reference Centre for Rare or Early-Onset Dementias, IM2A, Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France
| | - Marie Noguès-Lassiaille
- Reference Centre for Rare or Early-Onset Dementias, IM2A, Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France
| | - Simona Bottani
- Sorbonne Université, Paris Brain Institute - Institut Du Cerveau - ICM, Inserm U1127, CNRS UMR 7225, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Aramis Project Team, Inria Research Center of Paris, Paris, France
| | - Lorenzo Cipriano
- Sorbonne Université, Paris Brain Institute - Institut Du Cerveau - ICM, Inserm U1127, CNRS UMR 7225, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Department of Advanced Medical and Surgical Sciences, University of Campania "L. Vanvitelli" - Naples, Italy
| | - Marion Houot
- Sorbonne Université, Paris Brain Institute - Institut Du Cerveau - ICM, Inserm U1127, CNRS UMR 7225, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Reference Centre for Rare or Early-Onset Dementias, IM2A, Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Center of Excellence of Neurodegenerative Disease (CoEN), ICM, CIC Neurosciences, Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, Sorbonne Université, Paris, France
| | - Aurélie Funkiewiez
- Reference Centre for Rare or Early-Onset Dementias, IM2A, Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Paris Brain Institute - Institut Du Cerveau (ICM), FrontLab, Paris, France
| | - Agnès Camuzat
- Sorbonne Université, Paris Brain Institute - Institut Du Cerveau - ICM, Inserm U1127, CNRS UMR 7225, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; EPHE, PSL Research University, Paris, France
| | - Daisy Rinaldi
- Sorbonne Université, Paris Brain Institute - Institut Du Cerveau - ICM, Inserm U1127, CNRS UMR 7225, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Reference Centre for Rare or Early-Onset Dementias, IM2A, Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France
| | - Marc Teichmann
- Sorbonne Université, Paris Brain Institute - Institut Du Cerveau - ICM, Inserm U1127, CNRS UMR 7225, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Reference Centre for Rare or Early-Onset Dementias, IM2A, Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Paris Brain Institute - Institut Du Cerveau (ICM), FrontLab, Paris, France
| | - Jérémie Pariente
- Department of Neurology, Toulouse University Hospital, Toulouse, France; ToNIC, Toulouse NeuroImaging Centre, Inserm, UPS, University of Toulouse, Toulouse, France
| | | | | | - Sophie Auriacombe
- CMRR Nouvelle Aquitaine / Institut des Maladies Neurodégénératives Clinique (IMNc), CHU de Bordeaux Hôpital Pellegrin, Bordeaux, France
| | | | - Richard Levy
- Sorbonne Université, Paris Brain Institute - Institut Du Cerveau - ICM, Inserm U1127, CNRS UMR 7225, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Reference Centre for Rare or Early-Onset Dementias, IM2A, Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Paris Brain Institute - Institut Du Cerveau (ICM), FrontLab, Paris, France
| | - Raffaella Migliaccio
- Sorbonne Université, Paris Brain Institute - Institut Du Cerveau - ICM, Inserm U1127, CNRS UMR 7225, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Reference Centre for Rare or Early-Onset Dementias, IM2A, Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Paris Brain Institute - Institut Du Cerveau (ICM), FrontLab, Paris, France
| | - Eino Solje
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland; Neuro Center, Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Isabelle Le Ber
- Sorbonne Université, Paris Brain Institute - Institut Du Cerveau - ICM, Inserm U1127, CNRS UMR 7225, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Reference Centre for Rare or Early-Onset Dementias, IM2A, Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Paris Brain Institute - Institut Du Cerveau (ICM), FrontLab, Paris, France.
| | | |
Collapse
|