1
|
Forrest SL, Kovacs GG. Current concepts and molecular pathology of neurodegenerative diseases. Pathology 2025; 57:178-190. [PMID: 39672768 DOI: 10.1016/j.pathol.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 10/27/2024] [Indexed: 12/15/2024]
Abstract
Neurodegenerative diseases are a pathologically, clinically and genetically diverse group of diseases characterised by selective dysfunction, loss of synaptic connectivity and neurodegeneration, and are associated with the deposition of misfolded proteins in neurons and/or glia. Molecular studies have highlighted the role of conformationally altered proteins in the pathogenesis of neurodegenerative diseases and have paved the way for developing disease-specific biomarkers that capture and differentiate the main type/s of protein abnormality responsible for neurodegenerative diseases, some of which are currently used in clinical practice. These proteins follow sequential patterns of anatomical involvement and disease spread in the brain and may also be detected in peripheral organs. Recent studies suggest that glia are likely to have an important role in pathological spread throughout the brain and even follow distinct progression patterns from neurons. In addition to morphological and molecular approaches to the classification of these disorders, a further new stratification level incorporates the structure of protein filaments detected by cryogenic electron microscopy. Rather than occurring in isolation, combined deposition of tau, amyloid-β, α-synuclein and TDP-43 are frequently observed in neurodegenerative diseases and in the ageing brain. These can be overlooked, and their clinicopathological relevance is difficult to interpret. This review provides an overview of disease pathogenesis and diagnostic implications, recent molecular and ultrastructural classification of neurodegenerative diseases, how to approach ageing-related and mixed pathologies, and the importance of the protein-based classification system for practising neuropathologists and clinicians. This review also informs general pathologists about the relevance of ongoing full body autopsy studies to understand the spectrum and pathogenesis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Shelley L Forrest
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, ON, Canada; Laboratory Medicine Program and Krembil Brain Institute, University Health Network, Toronto, ON, Canada; Dementia Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Gabor G Kovacs
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, ON, Canada; Laboratory Medicine Program and Krembil Brain Institute, University Health Network, Toronto, ON, Canada; Dementia Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia; Edmond J. Safra Program in Parkinson's Disease, Rossy PSP Centre and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, ON, Canada; Department of Laboratory Medicine and Pathobiology and Department of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
2
|
Webster CP, Hall B, Crossley OM, Dauletalina D, King M, Lin YH, Castelli LM, Yang ZL, Coldicott I, Kyrgiou-Balli E, Higginbottom A, Ferraiuolo L, De Vos KJ, Hautbergue GM, Shaw PJ, West RJ, Azzouz M. RuvBL1/2 reduce toxic dipeptide repeat protein burden in multiple models of C9orf72-ALS/FTD. Life Sci Alliance 2025; 8:e202402757. [PMID: 39638345 PMCID: PMC11629685 DOI: 10.26508/lsa.202402757] [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: 04/05/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024] Open
Abstract
A G4C2 hexanucleotide repeat expansion in C9orf72 is the most common cause of amyotrophic lateral sclerosis and frontotemporal dementia (C9ALS/FTD). Bidirectional transcription and subsequent repeat-associated non-AUG (RAN) translation of sense and antisense transcripts leads to the formation of five dipeptide repeat (DPR) proteins. These DPRs are toxic in a wide range of cell and animal models. Therefore, decreasing RAN-DPRs may be of therapeutic benefit in the context of C9ALS/FTD. In this study, we found that C9ALS/FTD patients have reduced expression of the AAA+ family members RuvBL1 and RuvBL2, which have both been implicated in aggregate clearance. We report that overexpression of RuvBL1, but to a greater extent RuvBL2, reduced C9orf72-associated DPRs in a range of in vitro systems including cell lines, primary neurons from the C9-500 transgenic mouse model, and patient-derived iPSC motor neurons. In vivo, we further demonstrated that RuvBL2 overexpression and consequent DPR reduction in our Drosophila model was sufficient to rescue a number of DPR-related motor phenotypes. Thus, modulating RuvBL levels to reduce DPRs may be of therapeutic potential in C9ALS/FTD.
Collapse
Affiliation(s)
- Christopher P Webster
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, School of Medicine and Population Health, Faculty of Health, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Bradley Hall
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, School of Medicine and Population Health, Faculty of Health, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Olivia M Crossley
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, School of Medicine and Population Health, Faculty of Health, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Dana Dauletalina
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, School of Medicine and Population Health, Faculty of Health, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Marianne King
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, School of Medicine and Population Health, Faculty of Health, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Ya-Hui Lin
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, School of Medicine and Population Health, Faculty of Health, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Lydia M Castelli
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, School of Medicine and Population Health, Faculty of Health, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Zih-Liang Yang
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, School of Medicine and Population Health, Faculty of Health, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Ian Coldicott
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, School of Medicine and Population Health, Faculty of Health, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Ergita Kyrgiou-Balli
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, School of Medicine and Population Health, Faculty of Health, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Adrian Higginbottom
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, School of Medicine and Population Health, Faculty of Health, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Laura Ferraiuolo
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, School of Medicine and Population Health, Faculty of Health, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Kurt J De Vos
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, School of Medicine and Population Health, Faculty of Health, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Guillaume M Hautbergue
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, School of Medicine and Population Health, Faculty of Health, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
- Healthy Lifespan Institute (HELSI), University of Sheffield, Sheffield, UK
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, School of Medicine and Population Health, Faculty of Health, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Ryan Jh West
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, School of Medicine and Population Health, Faculty of Health, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Mimoun Azzouz
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, School of Medicine and Population Health, Faculty of Health, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
- Gene Therapy Innovation and Manufacturing Centre (GTIMC), Division of Neuroscience, University of Sheffield, Sheffield, UK
| |
Collapse
|
3
|
Luan T, Li Q, Huang Z, Feng Y, Xu D, Zhou Y, Hu Y, Wang T. Axonopathy Underlying Amyotrophic Lateral Sclerosis: Unraveling Complex Pathways and Therapeutic Insights. Neurosci Bull 2024; 40:1789-1810. [PMID: 39097850 PMCID: PMC11607281 DOI: 10.1007/s12264-024-01267-2] [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: 02/15/2024] [Accepted: 04/08/2024] [Indexed: 08/05/2024] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a complex neurodegenerative disorder characterized by progressive axonopathy, jointly leading to the dying back of the motor neuron, disrupting both nerve signaling and motor control. In this review, we highlight the roles of axonopathy in ALS progression, driven by the interplay of multiple factors including defective trafficking machinery, protein aggregation, and mitochondrial dysfunction. Dysfunctional intracellular transport, caused by disruptions in microtubules, molecular motors, and adaptors, has been identified as a key contributor to disease progression. Aberrant protein aggregation involving TDP-43, FUS, SOD1, and dipeptide repeat proteins further amplifies neuronal toxicity. Mitochondrial defects lead to ATP depletion, oxidative stress, and Ca2+ imbalance, which are regarded as key factors underlying the loss of neuromuscular junctions and axonopathy. Mitigating these defects through interventions including neurotrophic treatments offers therapeutic potential. Collaborative research efforts aim to unravel ALS complexities, opening avenues for holistic interventions that target diverse pathological mechanisms.
Collapse
Affiliation(s)
- Tongshu Luan
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Qing Li
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Zhi Huang
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Yu Feng
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Duo Xu
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Yujie Zhou
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Yiqing Hu
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Tong Wang
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| |
Collapse
|
4
|
van der Geest AT, Jakobs CE, Ljubikj T, Huffels CFM, Cañizares Luna M, Vieira de Sá R, Adolfs Y, de Wit M, Rutten DH, Kaal M, Zwartkruis MM, Carcolé M, Groen EJN, Hol EM, Basak O, Isaacs AM, Westeneng HJ, van den Berg LH, Veldink JH, Schlegel DK, Pasterkamp RJ. Molecular pathology, developmental changes and synaptic dysfunction in (pre-) symptomatic human C9ORF72-ALS/FTD cerebral organoids. Acta Neuropathol Commun 2024; 12:152. [PMID: 39289761 PMCID: PMC11409520 DOI: 10.1186/s40478-024-01857-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 08/24/2024] [Indexed: 09/19/2024] Open
Abstract
A hexanucleotide repeat expansion (HRE) in C9ORF72 is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Human brain imaging and experimental studies indicate early changes in brain structure and connectivity in C9-ALS/FTD, even before symptom onset. Because these early disease phenotypes remain incompletely understood, we generated iPSC-derived cerebral organoid models from C9-ALS/FTD patients, presymptomatic C9ORF72-HRE (C9-HRE) carriers, and controls. Our work revealed the presence of all three C9-HRE-related molecular pathologies and developmental stage-dependent size phenotypes in cerebral organoids from C9-ALS/FTD patients. In addition, single-cell RNA sequencing identified changes in cell type abundance and distribution in C9-ALS/FTD organoids, including a reduction in the number of deep layer cortical neurons and the distribution of neural progenitors. Further, molecular and cellular analyses and patch-clamp electrophysiology detected various changes in synapse structure and function. Intriguingly, organoids from all presymptomatic C9-HRE carriers displayed C9-HRE molecular pathology, whereas the extent to which more downstream cellular defects, as found in C9-ALS/FTD models, were detected varied for the different presymptomatic C9-HRE cases. Together, these results unveil early changes in 3D human brain tissue organization and synaptic connectivity in C9-ALS/FTD that likely constitute initial pathologies crucial for understanding disease onset and the design of therapeutic strategies.
Collapse
Affiliation(s)
- Astrid T van der Geest
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Channa E Jakobs
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Tijana Ljubikj
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Christiaan F M Huffels
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Marta Cañizares Luna
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Renata Vieira de Sá
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Youri Adolfs
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Marina de Wit
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Daan H Rutten
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Marthe Kaal
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Maria M Zwartkruis
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Genetics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Mireia Carcolé
- UK Dementia Research Institute at UCL and Dept. of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Ewout J N Groen
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Elly M Hol
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Onur Basak
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Adrian M Isaacs
- UK Dementia Research Institute at UCL and Dept. of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Henk-Jan Westeneng
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Leonard H van den Berg
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jan H Veldink
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Domino K Schlegel
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
5
|
Yokota O, Miki T, Nakashima-Yasuda H, Ishizu H, Haraguchi T, Ikeda C, Hasegawa M, Miyashita A, Ikeuchi T, Nishikawa N, Takenoshita S, Sudo K, Terada S, Takaki M. Pure argyrophilic grain disease revisited: independent effects on limbic, neocortical, and striato-pallido-nigral degeneration and the development of dementia in a series with a low to moderate Braak stage. Acta Neuropathol Commun 2024; 12:121. [PMID: 39085955 PMCID: PMC11290173 DOI: 10.1186/s40478-024-01828-6] [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: 06/15/2024] [Accepted: 06/18/2024] [Indexed: 08/02/2024] Open
Abstract
Agyrophilic grains (AGs) are age-related limbic-predominant lesions in which four-repeat tau is selectively accumulated. Because previous methodologically heterogeneous studies have demonstrated inconsistent findings on the relationship between AGs and dementia, whether AGs affect cognitive function remains unclear. To address this question, we first comprehensively evaluated the distribution and quantity of Gallyas-positive AGs and the severity of neuronal loss in the limbic, neocortical, and subcortical regions in 30 cases of pure argyrophilic grain disease (pAGD) in Braak stages I-IV and without other degenerative diseases, and 34 control cases that had only neurofibrillary tangles with Braak stages I-IV and no or minimal Aβ deposits. Then, we examined whether AGs have independent effects on neuronal loss and dementia by employing multivariate ordered logistic regression and binomial logistic regression. Of 30 pAGD cases, three were classified in diffuse form pAGD, which had evident neuronal loss not only in the limbic region but also in the neocortex and subcortical nuclei. In all 30 pAGD cases, neuronal loss developed first in the amygdala, followed by temporo-frontal cortex, hippocampal CA1, substantia nigra, and finally, the striatum and globus pallidus with the progression of Saito AG stage. In multivariate analyses of 30 pAGD and 34 control cases, the Saito AG stage affected neuronal loss in the amygdala, hippocampal CA1, temporo-frontal cortex, striatum, globus pallidus, and substantia nigra independent of the age, Braak stage, and limbic-predominant age-related TDP-43 encephalopathy (LATE-NC) stage. In multivariate analyses of 23 pAGD and 28 control cases that lacked two or more lacunae and/or one or more large infarctions, 100 or more AGs per × 400 visual field in the amygdala (OR 10.02, 95% CI 1.12-89.43) and hippocampal CA1 (OR 12.22, 95% CI 1.70-87.81), and the presence of AGs in the inferior temporal cortex (OR 8.18, 95% CI 1.03-65.13) affected dementia independent of age, moderate Braak stages (III-IV), and LATE-NC. Given these findings, the high density of limbic AGs and the increase of AGs in the inferior temporal gyrus may contribute to the occurrence of dementia through neuronal loss, at least in cases in a low to moderate Braak stage.
Collapse
Affiliation(s)
- Osamu Yokota
- Department of Neuropsychiatry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama, 700-8558, Japan.
- Okayama University Medical School, Okayama, Japan.
- Department of Psychiatry, Kinoko Espoir Hospital, Okayama, Japan.
| | - Tomoko Miki
- Department of Neuropsychiatry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama, 700-8558, Japan
- Department of Neuropathology, Pitié-Salpêtrière Hospital, AP-HP, Sorbonne University, Paris, France
- Institut du Cerveau - Paris Brain Institute - ICM, Inserm U1127, CNRS UMR7225, AP-HP, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
| | - Hanae Nakashima-Yasuda
- Okayama University Medical School, Okayama, Japan
- Department of Psychiatry, Zikei Hospital, Okayama, Japan
| | - Hideki Ishizu
- Okayama University Medical School, Okayama, Japan
- Department of Psychiatry, Zikei Hospital, Okayama, Japan
| | - Takashi Haraguchi
- Department of Neurology, National Hospital Organization Minami-Okayama Medical Center, Okayama, Japan
| | - Chikako Ikeda
- Okayama University Medical School, Okayama, Japan
- Department of Psychiatry, Zikei Hospital, Okayama, Japan
| | - Masato Hasegawa
- Dementia Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Akinori Miyashita
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Naoto Nishikawa
- Department of Neuropsychiatry, Okayama University Hospital, Okayama, Japan
| | | | - Koichiro Sudo
- Department of Psychiatry, Tosa Hospital, Kochi, Japan
| | - Seishi Terada
- Department of Neuropsychiatry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama, 700-8558, Japan
- Department of Neuropsychiatry, Okayama University Hospital, Okayama, Japan
- Department of Neuropsychiatry, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Manabu Takaki
- Department of Neuropsychiatry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama, 700-8558, Japan
- Department of Neuropsychiatry, Okayama University Hospital, Okayama, Japan
- Department of Neuropsychiatry, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
6
|
Tahedl M, Tan EL, Kleinerova J, Delaney S, Hengeveld JC, Doherty MA, Mclaughlin RL, Pradat PF, Raoul C, Ango F, Hardiman O, Chang KM, Lope J, Bede P. Progressive Cerebrocerebellar Uncoupling in Sporadic and Genetic Forms of Amyotrophic Lateral Sclerosis. Neurology 2024; 103:e209623. [PMID: 38900989 DOI: 10.1212/wnl.0000000000209623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Amyotrophic lateral sclerosis (ALS) is predominantly associated with motor cortex, corticospinal tract (CST), brainstem, and spinal cord degeneration, and cerebellar involvement is much less well characterized. However, some of the cardinal clinical features of ALS, such as dysarthria, dysphagia, gait impairment, falls, and impaired dexterity, are believed to be exacerbated by coexisting cerebellar pathology. Cerebellar pathology may also contribute to cognitive, behavioral, and pseudobulbar manifestations. Our objective was to systematically assess both intracerebellar pathology and cerebrocerebellar connectivity alterations in a genetically stratified cohort of ALS. METHODS A prospective, multimodal neuroimaging study was conducted to evaluate the longitudinal evolution of intracerebellar pathology and cerebrocerebellar connectivity, using structural and functional measures. RESULTS A total of 113 healthy controls and 212 genetically stratified individuals with ALS were included: (1) C9orf72 hexanucleotide carriers ("C9POS"), (2) sporadic patients who tested negative for ALS-associated genetic variants, and (3) intermediate-length CAG trinucleotide carriers in ATXN2 ("ATXN2"). Flocculonodular lobule (padj = 0.014, 95% CI -5.06e-5 to -3.98e-6) and crura (padj = 0.031, 95% CI -1.63e-3 to -5.55e-5) volume reductions were detected at baseline in sporadic patients. Cerebellofrontal and cerebelloparietal structural connectivity impairment was observed in both C9POS and sporadic patients at baseline, and both projections deteriorated further over time in sporadic patients (padj = 0.003, t(249) = 3.04 and padj = 0.05, t(249) = 1.93). Functional cerebelloparietal uncoupling was evident in sporadic patients at baseline (padj = 0.004, 95% CI -0.19 to -0.03). ATXN2 patients exhibited decreased cerebello-occipital functional connectivity at baseline (padj = 0.004, 95% CI -0.63 to -0.06), progressive cerebellotemporal functional disconnection (padj = 0.025, t(199) = -2.26), and progressive flocculonodular lobule degeneration (padj = 0.017, t(249) = -2.24). C9POS patients showed progressive ventral dentate atrophy (padj = 0.007, t(249) = -2.75). The CSTs (padj < 0.001, 95% CI 4.89e-5 to 1.14e-4) and transcallosal interhemispheric fibers (padj < 0.001, 95% CI 5.21e-5 to 1.31e-4) were affected at baseline in C9POS and exhibited rapid degeneration over the 4 time points. The rate of decline in CST and corpus callosum integrity was faster than the rate of cerebrocerebellar disconnection (padj = 0.001, t(190) = 6.93). DISCUSSION ALS is associated with accruing intracerebellar disease burden as well as progressive corticocerebellar uncoupling. Contrary to previous suggestions, we have not detected evidence of compensatory structural or functional changes in response to supratentorial degeneration. The contribution of cerebellar disease burden to dysarthria, dysphagia, gait impairment, pseudobulbar affect, and cognitive deficits should be carefully considered in clinical assessments, monitoring, and multidisciplinary interventions.
Collapse
Affiliation(s)
- Marlene Tahedl
- From the Computational Neuroimaging Group (CNG) (M.T., E.L.T., J.K., S.D., O.H., K.M.C., J.L., P.B.), School of Medicine, Trinity College Dublin; Department of Neurology (S.D., P.B.), St James's Hospital, Dublin; Smurfit Institute of Genetics (J.C.H., M.A.D., R.L.M.), Trinity College Dublin, Ireland; Department of Neurology (P.-F.P.), Pitié-Salpêtrière University Hospital, Paris; The Neuroscience Institute of Montpellier (INM) (C.R., F.A.), INSERM, CNRS; and ALS Centre (C.R.), University of Montpellier, CHU Montpellier, France
| | - Ee Ling Tan
- From the Computational Neuroimaging Group (CNG) (M.T., E.L.T., J.K., S.D., O.H., K.M.C., J.L., P.B.), School of Medicine, Trinity College Dublin; Department of Neurology (S.D., P.B.), St James's Hospital, Dublin; Smurfit Institute of Genetics (J.C.H., M.A.D., R.L.M.), Trinity College Dublin, Ireland; Department of Neurology (P.-F.P.), Pitié-Salpêtrière University Hospital, Paris; The Neuroscience Institute of Montpellier (INM) (C.R., F.A.), INSERM, CNRS; and ALS Centre (C.R.), University of Montpellier, CHU Montpellier, France
| | - Jana Kleinerova
- From the Computational Neuroimaging Group (CNG) (M.T., E.L.T., J.K., S.D., O.H., K.M.C., J.L., P.B.), School of Medicine, Trinity College Dublin; Department of Neurology (S.D., P.B.), St James's Hospital, Dublin; Smurfit Institute of Genetics (J.C.H., M.A.D., R.L.M.), Trinity College Dublin, Ireland; Department of Neurology (P.-F.P.), Pitié-Salpêtrière University Hospital, Paris; The Neuroscience Institute of Montpellier (INM) (C.R., F.A.), INSERM, CNRS; and ALS Centre (C.R.), University of Montpellier, CHU Montpellier, France
| | - Siobhan Delaney
- From the Computational Neuroimaging Group (CNG) (M.T., E.L.T., J.K., S.D., O.H., K.M.C., J.L., P.B.), School of Medicine, Trinity College Dublin; Department of Neurology (S.D., P.B.), St James's Hospital, Dublin; Smurfit Institute of Genetics (J.C.H., M.A.D., R.L.M.), Trinity College Dublin, Ireland; Department of Neurology (P.-F.P.), Pitié-Salpêtrière University Hospital, Paris; The Neuroscience Institute of Montpellier (INM) (C.R., F.A.), INSERM, CNRS; and ALS Centre (C.R.), University of Montpellier, CHU Montpellier, France
| | - Jennifer C Hengeveld
- From the Computational Neuroimaging Group (CNG) (M.T., E.L.T., J.K., S.D., O.H., K.M.C., J.L., P.B.), School of Medicine, Trinity College Dublin; Department of Neurology (S.D., P.B.), St James's Hospital, Dublin; Smurfit Institute of Genetics (J.C.H., M.A.D., R.L.M.), Trinity College Dublin, Ireland; Department of Neurology (P.-F.P.), Pitié-Salpêtrière University Hospital, Paris; The Neuroscience Institute of Montpellier (INM) (C.R., F.A.), INSERM, CNRS; and ALS Centre (C.R.), University of Montpellier, CHU Montpellier, France
| | - Mark A Doherty
- From the Computational Neuroimaging Group (CNG) (M.T., E.L.T., J.K., S.D., O.H., K.M.C., J.L., P.B.), School of Medicine, Trinity College Dublin; Department of Neurology (S.D., P.B.), St James's Hospital, Dublin; Smurfit Institute of Genetics (J.C.H., M.A.D., R.L.M.), Trinity College Dublin, Ireland; Department of Neurology (P.-F.P.), Pitié-Salpêtrière University Hospital, Paris; The Neuroscience Institute of Montpellier (INM) (C.R., F.A.), INSERM, CNRS; and ALS Centre (C.R.), University of Montpellier, CHU Montpellier, France
| | - Russell L Mclaughlin
- From the Computational Neuroimaging Group (CNG) (M.T., E.L.T., J.K., S.D., O.H., K.M.C., J.L., P.B.), School of Medicine, Trinity College Dublin; Department of Neurology (S.D., P.B.), St James's Hospital, Dublin; Smurfit Institute of Genetics (J.C.H., M.A.D., R.L.M.), Trinity College Dublin, Ireland; Department of Neurology (P.-F.P.), Pitié-Salpêtrière University Hospital, Paris; The Neuroscience Institute of Montpellier (INM) (C.R., F.A.), INSERM, CNRS; and ALS Centre (C.R.), University of Montpellier, CHU Montpellier, France
| | - Pierre-Francois Pradat
- From the Computational Neuroimaging Group (CNG) (M.T., E.L.T., J.K., S.D., O.H., K.M.C., J.L., P.B.), School of Medicine, Trinity College Dublin; Department of Neurology (S.D., P.B.), St James's Hospital, Dublin; Smurfit Institute of Genetics (J.C.H., M.A.D., R.L.M.), Trinity College Dublin, Ireland; Department of Neurology (P.-F.P.), Pitié-Salpêtrière University Hospital, Paris; The Neuroscience Institute of Montpellier (INM) (C.R., F.A.), INSERM, CNRS; and ALS Centre (C.R.), University of Montpellier, CHU Montpellier, France
| | - Cédric Raoul
- From the Computational Neuroimaging Group (CNG) (M.T., E.L.T., J.K., S.D., O.H., K.M.C., J.L., P.B.), School of Medicine, Trinity College Dublin; Department of Neurology (S.D., P.B.), St James's Hospital, Dublin; Smurfit Institute of Genetics (J.C.H., M.A.D., R.L.M.), Trinity College Dublin, Ireland; Department of Neurology (P.-F.P.), Pitié-Salpêtrière University Hospital, Paris; The Neuroscience Institute of Montpellier (INM) (C.R., F.A.), INSERM, CNRS; and ALS Centre (C.R.), University of Montpellier, CHU Montpellier, France
| | - Fabrice Ango
- From the Computational Neuroimaging Group (CNG) (M.T., E.L.T., J.K., S.D., O.H., K.M.C., J.L., P.B.), School of Medicine, Trinity College Dublin; Department of Neurology (S.D., P.B.), St James's Hospital, Dublin; Smurfit Institute of Genetics (J.C.H., M.A.D., R.L.M.), Trinity College Dublin, Ireland; Department of Neurology (P.-F.P.), Pitié-Salpêtrière University Hospital, Paris; The Neuroscience Institute of Montpellier (INM) (C.R., F.A.), INSERM, CNRS; and ALS Centre (C.R.), University of Montpellier, CHU Montpellier, France
| | - Orla Hardiman
- From the Computational Neuroimaging Group (CNG) (M.T., E.L.T., J.K., S.D., O.H., K.M.C., J.L., P.B.), School of Medicine, Trinity College Dublin; Department of Neurology (S.D., P.B.), St James's Hospital, Dublin; Smurfit Institute of Genetics (J.C.H., M.A.D., R.L.M.), Trinity College Dublin, Ireland; Department of Neurology (P.-F.P.), Pitié-Salpêtrière University Hospital, Paris; The Neuroscience Institute of Montpellier (INM) (C.R., F.A.), INSERM, CNRS; and ALS Centre (C.R.), University of Montpellier, CHU Montpellier, France
| | - Kai Ming Chang
- From the Computational Neuroimaging Group (CNG) (M.T., E.L.T., J.K., S.D., O.H., K.M.C., J.L., P.B.), School of Medicine, Trinity College Dublin; Department of Neurology (S.D., P.B.), St James's Hospital, Dublin; Smurfit Institute of Genetics (J.C.H., M.A.D., R.L.M.), Trinity College Dublin, Ireland; Department of Neurology (P.-F.P.), Pitié-Salpêtrière University Hospital, Paris; The Neuroscience Institute of Montpellier (INM) (C.R., F.A.), INSERM, CNRS; and ALS Centre (C.R.), University of Montpellier, CHU Montpellier, France
| | - Jasmin Lope
- From the Computational Neuroimaging Group (CNG) (M.T., E.L.T., J.K., S.D., O.H., K.M.C., J.L., P.B.), School of Medicine, Trinity College Dublin; Department of Neurology (S.D., P.B.), St James's Hospital, Dublin; Smurfit Institute of Genetics (J.C.H., M.A.D., R.L.M.), Trinity College Dublin, Ireland; Department of Neurology (P.-F.P.), Pitié-Salpêtrière University Hospital, Paris; The Neuroscience Institute of Montpellier (INM) (C.R., F.A.), INSERM, CNRS; and ALS Centre (C.R.), University of Montpellier, CHU Montpellier, France
| | - Peter Bede
- From the Computational Neuroimaging Group (CNG) (M.T., E.L.T., J.K., S.D., O.H., K.M.C., J.L., P.B.), School of Medicine, Trinity College Dublin; Department of Neurology (S.D., P.B.), St James's Hospital, Dublin; Smurfit Institute of Genetics (J.C.H., M.A.D., R.L.M.), Trinity College Dublin, Ireland; Department of Neurology (P.-F.P.), Pitié-Salpêtrière University Hospital, Paris; The Neuroscience Institute of Montpellier (INM) (C.R., F.A.), INSERM, CNRS; and ALS Centre (C.R.), University of Montpellier, CHU Montpellier, France
| |
Collapse
|
7
|
Christidi F, Kleinerova J, Tan EL, Delaney S, Tacheva A, Hengeveld JC, Doherty MA, McLaughlin RL, Hardiman O, Siah WF, Chang KM, Lope J, Bede P. Limbic Network and Papez Circuit Involvement in ALS: Imaging and Clinical Profiles in GGGGCC Hexanucleotide Carriers in C9orf72 and C9orf72-Negative Patients. BIOLOGY 2024; 13:504. [PMID: 39056697 PMCID: PMC11273537 DOI: 10.3390/biology13070504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 06/26/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024]
Abstract
Background: While frontotemporal involvement is increasingly recognized in Amyotrophic lateral sclerosis (ALS), the degeneration of limbic networks remains poorly characterized, despite growing evidence of amnestic deficits, impaired emotional processing and deficits in social cognition. Methods: A prospective neuroimaging study was conducted with 204 individuals with ALS and 111 healthy controls. Patients were stratified for hexanucleotide expansion status in C9orf72. A deep-learning-based segmentation approach was implemented to segment the nucleus accumbens, hypothalamus, fornix, mammillary body, basal forebrain and septal nuclei. The cortical, subcortical and white matter components of the Papez circuit were also systematically evaluated. Results: Hexanucleotide repeat expansion carriers exhibited bilateral amygdala, hypothalamus and nucleus accumbens atrophy, and C9orf72 negative patients showed bilateral basal forebrain volume reductions compared to controls. Both patient groups showed left rostral anterior cingulate atrophy, left entorhinal cortex thinning and cingulum and fornix alterations, irrespective of the genotype. Fornix, cingulum, posterior cingulate, nucleus accumbens, amygdala and hypothalamus degeneration was more marked in C9orf72-positive ALS patients. Conclusions: Our results highlighted that mesial temporal and parasagittal subcortical degeneration is not unique to C9orf72 carriers. Our radiological findings were consistent with neuropsychological observations and highlighted the importance of comprehensive neuropsychological testing in ALS, irrespective of the underlying genotype.
Collapse
Affiliation(s)
- Foteini Christidi
- Computational Neuroimaging Group (CNG), School of Medicine, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Jana Kleinerova
- Computational Neuroimaging Group (CNG), School of Medicine, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Ee Ling Tan
- Computational Neuroimaging Group (CNG), School of Medicine, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Siobhan Delaney
- Computational Neuroimaging Group (CNG), School of Medicine, Trinity College Dublin, D08 W9RT Dublin, Ireland
- Department of Neurology, St James’s Hospital, D08 KC95 Dublin, Ireland
| | - Asya Tacheva
- Computational Neuroimaging Group (CNG), School of Medicine, Trinity College Dublin, D08 W9RT Dublin, Ireland
- Department of Neurology, St James’s Hospital, D08 KC95 Dublin, Ireland
| | | | - Mark A. Doherty
- Smurfit Institute of Genetics, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | | | - Orla Hardiman
- Computational Neuroimaging Group (CNG), School of Medicine, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - We Fong Siah
- Computational Neuroimaging Group (CNG), School of Medicine, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Kai Ming Chang
- Computational Neuroimaging Group (CNG), School of Medicine, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Jasmin Lope
- Computational Neuroimaging Group (CNG), School of Medicine, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Peter Bede
- Computational Neuroimaging Group (CNG), School of Medicine, Trinity College Dublin, D08 W9RT Dublin, Ireland
- Department of Neurology, St James’s Hospital, D08 KC95 Dublin, Ireland
| |
Collapse
|
8
|
Carbayo Á, Borrego-Écija S, Turon-Sans J, Cortés-Vicente E, Molina-Porcel L, Gascón-Bayarri J, Rubio MÁ, Povedano M, Gámez J, Sotoca J, Juntas-Morales R, Almendrote M, Marquié M, Sánchez-Valle R, Illán-Gala I, Dols-Icardo O, Rubio-Guerra S, Bernal S, Caballero-Ávila M, Vesperinas A, Gelpi E, Rojas-García R. Clinicopathological correlates in the frontotemporal lobar degeneration-motor neuron disease spectrum. Brain 2024; 147:2357-2367. [PMID: 38227807 PMCID: PMC11224598 DOI: 10.1093/brain/awae011] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/05/2023] [Accepted: 01/02/2024] [Indexed: 01/18/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating motor neuron disease (MND) that shares a common clinical, genetic and pathologic spectrum with frontotemporal dementia (FTD). It is highly heterogeneous in its presentation and features. Up to 50% of patients with MND develop cognitive-behavioural symptoms during the course of the disease, meeting criteria for FTD in 10%-15% of cases. In the absence of a precise biomarker, neuropathology is still a valuable tool to understand disease nosology, reach a definite diagnostic confirmation and help define specific subgroups of patients with common phenotypic, genetic and biomarker profiles. However, few neuropathological series have been published, and the frequency of frontotemporal lobar degeneration (FTLD) in MND is difficult to estimate. In this work we describe a large clinicopathological series of MND patients, analysing the frequency of concurrent FTLD changes and trying to define specific subgroups of patients based on their clinical, genetic and pathological characteristics. We performed an observational, retrospective, multicentre case study. We included all cases meeting neuropathological criteria for MND from the Neurological Tissue Bank of the FRCB-IDIBAPS-Hospital Clínic Barcelona Biobank between 1994 and 2022, regardless of their last clinical diagnosis. While brain donation is encouraged in all patients, it is performed in very few, and representativeness of the cohort might not be precise for all patients with MND. We retrospectively reviewed clinical and neuropathological data and describe the main clinical, genetic and pathogenic features, comparing neuropathologic groups between MND with and without FTLD changes and aiming to define specific subgroups. We included brain samples from 124 patients, 44 of whom (35.5%) had FTLD neuropathologic features (i.e. FTLD-MND). Pathologic TDP-43 aggregates were present in 93.6% of the cohort and were more extensive (higher Brettschneider stage) in those with concurrent FTLD (P < 0.001). Motor symptom onset was more frequent in the bulbar region in FTLD-MND cases than in those with isolated MND (P = 0.023), with no differences in survival. We observed a better clinicopathological correlation in the MND group than in the FTLD-MND group (93.8% versus 61.4%; P < 0.001). Pathogenic genetic variants were more common in the FTLD-MND group, especially C9orf72. We describe a frequency of FTLD of 35.5% in our series of neuropathologically confirmed cases of MND. The FTLD-MND spectrum is highly heterogeneous in all aspects, especially in patients with FTLD, in whom it is particularly difficult to define specific subgroups. In the absence of definite biomarkers, neuropathology remains a valuable tool for a definite diagnosis, increasing our knowledge in disease nosology.
Collapse
Affiliation(s)
- Álvaro Carbayo
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute (IIB Sant Pau) Sant Pau, Barcelona 08025, Spain
- Department of Medicine, Universitat Autònoma de Barcelona (UAB), Barcelona 08025, Spain
- Biomedical Network Research Centre on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Sergi Borrego-Écija
- Alzheimer’s Disease and Other Cognitive Disorders Unit, Neurology Department, Hospital Clínic, Institut d’Investigacions Biomediques August Pi i Sunyer, University of Barcelona, Barcelona 08036, Spain
| | - Janina Turon-Sans
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute (IIB Sant Pau) Sant Pau, Barcelona 08025, Spain
- Department of Medicine, Universitat Autònoma de Barcelona (UAB), Barcelona 08025, Spain
- Biomedical Network Research Centre on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Elena Cortés-Vicente
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute (IIB Sant Pau) Sant Pau, Barcelona 08025, Spain
- Department of Medicine, Universitat Autònoma de Barcelona (UAB), Barcelona 08025, Spain
- Biomedical Network Research Centre on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Laura Molina-Porcel
- Alzheimer’s Disease and Other Cognitive Disorders Unit, Neurology Department, Hospital Clínic, Institut d’Investigacions Biomediques August Pi i Sunyer, University of Barcelona, Barcelona 08036, Spain
- Neurological Tissue Bank, Biobanc-Hospital Clínic-FRCB-IDIBAPS, Barcelona 08036, Spain
| | - Jordi Gascón-Bayarri
- Department of Neurology, Bellvitge University Hospital, L’Hospitalet de Llobregat, Barcelona 08907, Spain
| | - Miguel Ángel Rubio
- Neuromuscular Unit, Department of Neurology, Hospital del Mar, Barcelona 08003, Spain
| | - Mónica Povedano
- Department of Neurology, Motor Neuron Unit, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Bellvitge University Hospital, Hospitalet de Llobregat, Barcelona 08907, Spain
| | - Josep Gámez
- GMA Clinic, Neurology Department, European Reference Network On Rare Neuromuscular Diseases (ERN EURO-NMD), Barcelona 08029, Spain
| | - Javier Sotoca
- Neuromuscular Diseases Unit, Department of Neurology, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, 08035, Spain
| | - Raúl Juntas-Morales
- Neuromuscular Diseases Unit, Department of Neurology, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, 08035, Spain
| | - Miriam Almendrote
- Neurology Department, Hospital Germans Trias i Pujol, Badalona 08916, Spain
| | - Marta Marquié
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), Barcelona 08028, Spain
| | - Raquel Sánchez-Valle
- Alzheimer’s Disease and Other Cognitive Disorders Unit, Neurology Department, Hospital Clínic, Institut d’Investigacions Biomediques August Pi i Sunyer, University of Barcelona, Barcelona 08036, Spain
| | - Ignacio Illán-Gala
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Barcelona 08025, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid 28029, Spain
| | - Oriol Dols-Icardo
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Barcelona 08025, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid 28029, Spain
| | - Sara Rubio-Guerra
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Barcelona 08025, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid 28029, Spain
| | - Sara Bernal
- Biomedical Network Research Centre on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid 28029, Spain
- Genetics Department, Hospital de la Santa Creu i Sant Pau, Barcelona 08025, Spain
| | - Marta Caballero-Ávila
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute (IIB Sant Pau) Sant Pau, Barcelona 08025, Spain
- Department of Medicine, Universitat Autònoma de Barcelona (UAB), Barcelona 08025, Spain
- Biomedical Network Research Centre on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Ana Vesperinas
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute (IIB Sant Pau) Sant Pau, Barcelona 08025, Spain
- Department of Medicine, Universitat Autònoma de Barcelona (UAB), Barcelona 08025, Spain
- Biomedical Network Research Centre on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Ellen Gelpi
- Neurological Tissue Bank, Biobanc-Hospital Clínic-FRCB-IDIBAPS, Barcelona 08036, Spain
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna 1090, Austria
| | - Ricard Rojas-García
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute (IIB Sant Pau) Sant Pau, Barcelona 08025, Spain
- Department of Medicine, Universitat Autònoma de Barcelona (UAB), Barcelona 08025, Spain
- Biomedical Network Research Centre on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid 28029, Spain
| |
Collapse
|
9
|
Fontana E, Bongianni M, Benussi A, Bronzato E, Scialo C, Sacchetto L, Cagnin A, Castriciano S, Buratti E, Gardoni F, Italia M, Schreiber A, Ferracin C, Fiorini M, Newell KL, Cracco L, Garringer HJ, Cecchini MP, Polymenidou M, Padovani A, Monaco S, Legname G, Ghetti B, Borroni B, Zanusso G. Detection of TDP-43 seeding activity in the olfactory mucosa from patients with frontotemporal dementia. Alzheimers Dement 2024; 20:1156-1165. [PMID: 37908186 PMCID: PMC10917048 DOI: 10.1002/alz.13541] [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: 07/25/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 11/02/2023]
Abstract
INTRODUCTION We assessed TAR DNA-binding protein 43 (TDP-43) seeding activity and aggregates detection in olfactory mucosa of patients with frontotemporal lobar degeneration with TDP-43-immunoreactive pathology (FTLD-TDP) by TDP-43 seeding amplification assay (TDP43-SAA) and immunocytochemical analysis. METHODS The TDP43-SAA was optimized using frontal cortex samples from 16 post mortem cases with FTLD-TDP, FTLD with tau inclusions, and controls. Subsequently, olfactory mucosa samples were collected from 17 patients with FTLD-TDP, 15 healthy controls, and three patients carrying MAPT variants. RESULTS TDP43-SAA discriminated with 100% accuracy post mortem cases presenting or lacking TDP-43 neuropathology. TDP-43 seeding activity was detectable in the olfactory mucosa, and 82.4% of patients with FTLD-TDP tested positive, whereas 86.7% of controls tested negative (P < 0.001). Two out of three patients with MAPT mutations tested negative. In TDP43-SAA positive samples, cytoplasmatic deposits of phosphorylated TDP-43 in the olfactory neural cells were detected. DISCUSSION TDP-43 aggregates can be detectable in olfactory mucosa, suggesting that TDP43-SAA might be useful for identifying and monitoring FTLD-TDP in living patients.
Collapse
Affiliation(s)
- Elena Fontana
- Department of Neuroscience, Biomedicine and Movement SciencesUniversity of VeronaVeronaItaly
| | - Matilde Bongianni
- Department of Neuroscience, Biomedicine and Movement SciencesUniversity of VeronaVeronaItaly
| | - Alberto Benussi
- Department of Clinical and Experimental SciencesUniversity of BresciaBresciaItaly
- Department of Continuity of Care and FrailtyASST Spedali Civili Brescia HospitalBresciaItaly
| | - Erika Bronzato
- Department of Neuroscience, Biomedicine and Movement SciencesUniversity of VeronaVeronaItaly
| | - Carlo Scialo
- Department of Quantitative BiomedicineUniversity of ZurichZurichSwitzerland
| | - Luca Sacchetto
- Department of SurgeryDentistryPaediatrics and GynaecologyOtolaryngology SectionUniversity of VeronaVeronaItaly
| | - Annachiara Cagnin
- Neurology UnitDepartment of NeuroscienceUniversity of PadovaPaduaItaly
- Padova Neuroscience Center (PNC)University of PadovaPaduaItaly
| | | | - Emanuele Buratti
- International Centre for Genetic Engineering and Biotechnology (ICGEB)TriesteItaly
| | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences (DiSFeB) “Rodolfo Paoletti,”University of MilanMilanItaly
| | - Maria Italia
- Department of Pharmacological and Biomolecular Sciences (DiSFeB) “Rodolfo Paoletti,”University of MilanMilanItaly
| | - Alberto Schreiber
- Otorhinolaryngology Unit, Head and Neck SurgeryASST Spedali CiviliUniversity of BresciaBresciaItaly
| | - Chiara Ferracin
- Laboratory of Prion BiologyDepartment of NeuroscienceScuola Internazionale Superiore Di Studi Avanzati (SISSA)TriesteItaly
| | - Michele Fiorini
- Department of Neuroscience, Biomedicine and Movement SciencesUniversity of VeronaVeronaItaly
| | - Kathy L. Newell
- Department of Pathology and Laboratory MedicineSchool of MedicineIndiana UniversityIndianapolisIndianaUSA
| | - Laura Cracco
- Department of Pathology and Laboratory MedicineSchool of MedicineIndiana UniversityIndianapolisIndianaUSA
| | - Holly J. Garringer
- Department of Pathology and Laboratory MedicineSchool of MedicineIndiana UniversityIndianapolisIndianaUSA
| | - Maria Paola Cecchini
- Department of Neuroscience, Biomedicine and Movement SciencesUniversity of VeronaVeronaItaly
| | | | - Alessandro Padovani
- Department of Clinical and Experimental SciencesUniversity of BresciaBresciaItaly
- Department of Continuity of Care and FrailtyASST Spedali Civili Brescia HospitalBresciaItaly
| | - Salvatore Monaco
- Department of Neuroscience, Biomedicine and Movement SciencesUniversity of VeronaVeronaItaly
| | - Giuseppe Legname
- Laboratory of Prion BiologyDepartment of NeuroscienceScuola Internazionale Superiore Di Studi Avanzati (SISSA)TriesteItaly
| | - Bernardino Ghetti
- Department of Pathology and Laboratory MedicineSchool of MedicineIndiana UniversityIndianapolisIndianaUSA
| | - Barbara Borroni
- Department of Clinical and Experimental SciencesUniversity of BresciaBresciaItaly
- Department of Continuity of Care and FrailtyASST Spedali Civili Brescia HospitalBresciaItaly
| | - Gianluigi Zanusso
- Department of Neuroscience, Biomedicine and Movement SciencesUniversity of VeronaVeronaItaly
| |
Collapse
|
10
|
Rothstein JD, Baskerville V, Rapuri S, Mehlhop E, Jafar-Nejad P, Rigo F, Bennett F, Mizielinska S, Isaacs A, Coyne AN. G 2C 4 targeting antisense oligonucleotides potently mitigate TDP-43 dysfunction in human C9orf72 ALS/FTD induced pluripotent stem cell derived neurons. Acta Neuropathol 2023; 147:1. [PMID: 38019311 PMCID: PMC10840905 DOI: 10.1007/s00401-023-02652-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/09/2023] [Accepted: 10/26/2023] [Indexed: 11/30/2023]
Abstract
The G4C2 repeat expansion in the C9orf72 gene is the most common genetic cause of Amyotrophic Lateral Sclerosis and Frontotemporal Dementia. Many studies suggest that dipeptide repeat proteins produced from this repeat are toxic, yet, the contribution of repeat RNA toxicity is under investigated and even less is known regarding the pathogenicity of antisense repeat RNA. Recently, two clinical trials targeting G4C2 (sense) repeat RNA via antisense oligonucleotide failed despite a robust decrease in sense-encoded dipeptide repeat proteins demonstrating target engagement. Here, in this brief report, we show that G2C4 antisense, but not G4C2 sense, repeat RNA is sufficient to induce TDP-43 dysfunction in induced pluripotent stem cell (iPSC) derived neurons (iPSNs). Unexpectedly, only G2C4, but not G4C2 sense strand targeting, ASOs mitigate deficits in TDP-43 function in authentic C9orf72 ALS/FTD patient iPSNs. Collectively, our data suggest that the G2C4 antisense repeat RNA may be an important therapeutic target and provide insights into a possible explanation for the recent G4C2 ASO clinical trial failure.
Collapse
Affiliation(s)
- Jeffrey D Rothstein
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Victoria Baskerville
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Sampath Rapuri
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Emma Mehlhop
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | | | - Frank Rigo
- Ionis Pharmaceuticals, Carlsbad, CA, 92010, USA
| | | | - Sarah Mizielinska
- UK Dementia Research Institute at King's College London, London, UK
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
| | - Adrian Isaacs
- UK Dementia Research Institute at UCL, London, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UCL Queen Square Motor Neuron Disease Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Alyssa N Coyne
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
11
|
Fu RH, Chen HJ, Hong SY. Glycine-Alanine Dipeptide Repeat Protein from C9-ALS Interacts with Sulfide Quinone Oxidoreductase (SQOR) to Induce the Activity of the NLRP3 Inflammasome in HMC3 Microglia: Irisflorentin Reverses This Interaction. Antioxidants (Basel) 2023; 12:1896. [PMID: 37891975 PMCID: PMC10604625 DOI: 10.3390/antiox12101896] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/07/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal rare disease of progressive degeneration of motor neurons. The most common genetic mutation in ALS is the hexanucleotide repeat expansion (HRE) located in the first intron of the C9orf72 gene (C9-ALS). HRE can produce dipeptide repeat proteins (DPRs) such as poly glycine-alanine (GA) in a repeat-associated non-ATG (RAN) translation. GA-DPR has been shown to be toxic to motor neurons in various biological models. However, its effects on microglia involved in C9-ALS have not been reported. Here, we show that GA-DPR (GA50) activates the NLR family pyrin domain containing 3 (NLRP3) inflammasome in a human HMC3 microglia model. MCC950 (specific inhibitor of the NLRP3) treatment can abrogate this activity. Next, using yeast two-hybrid screening, we identified sulfide quinone oxidoreductase (SQOR) as a GA50 interacting protein. SQOR knockdown in HMC3 cells can significantly induce the activity of the NLRP3 inflammasome by upregulating the level of intracellular reactive oxygen species and the cytoplasmic escape of mitochondrial DNA. Furthermore, we obtained irisflorentin as an effective blocker of the interaction between SQOR and GA50, thus inhibiting NLRP3 inflammasome activity in GA50-expressing HMC3 cells. These results imply the association of GA-DPR, SQOR, and NLRP3 inflammasomes in microglia and establish a treatment strategy for C9-ALS with irisflorentin.
Collapse
Affiliation(s)
- Ru-Huei Fu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- Translational Medicine Research Center, China Medical University Hospital, Taichung 40447, Taiwan
- Ph.D. Program for Aging, China Medical University, Taichung 40402, Taiwan
| | - Hui-Jye Chen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
| | - Syuan-Yu Hong
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- Department of Medicine, School of Medicine, China Medical University, Taichung 40447, Taiwan
- Division of Pediatric Neurology, China Medical University Children’s Hospital, Taichung 40447, Taiwan
| |
Collapse
|
12
|
Zhang S, Tong M, Zheng D, Huang H, Li L, Ungermann C, Pan Y, Luo H, Lei M, Tang Z, Fu W, Chen S, Liu X, Zhong Q. C9orf72-catalyzed GTP loading of Rab39A enables HOPS-mediated membrane tethering and fusion in mammalian autophagy. Nat Commun 2023; 14:6360. [PMID: 37821429 PMCID: PMC10567733 DOI: 10.1038/s41467-023-42003-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 09/25/2023] [Indexed: 10/13/2023] Open
Abstract
The multi-subunit homotypic fusion and vacuole protein sorting (HOPS) membrane-tethering complex is required for autophagosome-lysosome fusion in mammals, yet reconstituting the mammalian HOPS complex remains a challenge. Here we propose a "hook-up" model for mammalian HOPS complex assembly, which requires two HOPS sub-complexes docking on membranes via membrane-associated Rabs. We identify Rab39A as a key small GTPase that recruits HOPS onto autophagic vesicles. Proper pairing with Rab2 and Rab39A enables HOPS complex assembly between proteoliposomes for its tethering function, facilitating efficient membrane fusion. GTP loading of Rab39A is important for the recruitment of HOPS to autophagic membranes. Activation of Rab39A is catalyzed by C9orf72, a guanine exchange factor associated with amyotrophic lateral sclerosis and familial frontotemporal dementia. Constitutive activation of Rab39A can rescue autophagy defects caused by C9orf72 depletion. These results therefore reveal a crucial role for the C9orf72-Rab39A-HOPS axis in autophagosome-lysosome fusion.
Collapse
Affiliation(s)
- Shen Zhang
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mindan Tong
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Denghao Zheng
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huiying Huang
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Linsen Li
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Christian Ungermann
- Osnabrück University, Department of Biology/Chemistry, Biochemistry section, Osnabrück, Germany
- Center of Cellular Nanoanalytic Osnabrück (CellNanOs), Osnabrück University, Osnabrück, Germany
| | - Yi Pan
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hanyan Luo
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming Lei
- State Key Laboratory of Oncogenes and Related Genes, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200011, Shanghai, China
- Shanghai Institute of Precision Medicine, 200125, Shanghai, China
| | - Zaiming Tang
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wan Fu
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - She Chen
- National Institute of Biological Sciences, 102206, Beijing, China
| | - Xiaoxia Liu
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Qing Zhong
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
13
|
Cristi AC, Rapuri S, Coyne AN. Nuclear pore complex and nucleocytoplasmic transport disruption in neurodegeneration. FEBS Lett 2023; 597:2546-2566. [PMID: 37657945 PMCID: PMC10612469 DOI: 10.1002/1873-3468.14729] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/29/2023] [Accepted: 08/24/2023] [Indexed: 09/03/2023]
Abstract
Nuclear pore complexes (NPCs) play a critical role in maintaining the equilibrium between the nucleus and cytoplasm, enabling bidirectional transport across the nuclear envelope, and are essential for proper nuclear organization and gene regulation. Perturbations in the regulatory mechanisms governing NPCs and nuclear envelope homeostasis have been implicated in the pathogenesis of several neurodegenerative diseases. The ESCRT-III pathway emerges as a critical player in the surveillance and preservation of well-assembled, functional NPCs, as well as nuclear envelope sealing. Recent studies have provided insights into the involvement of nuclear ESCRT-III in the selective reduction of specific nucleoporins associated with neurodegenerative pathologies. Thus, maintaining quality control of the nuclear envelope and NPCs represents a pivotal element in the pathological cascade leading to neurodegenerative diseases. This review describes the constituents of the nuclear-cytoplasmic transport machinery, encompassing the nuclear envelope, NPC, and ESCRT proteins, and how their structural and functional alterations contribute to the development of neurodegenerative diseases.
Collapse
Affiliation(s)
- América Chandía Cristi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore MD 21205, USA
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore MD 21205, USA
| | - Sampath Rapuri
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore MD 21205, USA
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore MD 21205, USA
| | - Alyssa N Coyne
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore MD 21205, USA
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore MD 21205, USA
| |
Collapse
|
14
|
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
|
15
|
Beckers J, Tharkeshwar AK, Fumagalli L, Contardo M, Van Schoor E, Fazal R, Thal DR, Chandran S, Mancuso R, Van Den Bosch L, Van Damme P. A toxic gain-of-function mechanism in C9orf72 ALS impairs the autophagy-lysosome pathway in neurons. Acta Neuropathol Commun 2023; 11:151. [PMID: 37723585 PMCID: PMC10506245 DOI: 10.1186/s40478-023-01648-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/20/2023] Open
Abstract
BACKGROUND Motor neurons (MNs), which are primarily affected in amyotrophic lateral sclerosis (ALS), are a specialized type of neurons that are long and non-dividing. Given their unique structure, these cells heavily rely on transport of organelles along their axons and the process of autophagy to maintain their cellular homeostasis. It has been shown that disruption of the autophagy pathway is sufficient to cause progressive neurodegeneration and defects in autophagy have been associated with various subtypes of ALS, including those caused by hexanucleotide repeat expansions in the C9orf72 gene. A more comprehensive understanding of the dysfunctional cellular mechanisms will help rationalize the design of potent and selective therapies for C9orf72-ALS. METHODS In this study, we used induced pluripotent stem cell (iPSC)-derived MNs from C9orf72-ALS patients and isogenic control lines to identify the underlying mechanisms causing dysregulations of the autophagy-lysosome pathway. Additionally, to ascertain the potential impact of C9orf72 loss-of-function on autophagic defects, we characterized the observed phenotypes in a C9orf72 knockout iPSC line (C9-KO). RESULTS Despite the evident presence of dysfunctions in several aspects of the autophagy-lysosome pathway, such as disrupted lysosomal homeostasis, abnormal lysosome morphology, inhibition of autophagic flux, and accumulation of p62 in C9orf72-ALS MNs, we were surprised to find that C9orf72 loss-of-function had minimal influence on these phenotypes. Instead, we primarily observed impairment in endosome maturation as a result of C9orf72 loss-of-function. Additionally, our study shed light on the pathological mechanisms underlying C9orf72-ALS, as we detected an increased TBK1 phosphorylation at S172 in MNs derived from C9orf72 ALS patients. CONCLUSIONS Our data provides further insight into the involvement of defects in the autophagy-lysosome pathway in C9orf72-ALS and strongly indicate that those defects are mainly due to the toxic gain-of-function mechanisms underlying C9orf72-ALS.
Collapse
Affiliation(s)
- Jimmy Beckers
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Louvain - University of Leuven, Leuven, Belgium.
- Center for Brain and Disease Research, Laboratory of Neurobiology, VIB, Leuven, Belgium.
| | - Arun Kumar Tharkeshwar
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Louvain - University of Leuven, Leuven, Belgium
- Center for Brain and Disease Research, Laboratory of Neurobiology, VIB, Leuven, Belgium
- Department of Human Genetics, KU Leuven, Louvain, Belgium
| | - Laura Fumagalli
- Center for Molecular Neurology, Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Matilde Contardo
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Louvain - University of Leuven, Leuven, Belgium
- Center for Brain and Disease Research, Laboratory of Neurobiology, VIB, Leuven, Belgium
| | - Evelien Van Schoor
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Louvain - University of Leuven, Leuven, Belgium
- Center for Brain and Disease Research, Laboratory of Neurobiology, VIB, Leuven, Belgium
- Laboratory of Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), KU Louvain - University of Leuven, Leuven, Belgium
| | - Raheem Fazal
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Louvain - University of Leuven, Leuven, Belgium
- Center for Brain and Disease Research, Laboratory of Neurobiology, VIB, Leuven, Belgium
| | - Dietmar Rudolf Thal
- Laboratory of Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), KU Louvain - University of Leuven, Leuven, Belgium
- Department of Pathology, University Hospitals Leuven, Louvain, Belgium
| | - Siddharthan Chandran
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, EH16 4SB, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Renzo Mancuso
- Center for Molecular Neurology, Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Louvain - University of Leuven, Leuven, Belgium
- Center for Brain and Disease Research, Laboratory of Neurobiology, VIB, Leuven, Belgium
| | - Philip Van Damme
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Louvain - University of Leuven, Leuven, Belgium.
- Center for Brain and Disease Research, Laboratory of Neurobiology, VIB, Leuven, Belgium.
- Department of Neurology, University Hospitals Leuven, Louvain, Belgium.
| |
Collapse
|
16
|
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: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [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
|
17
|
Ma L, Liang C, Wang J, Chang Q, Wang Y, Zhang W, Du Y, Sadan J, Chen JF. Reversing lysosome-ribosome circuit dysregulation mitigates C9FTD/ALS neurodegeneration and behaviors. Hum Mol Genet 2023; 32:1252-1265. [PMID: 36322143 PMCID: PMC10077508 DOI: 10.1093/hmg/ddac271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 10/22/2022] [Accepted: 10/27/2022] [Indexed: 12/14/2022] Open
Abstract
G4C2 repeat expansion in C9orf72 causes the most common familial frontotemporal dementia and amyotrophic lateral sclerosis (C9FTD/ALS). The pathogenesis includes haploinsufficiency of C9orf72, which forms a protein complex with Smcr8, as well as G4C2 repeat-induced gain of function including toxic dipeptide repeats (DPRs). The key in vivo disease-driving mechanisms and how loss- and gain-of-function interplay remain poorly understood. Here, we identified dysregulation of a lysosome-ribosome biogenesis circuit as an early and key disease mechanism using a physiologically relevant mouse model with combined loss- and gain-of-function across the aging process. C9orf72 deficiency exacerbates FTD/ALS-like pathologies and behaviors in C9ORF72 bacterial artificial chromosome (C9-BAC) mice with G4C2 repeats under endogenous regulatory elements from patients. Single nucleus RNA sequencing (snRNA-seq) and bulk RNA-seq revealed that C9orf72 depletion disrupts lysosomes in neurons and leads to transcriptional dysregulation of ribosomal protein genes, which are likely due to the proteotoxic stress response and resemble ribosomopathy defects. Importantly, ectopic expression of C9orf72 or its partner Smcr8 in C9FTD/ALS mutant mice promotes lysosomal functions and restores ribosome biogenesis gene transcription, resulting in the mitigation of DPR accumulation, neurodegeneration as well as FTD/ALS-like motor and cognitive behaviors. Therefore, we conclude that loss- and gain-of-function crosstalk in C9FTD/ALS converges on neuronal dysregulation of a lysosome-ribosome biogenesis circuit leading to proteotoxicity, neurodegeneration and behavioral defects.
Collapse
Affiliation(s)
- Li Ma
- Center for Craniofacial Molecular Biology, University of Southern California (USC), Los Angeles, CA 90033, USA
| | - Chen Liang
- Center for Craniofacial Molecular Biology, University of Southern California (USC), Los Angeles, CA 90033, USA
| | - Jing Wang
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Qing Chang
- Center for Craniofacial Molecular Biology, University of Southern California (USC), Los Angeles, CA 90033, USA
| | - Yuan Wang
- Center for Craniofacial Molecular Biology, University of Southern California (USC), Los Angeles, CA 90033, USA
| | - Wei Zhang
- Center for Craniofacial Molecular Biology, University of Southern California (USC), Los Angeles, CA 90033, USA
| | - Yuanning Du
- Center for Craniofacial Molecular Biology, University of Southern California (USC), Los Angeles, CA 90033, USA
| | - Jotham Sadan
- Center for Craniofacial Molecular Biology, University of Southern California (USC), Los Angeles, CA 90033, USA
| | - Jian-Fu Chen
- Center for Craniofacial Molecular Biology, University of Southern California (USC), Los Angeles, CA 90033, USA
| |
Collapse
|
18
|
Nelson PT, Lee EB, Cykowski MD, Alafuzoff I, Arfanakis K, Attems J, Brayne C, Corrada MM, Dugger BN, Flanagan ME, Ghetti B, Grinberg LT, Grossman M, Grothe MJ, Halliday GM, Hasegawa M, Hokkanen SRK, Hunter S, Jellinger K, Kawas CH, Keene CD, Kouri N, Kovacs GG, Leverenz JB, Latimer CS, Mackenzie IR, Mao Q, McAleese KE, Merrick R, Montine TJ, Murray ME, Myllykangas L, Nag S, Neltner JH, Newell KL, Rissman RA, Saito Y, Sajjadi SA, Schwetye KE, Teich AF, Thal DR, Tomé SO, Troncoso JC, Wang SHJ, White CL, Wisniewski T, Yang HS, Schneider JA, Dickson DW, Neumann M. LATE-NC staging in routine neuropathologic diagnosis: an update. Acta Neuropathol 2023; 145:159-173. [PMID: 36512061 PMCID: PMC9849315 DOI: 10.1007/s00401-022-02524-2] [Citation(s) in RCA: 81] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 12/15/2022]
Abstract
An international consensus report in 2019 recommended a classification system for limbic-predominant age-related TDP-43 encephalopathy neuropathologic changes (LATE-NC). The suggested neuropathologic staging system and nomenclature have proven useful for autopsy practice and dementia research. However, some issues remain unresolved, such as cases with unusual features that do not fit with current diagnostic categories. The goal of this report is to update the neuropathologic criteria for the diagnosis and staging of LATE-NC, based primarily on published data. We provide practical suggestions about how to integrate available genetic information and comorbid pathologies [e.g., Alzheimer's disease neuropathologic changes (ADNC) and Lewy body disease]. We also describe recent research findings that have enabled more precise guidance on how to differentiate LATE-NC from other subtypes of TDP-43 pathology [e.g., frontotemporal lobar degeneration (FTLD) and amyotrophic lateral sclerosis (ALS)], and how to render diagnoses in unusual situations in which TDP-43 pathology does not follow the staging scheme proposed in 2019. Specific recommendations are also made on when not to apply this diagnostic term based on current knowledge. Neuroanatomical regions of interest in LATE-NC are described in detail and the implications for TDP-43 immunohistochemical results are specified more precisely. We also highlight questions that remain unresolved and areas needing additional study. In summary, the current work lays out a number of recommendations to improve the precision of LATE-NC staging based on published reports and diagnostic experience.
Collapse
Affiliation(s)
- Peter T Nelson
- University of Kentucky, Rm 575 Todd Building, Lexington, KY, USA.
| | - Edward B Lee
- University of Pennsylvania, Philadelphia, PA, USA
| | | | | | - Konstantinos Arfanakis
- Rush University Medical Center, Chicago, IL, USA
- Illinois Institute of Technology, Chicago, IL, USA
| | | | | | | | | | | | | | | | | | - Michel J Grothe
- Unidad de Trastornos del Movimiento, Servicio de Neurología Y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | | | - Masato Hasegawa
- Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | | | | | | | | | | | | | - Gabor G Kovacs
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Laboratory Medicine Program, University Health Network, Toronto, Canada
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | | | | | | | - Qinwen Mao
- University of Utah, Salt Lake City, UT, USA
| | | | | | | | | | - Liisa Myllykangas
- University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Sukriti Nag
- Rush University Medical Center, Chicago, IL, USA
| | - Janna H Neltner
- University of Kentucky, Rm 575 Todd Building, Lexington, KY, USA
| | | | | | - Yuko Saito
- Tokyo Metropolitan Geriatric Hospital & Institute of Gerontology, Tokyo, Japan
| | | | | | | | - Dietmar R Thal
- Laboratory for Neuropathology, Department of Imaging and Pathoogy, and Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Pathology, University Hospital Leuven, Leuven, Belgium
| | - Sandra O Tomé
- Laboratory for Neuropathology, Department of Imaging and Pathoogy, and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | | | | | - Charles L White
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Hyun-Sik Yang
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, BostonBoston, MAMA, USA
| | | | | | | |
Collapse
|
19
|
Viader F. La sclérose latérale amyotrophique : une maladie neurodégénérative emblématique. BULLETIN DE L'ACADÉMIE NATIONALE DE MÉDECINE 2023. [DOI: 10.1016/j.banm.2023.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
20
|
Lopez-Herdoiza MB, Bauché S, Wilmet B, Le Duigou C, Roussel D, Frah M, Béal J, Devely G, Boluda S, Frick P, Bouteiller D, Dussaud S, Guillabert P, Dalle C, Dumont M, Camuzat A, Saracino D, Barbier M, Bruneteau G, Ravassard P, Neumann M, Nicole S, Le Ber I, Brice A, Latouche M. C9ORF72 knockdown triggers FTD-like symptoms and cell pathology in mice. Front Cell Neurosci 2023; 17:1155929. [PMID: 37138765 PMCID: PMC10149765 DOI: 10.3389/fncel.2023.1155929] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/27/2023] [Indexed: 05/05/2023] Open
Abstract
The GGGGCC intronic repeat expansion within C9ORF72 is the most common genetic cause of ALS and FTD. This mutation results in toxic gain of function through accumulation of expanded RNA foci and aggregation of abnormally translated dipeptide repeat proteins, as well as loss of function due to impaired transcription of C9ORF72. A number of in vivo and in vitro models of gain and loss of function effects have suggested that both mechanisms synergize to cause the disease. However, the contribution of the loss of function mechanism remains poorly understood. We have generated C9ORF72 knockdown mice to mimic C9-FTD/ALS patients haploinsufficiency and investigate the role of this loss of function in the pathogenesis. We found that decreasing C9ORF72 leads to anomalies of the autophagy/lysosomal pathway, cytoplasmic accumulation of TDP-43 and decreased synaptic density in the cortex. Knockdown mice also developed FTD-like behavioral deficits and mild motor phenotypes at a later stage. These findings show that C9ORF72 partial loss of function contributes to the damaging events leading to C9-FTD/ALS.
Collapse
Affiliation(s)
| | - Stephanie Bauché
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Baptiste Wilmet
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Caroline Le Duigou
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Delphine Roussel
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Magali Frah
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Jonas Béal
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Gabin Devely
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Susana Boluda
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Petra Frick
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | | | - Sébastien Dussaud
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Pierre Guillabert
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Carine Dalle
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Magali Dumont
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Agnes Camuzat
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Dario Saracino
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Mathieu Barbier
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Gaelle Bruneteau
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | | | - Manuela Neumann
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Neuropathology, Tübingen University Hospital, Tübingen, Germany
| | - Sophie Nicole
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Isabelle Le Ber
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Alexis Brice
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Morwena Latouche
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
- EPHE, Neurogenetics Team, PSL Research University, Paris, France
- *Correspondence: Morwena Latouche,
| |
Collapse
|
21
|
Tsoi KKF, Jia P, Dowling NM, Titiner JR, Wagner M, Capuano AW, Donohue MC. Applications of artificial intelligence in dementia research. CAMBRIDGE PRISMS. PRECISION MEDICINE 2022; 1:e9. [PMID: 38550934 PMCID: PMC10953738 DOI: 10.1017/pcm.2022.10] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 10/24/2022] [Accepted: 11/08/2022] [Indexed: 11/06/2024]
Abstract
More than 50 million older people worldwide are suffering from dementia, and this number is estimated to increase to 150 million by 2050. Greater caregiver burdens and financial impacts on the healthcare system are expected as we wait for an effective treatment for dementia. Researchers are constantly exploring new therapies and screening approaches for the early detection of dementia. Artificial intelligence (AI) is widely applied in dementia research, including machine learning and deep learning methods for dementia diagnosis and progression detection. Computerized apps are also convenient tools for patients and caregivers to monitor cognitive function changes. Furthermore, social robots can potentially provide daily life support or guidance for the elderly who live alone. This review aims to provide an overview of AI applications in dementia research. We divided the applications into three categories according to different stages of cognitive impairment: (1) cognitive screening and training, (2) diagnosis and prognosis for dementia, and (3) dementia care and interventions. There are numerous studies on AI applications for dementia research. However, one challenge that remains is comparing the effectiveness of different AI methods in real clinical settings.
Collapse
Affiliation(s)
- Kelvin K. F. Tsoi
- JC School of Public Health and Primary Care, The Chinese University of Hong Kong, Sha Tin, Hong Kong
- Stanley Ho Big Data Decision Analytics Research Centre, The Chinese University of Hong Kong, Sha Tin, Hong Kong
| | - Pingping Jia
- JC School of Public Health and Primary Care, The Chinese University of Hong Kong, Sha Tin, Hong Kong
| | - N. Maritza Dowling
- Department of Acute and Chronic tableCare, School of Nursing, The George Washington University, Washington, DC, USA
- Department of Epidemiology and Biostatistics, Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
| | | | - Maude Wagner
- Department of Neurological Sciences, Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Ana W. Capuano
- Department of Neurological Sciences, Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Michael C. Donohue
- Alzheimer’s Therapeutic Research Institute (ATRI), University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
22
|
Wang Y, Duan X, Zhou X, Wang R, Zhang X, Cao Z, Wang X, Zhou Z, Sun Y, Peng D. ANXA11 mutations are associated with amyotrophic lateral sclerosis–frontotemporal dementia. Front Neurol 2022; 13:886887. [PMID: 36226077 PMCID: PMC9549789 DOI: 10.3389/fneur.2022.886887] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 08/05/2022] [Indexed: 11/25/2022] Open
Abstract
Background The Annexin A11 (ANXA11) gene has been newly identified as a causative gene of amyotrophic lateral sclerosis (ALS) with or without frontotemporal dementia (FTD). The current study aimed to investigate the ANXA11 mutations in a Chinese ALS–FTD or FTD cohort. Methods We included ten probands/patients with suspected ALS–FTD or FTD. Mutational analysis of ANXA11 was performed through Next Generation Sequencing (NGS) and Sanger sequencing. We collected and reviewed clinical presentation, neuropsychology test results, brain-imaging findings, and electrophysiological examination findings. Results In total, six probands presented with ALS–FTD, and four with behavior variant FTD (bv-FTD). We identified a non-synonymous heterozygous mutation (c.119A>G, p.D40G) of ANXA11 in proband 1, which is associated with ALS. However, this is the first report of the mutation causing ALS–FTD. Proband 1 started with abnormal behavior and progressed to classic upper motor nervous disease. Magnetic resonance imaging (MRI) showed significant bilateral temporal lobe atrophy and bilateral hyperintensities along the corticospinal tracts.18F-AV45-PET imaging showed negative amyloid deposits. Conclusion ANXA11-related diseases have high clinical and genetic heterogeneity. Our study confirmed the contribution of ANXA11 mutations to ALS–FTD. The ANXA11 mutations established a complex genotype–phenotype correlation in ALS–FTD. Our research further elucidated the genetic mechanism of ALS–FTD and contributed to setting the foundation of future targeted therapy.
Collapse
Affiliation(s)
- Yu Wang
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | - Xiaohui Duan
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | - Xiao Zhou
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | - Renbin Wang
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | - Xiangfei Zhang
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | | | | | - Zhi Zhou
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | - Yu Sun
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | - Dantao Peng
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
- *Correspondence: Dantao Peng
| |
Collapse
|
23
|
Bioenergetic and Autophagic Characterization of Skin Fibroblasts from C9orf72 Patients. Antioxidants (Basel) 2022; 11:antiox11061129. [PMID: 35740026 PMCID: PMC9219955 DOI: 10.3390/antiox11061129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/24/2022] Open
Abstract
The objective of this study is to describe the alterations occurring during the neurodegenerative process in skin fibroblast cultures from C9orf72 patients. We characterized the oxidative stress, autophagy flux, small ubiquitin-related protein SUMO2/3 levels as well as the mitochondrial function in skin fibroblast cultures from C9orf72 patients. All metabolic and bioenergetic findings were further correlated with gene expression data obtained from RNA sequencing analysis. Fibroblasts from C9orf72 patients showed a 30% reduced expression of C9orf72, ~3-fold increased levels of oxidative stress and impaired mitochondrial function obtained by measuring the enzymatic activities of mitochondrial respiratory chain complexes, specifically of complex III activity. Furthermore, the results also reveal that C9orf72 patients showed an accumulation of p62 protein levels, suggesting the alteration of the autophagy process, and significantly higher protein levels of SUMO2/3 (p = 0.03). Our results provide new data reinforcing that C9orf72 cells suffer from elevated oxidative damage to biomolecules and organelles and from increased protein loads, leading to insufficient autophagy and an increase in SUMOylation processes.
Collapse
|
24
|
C9orf72 Hexanucleotide Repeat Expansion-Related Neuropathology Is Attenuated by Nasal Rifampicin in Mice. Biomedicines 2022; 10:biomedicines10051080. [PMID: 35625816 PMCID: PMC9138602 DOI: 10.3390/biomedicines10051080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/27/2022] [Accepted: 05/02/2022] [Indexed: 12/02/2022] Open
Abstract
The non-coding GGGGCC hexanucleotide repeat expansion (HRE) in C9orf72 gene is a dominant cause of frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS). This intronic mutation elicits the formation of nuclear and cytoplasmic inclusions containing RNA, RNA-binding proteins, and HRE-derived dipeptide repeat proteins (DPRs), leading to neurodegeneration via the gain-of-toxic function or loss-of-function of relevant proteins. Using C9-500 mice harboring ~500 repeats of the GGGGCC sequence in human C9orf72 gene, we investigated the effects of rifampicin against HRE-related pathological phenotypes. Rifampicin was administered intranasally to 4.5- to 5-month-old mice for 1 month, and their cognitive function and neuropathology were assessed by the Morris water maze test and immunohistochemical staining. Rifampicin treatment reduced the formation of RNA foci and cytoplasmic inclusions containing DPRs or phosphorylated TDP-43, and furthermore, the levels of phosphorylated double-strand RNA-dependent protein kinase (PKR) that regulates repeat-associated non-ATG (RAN) translation. Synapse loss in the hippocampus and neuronal loss and microglial activation in the prefrontal and motor cortices were also attenuated, and mouse memory was significantly improved. Our findings suggest a therapeutic potential of nasal rifampicin in the prevention of C9orf72-linked neurodegenerative disorders.
Collapse
|
25
|
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
|
26
|
Del Rosso G, Carlomagno Y, Todd TW, Jones CY, Prudencio M, Daughrity LM, Yue M, Jansen-West K, Tong J, Shao W, Wu Y, Castanedes-Casey M, Tabassian L, Oskarsson B, Ling K, Rigo F, Dickson DW, Yao TP, Petrucelli L, Cook CN, Zhang YJ. HDAC6 Interacts With Poly (GA) and Modulates its Accumulation in c9FTD/ALS. Front Cell Dev Biol 2022; 9:809942. [PMID: 35096836 PMCID: PMC8790530 DOI: 10.3389/fcell.2021.809942] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/16/2021] [Indexed: 11/13/2022] Open
Abstract
The aberrant translation of a repeat expansion in chromosome 9 open reading frame 72 (C9orf72), the most common cause of frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS), results in the accumulation of toxic dipeptide repeat (DPR) proteins in the central nervous system We have found that, among the sense DPR proteins, HDAC6 specifically interacts with the poly (GA) and co-localizes with inclusions in both patient tissue and a mouse model of this disease (c9FTD/ALS). Overexpression of HDAC6 increased poly (GA) levels in cultured cells independently of HDAC6 deacetylase activity, suggesting that HDAC6 can modulate poly (GA) pathology through a mechanism that depends upon their physical interaction. Moreover, decreasing HDAC6 expression by stereotaxic injection of antisense oligonucleotides significantly reduced the number of poly (GA) inclusions in c9FTD/ALS mice. These findings suggest that pharmacologically reducing HDAC6 levels could be of therapeutic value in c9FTD/ALS.
Collapse
Affiliation(s)
- Giulia Del Rosso
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States.,Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, United States
| | - Yari Carlomagno
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
| | - Tiffany W Todd
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
| | - Caroline Y Jones
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
| | - Mercedes Prudencio
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States.,Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, United States
| | | | - Mei Yue
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
| | - Karen Jansen-West
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
| | - Jimei Tong
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
| | - Wei Shao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
| | - Yanwei Wu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
| | | | - Lilia Tabassian
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
| | - Björn Oskarsson
- Department of Neurology, Mayo Clinic, Jacksonville, FL, United States
| | - Karen Ling
- Ionis Pharmaceuticals, Carlsbad, CA, United States
| | - Frank Rigo
- Ionis Pharmaceuticals, Carlsbad, CA, United States
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States.,Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, United States
| | - Tso-Pang Yao
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, United States
| | - Leonard Petrucelli
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States.,Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, United States
| | - Casey N Cook
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States.,Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, United States
| | - Yong Jie Zhang
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States.,Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, United States
| |
Collapse
|
27
|
Reyes-Leiva D, Dols-Icardo O, Sirisi S, Cortés-Vicente E, Turon-Sans J, de Luna N, Blesa R, Belbin O, Montal V, Alcolea D, Fortea J, Lleó A, Rojas-García R, Illán-Gala I. Pathophysiological Underpinnings of Extra-Motor Neurodegeneration in Amyotrophic Lateral Sclerosis: New Insights From Biomarker Studies. Front Neurol 2022; 12:750543. [PMID: 35115992 PMCID: PMC8804092 DOI: 10.3389/fneur.2021.750543] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 12/09/2021] [Indexed: 11/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) lie at opposing ends of a clinical, genetic, and neuropathological continuum. In the last decade, it has become clear that cognitive and behavioral changes in patients with ALS are more frequent than previously recognized. Significantly, these non-motor features can impact the diagnosis, prognosis, and management of ALS. Partially overlapping neuropathological staging systems have been proposed to describe the distribution of TAR DNA-binding protein 43 (TDP-43) aggregates outside the corticospinal tract. However, the relationship between TDP-43 inclusions and neurodegeneration is not absolute and other pathophysiological processes, such as neuroinflammation (with a prominent role of microglia), cortical hyperexcitability, and synaptic dysfunction also play a central role in ALS pathophysiology. In the last decade, imaging and biofluid biomarker studies have revealed important insights into the pathophysiological underpinnings of extra-motor neurodegeneration in the ALS-FTLD continuum. In this review, we first summarize the clinical and pathophysiological correlates of extra-motor neurodegeneration in ALS. Next, we discuss the diagnostic and prognostic value of biomarkers in ALS and their potential to characterize extra-motor neurodegeneration. Finally, we debate about how biomarkers could improve the diagnosis and classification of ALS. Emerging imaging biomarkers of extra-motor neurodegeneration that enable the monitoring of disease progression are particularly promising. In addition, a growing arsenal of biofluid biomarkers linked to neurodegeneration and neuroinflammation are improving the diagnostic accuracy and identification of patients with a faster progression rate. The development and validation of biomarkers that detect the pathological aggregates of TDP-43 in vivo are notably expected to further elucidate the pathophysiological underpinnings of extra-motor neurodegeneration in ALS. Novel biomarkers tracking the different aspects of ALS pathophysiology are paving the way to precision medicine approaches in the ALS-FTLD continuum. These are essential steps to improve the diagnosis and staging of ALS and the design of clinical trials testing novel disease-modifying treatments.
Collapse
Affiliation(s)
- David Reyes-Leiva
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Oriol Dols-Icardo
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Sonia Sirisi
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Elena Cortés-Vicente
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Janina Turon-Sans
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Noemi de Luna
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Rafael Blesa
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Olivia Belbin
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Victor Montal
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Daniel Alcolea
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Alberto Lleó
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Ricard Rojas-García
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Ignacio Illán-Gala
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
- *Correspondence: Ignacio Illán-Gala
| |
Collapse
|
28
|
Alvarez-Mora MI, Podlesniy P, Riazuelo T, Molina-Porcel L, Gelpi E, Rodriguez-Revenga L. Reduced mtDNA Copy Number in the Prefrontal Cortex of C9ORF72 Patients. Mol Neurobiol 2022; 59:1230-1237. [PMID: 34978044 DOI: 10.1007/s12035-021-02673-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/25/2021] [Indexed: 11/29/2022]
Abstract
Hexanucleotide repeat expansion in C9ORF72 gene is the most common genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia (C9ALS/FTD). Loss of C9ORF72 protein function and a toxic gain-of-function directly by the RNA or RAN translation have been proposed as triggering pathological mechanisms, along with the accumulation of TDP-43 protein. In addition, mitochondrial defects have been described to be a major driver of disease initiation. Mitochondrial DNA copy number has been proposed as a useful biomarker of mitochondrial dysfunction. The aim of our study was to determine the presence of mtDNA copy number alterations in C9ALS/FTD patients. Therefore, we assessed mtDNA copy number in postmortem prefrontal cortex from 18 C9ORF72 brain donors and 9 controls using digital droplet PCR. A statistically significant decrease of 50% was obtained when comparing C9ORF72 samples and controls. This decrease was independent of age and sex. The reduction of mtDNA copy number was found to be higher in patients' samples presenting abundant TDP-43 protein inclusions. A growing number of studies demonstrated the influence of mtDNA copy number reduction on neurodegeneration. Our results provide new insights into the role of mitochondrial dysfunction in the pathogenesis of C9ALS/FTD.
Collapse
Affiliation(s)
- Maria Isabel Alvarez-Mora
- Biochemistry and Molecular Genetics Department, Hospital Clinic of Barcelona, C/Villarroel, 170, 08036, Barcelona, Spain.,CIBER of Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain.,Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Petar Podlesniy
- CIBER of Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Teresa Riazuelo
- Biochemistry and Molecular Genetics Department, Hospital Clinic of Barcelona, C/Villarroel, 170, 08036, Barcelona, Spain
| | - Laura Molina-Porcel
- Neurological Tissue Bank of the Biobank-Hospital Clinic-IDIBAPS, Barcelona, Spain.,Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic, Institut d'Investigacions Biomediques August Pi I Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Ellen Gelpi
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Laia Rodriguez-Revenga
- Biochemistry and Molecular Genetics Department, Hospital Clinic of Barcelona, C/Villarroel, 170, 08036, Barcelona, Spain. .,CIBER of Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain. .,Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain.
| |
Collapse
|
29
|
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: 14] [Impact Index Per Article: 3.5] [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
|
30
|
Bush JA, Aikawa H, Fuerst R, Li Y, Ursu A, Meyer SM, Benhamou RI, Chen JL, Khan T, Wagner-Griffin S, Van Meter MJ, Tong Y, Olafson H, McKee KK, Childs-Disney JL, Gendron TF, Zhang Y, Coyne AN, Wang ET, Yildirim I, Wang KW, Petrucelli L, Rothstein JD, Disney MD. Ribonuclease recruitment using a small molecule reduced c9ALS/FTD r(G 4C 2) repeat expansion in vitro and in vivo ALS models. Sci Transl Med 2021; 13:eabd5991. [PMID: 34705518 DOI: 10.1126/scitranslmed.abd5991] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Jessica A Bush
- Department of Chemistry, Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Haruo Aikawa
- Department of Chemistry, Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Rita Fuerst
- Department of Chemistry, Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Yue Li
- Department of Chemistry, Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Andrei Ursu
- Department of Chemistry, Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Samantha M Meyer
- Department of Chemistry, Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Raphael I Benhamou
- Department of Chemistry, Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Jonathan L Chen
- Department of Chemistry, Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Tanya Khan
- Department of Chemistry, Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Sarah Wagner-Griffin
- Department of Chemistry, Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Montina J Van Meter
- Department of Chemistry, Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Yuquan Tong
- Department of Chemistry, Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Hailey Olafson
- Center for NeuroGenetics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Kendra K McKee
- Center for NeuroGenetics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Jessica L Childs-Disney
- Department of Chemistry, Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Tania F Gendron
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Yongjie Zhang
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Alyssa N Coyne
- Robert Packard Center for ALS Research, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, USA
| | - Eric T Wang
- Center for NeuroGenetics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Ilyas Yildirim
- Department of Chemistry and Biochemistry, Florida Atlantic University, Jupiter, FL 33458, USA
| | - Kye Won Wang
- Department of Chemistry and Biochemistry, Florida Atlantic University, Jupiter, FL 33458, USA
| | - Leonard Petrucelli
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Jeffrey D Rothstein
- Robert Packard Center for ALS Research, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, USA
| | - Matthew D Disney
- Department of Chemistry, Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| |
Collapse
|
31
|
López-Cáceres A, Velasco-Rueda M, Garcia-Cifuentes E, Zarante I, Matallana D. Analysis of Heritability Across the Clinical Phenotypes of Frontotemporal Dementia and the Frequency of the C9ORF72 in a Colombian Population. Front Neurol 2021; 12:681595. [PMID: 34526954 PMCID: PMC8435669 DOI: 10.3389/fneur.2021.681595] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 06/30/2021] [Indexed: 12/04/2022] Open
Abstract
Frontotemporal dementia (FTD) is a highly heritable condition. Up to 40% of FTD is familial and an estimated 15% to 40% is due to single-gene mutations. It has been estimated that the G4C2 hexanucleotide repeat expansions in the C9ORF72 gene can explain up to 37.5% of the familial cases of FTD, especially in populations of Caucasian origin. The purpose of this paper is to evaluate hereditary risk across the clinical phenotypes of FTD and the frequency of the G4C2 expansion in a Colombian cohort diagnosed with FTD. Methods: A total of 132 FTD patients were diagnosed according to established criteria in the behavioral variant FTD, logopenic variant PPA, non-fluent agrammatic PPA, and semantic variant PPA. Hereditary risk across the clinical phenotypes was established in four categories that indicate the pathogenic relationship of the mutation: high, medium, low, and apparently sporadic, based on those proposed by Wood and collaborators. All subjects were also examined for C9ORF72 hexanucleotide expansion (defined as >30 repetitions). Results: There were no significant differences in the demographic characteristics of the patients between the clinical phenotypes of FTD. The higher rate phenotype was bvFTD (62.12%). In accordance with the risk classification, we found that 72 (54.4%) complied with the criteria for the sporadic cases; for the familial cases, 23 (17.4%) fulfilled the high-risk criteria, 23 (17.4%) fulfilled the low risk criteria, and 14 (10.6%) fulfilled the criteria to be classified as subject to medium risk. C9ORF72 expansion frequency was 0.76% (1/132). Conclusion: The FTD heritability presented in this research was very similar to the results reported in the literature. The C9ORF72 expansion frequency was low. Colombia is a triethnic country, with a high frequency of genetic Amerindian markers; this shows consistency with the present results of a low repetition frequency. This study provides an initial report of the frequency for the hexanucleotide repeat expansions in C9ORF72 in patients with FTD in a Colombian population and paves the way for further study of the possible genetic causes of FTD in Colombia.
Collapse
Affiliation(s)
- Andrea López-Cáceres
- School of Medicine, Instituto de Genética Humana, Pontificia Universidad Javeriana, Bogotá, Colombia
- Fundación Santa Fé de Bogotá, Bogotá, Colombia
| | - María Velasco-Rueda
- School of Medicine, Instituto de Genética Humana, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Elkin Garcia-Cifuentes
- School of Medicine, Departamento de Neurociencias, Unidad de neurología, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Ignacio Zarante
- School of Medicine, Instituto de Genética Humana, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Diana Matallana
- Fundación Santa Fé de Bogotá, Bogotá, Colombia
- School of Medicine, Instituto de Envejecimiento, Doctorado de Neurociencias, Psychiatry and Mental Health Department, Pontificia Universidad Javeriana, Bogotá, Colombia
- Centro de Memoria y Cognición Intellectus, Hospital Universitario San Ignacio, Bogotá, Colombia
| |
Collapse
|
32
|
Trudler D, Ghatak S, Lipton SA. Emerging hiPSC Models for Drug Discovery in Neurodegenerative Diseases. Int J Mol Sci 2021; 22:8196. [PMID: 34360966 PMCID: PMC8347370 DOI: 10.3390/ijms22158196] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 12/12/2022] Open
Abstract
Neurodegenerative diseases affect millions of people worldwide and are characterized by the chronic and progressive deterioration of neural function. Neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD), represent a huge social and economic burden due to increasing prevalence in our aging society, severity of symptoms, and lack of effective disease-modifying therapies. This lack of effective treatments is partly due to a lack of reliable models. Modeling neurodegenerative diseases is difficult because of poor access to human samples (restricted in general to postmortem tissue) and limited knowledge of disease mechanisms in a human context. Animal models play an instrumental role in understanding these diseases but fail to comprehensively represent the full extent of disease due to critical differences between humans and other mammals. The advent of human-induced pluripotent stem cell (hiPSC) technology presents an advantageous system that complements animal models of neurodegenerative diseases. Coupled with advances in gene-editing technologies, hiPSC-derived neural cells from patients and healthy donors now allow disease modeling using human samples that can be used for drug discovery.
Collapse
Affiliation(s)
- Dorit Trudler
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (D.T.); (S.G.)
| | - Swagata Ghatak
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (D.T.); (S.G.)
| | - Stuart A. Lipton
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (D.T.); (S.G.)
- Department of Neurosciences, University of California San Diego School of Medicine, La Jolla, CA 92093, USA
| |
Collapse
|
33
|
Molecular Pathology of ALS: What We Currently Know and What Important Information Is Still Missing. Diagnostics (Basel) 2021; 11:diagnostics11081365. [PMID: 34441299 PMCID: PMC8391180 DOI: 10.3390/diagnostics11081365] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/25/2021] [Accepted: 07/25/2021] [Indexed: 12/23/2022] Open
Abstract
Despite an early understanding of amyotrophic lateral sclerosis (ALS) as a disease affecting the motor system, including motoneurons in the motor cortex, brainstem, and spinal cord, today, many cases involving dementia and behavioral disorders are reported. Therefore, we currently divide ALS not only based on genetic predisposition into the most common sporadic variant (90% of cases) and the familial variant (10%), but also based on cognitive and/or behavioral symptoms, with five specific subgroups of clinical manifestation—ALS with cognitive impairment, ALS with behavioral impairment, ALS with combined cognitive and behavioral impairment, the fully developed behavioral variant of frontotemporal dementia in combination with ALS, and comorbid ALS and Alzheimer’s disease (AD). Generally, these cases are referred to as amyotrophic lateral sclerosis-frontotemporal spectrum disorder (ALS-FTSD). Clinical behaviors and the presence of the same pathognomonic deposits suggest that FTLD and ALS could be a continuum of one entity. This review was designed primarily to compare neuropathological findings in different types of ALS relative to their characteristic locations as well as the immunoreactivity of the inclusions, and thus, foster a better understanding of the immunoreactivity, distribution, and morphology of the pathological deposits in relation to genetic mutations, which can be useful in specifying the final diagnosis.
Collapse
|
34
|
Chua JP, De Calbiac H, Kabashi E, Barmada SJ. Autophagy and ALS: mechanistic insights and therapeutic implications. Autophagy 2021; 18:254-282. [PMID: 34057020 PMCID: PMC8942428 DOI: 10.1080/15548627.2021.1926656] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mechanisms of protein homeostasis are crucial for overseeing the clearance of misfolded and toxic proteins over the lifetime of an organism, thereby ensuring the health of neurons and other cells of the central nervous system. The highly conserved pathway of autophagy is particularly necessary for preventing and counteracting pathogenic insults that may lead to neurodegeneration. In line with this, mutations in genes that encode essential autophagy factors result in impaired autophagy and lead to neurodegenerative conditions such as amyotrophic lateral sclerosis (ALS). However, the mechanistic details underlying the neuroprotective role of autophagy, neuronal resistance to autophagy induction, and the neuron-specific effects of autophagy-impairing mutations remain incompletely defined. Further, the manner and extent to which non-cell autonomous effects of autophagy dysfunction contribute to ALS pathogenesis are not fully understood. Here, we review the current understanding of the interplay between autophagy and ALS pathogenesis by providing an overview of critical steps in the autophagy pathway, with special focus on pivotal factors impaired by ALS-causing mutations, their physiologic effects on autophagy in disease models, and the cell type-specific mechanisms regulating autophagy in non-neuronal cells which, when impaired, can contribute to neurodegeneration. This review thereby provides a framework not only to guide further investigations of neuronal autophagy but also to refine therapeutic strategies for ALS and related neurodegenerative diseases.Abbreviations: ALS: amyotrophic lateral sclerosis; Atg: autophagy-related; CHMP2B: charged multivesicular body protein 2B; DPR: dipeptide repeat; FTD: frontotemporal dementia; iPSC: induced pluripotent stem cell; LIR: LC3-interacting region; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MTOR: mechanistic target of rapamycin kinase; PINK1: PTEN induced kinase 1; RNP: ribonuclear protein; sALS: sporadic ALS; SPHK1: sphingosine kinase 1; TARDBP/TDP-43: TAR DNA binding protein; TBK1: TANK-binding kinase 1; TFEB: transcription factor EB; ULK: unc-51 like autophagy activating kinase; UPR: unfolded protein response; UPS: ubiquitin-proteasome system; VCP: valosin containing protein.
Collapse
Affiliation(s)
- Jason P Chua
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Hortense De Calbiac
- Recherche translationnelle sur les maladies neurologiques, Institut Imagine, UMR-1163 INSERM et Université Paris Descartes, Hôpital Universitaire Necker-Enfants Malades, Paris, France
| | - Edor Kabashi
- Recherche translationnelle sur les maladies neurologiques, Institut Imagine, UMR-1163 INSERM et Université Paris Descartes, Hôpital Universitaire Necker-Enfants Malades, Paris, France
| | - Sami J Barmada
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
35
|
Smeyers J, Banchi EG, Latouche M. C9ORF72: What It Is, What It Does, and Why It Matters. Front Cell Neurosci 2021; 15:661447. [PMID: 34025358 PMCID: PMC8131521 DOI: 10.3389/fncel.2021.661447] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 03/17/2021] [Indexed: 12/11/2022] Open
Abstract
When the non-coding repeat expansion in the C9ORF72 gene was discovered to be the most frequent cause of frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) in 2011, this gene and its derived protein, C9ORF72, were completely unknown. The mutation appeared to produce both haploinsufficiency and gain-of-function effects in the form of aggregating expanded RNAs and dipeptide repeat proteins (DPRs). An unprecedented effort was then unleashed to decipher the pathogenic mechanisms and the functions of C9ORF72 in order to design therapies. A decade later, while the toxicity of accumulating gain-of-function products has been established and therapeutic strategies are being developed to target it, the contribution of the loss of function starts to appear more clearly. This article reviews the current knowledge about the C9ORF72 protein, how it is affected by the repeat expansion in models and patients, and what could be the contribution of its haploinsufficiency to the disease in light of the most recent findings. We suggest that these elements should be taken into consideration to refine future therapeutic strategies, compensating for the decrease of C9ORF72 or at least preventing a further reduction.
Collapse
Affiliation(s)
- Julie Smeyers
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, DMU Neuroscience 6, Paris, France
- PSL Research university, EPHE, Neurogenetics team, Paris, France
| | - Elena-Gaia Banchi
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, DMU Neuroscience 6, Paris, France
| | - Morwena Latouche
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, DMU Neuroscience 6, Paris, France
- PSL Research university, EPHE, Neurogenetics team, Paris, France
| |
Collapse
|
36
|
Kmetzsch V, Anquetil V, Saracino D, Rinaldi D, Camuzat A, Gareau T, Jornea L, Forlani S, Couratier P, Wallon D, Pasquier F, Robil N, de la Grange P, Moszer I, Le Ber I, Colliot O, Becker E. Plasma microRNA signature in presymptomatic and symptomatic subjects with C9orf72-associated frontotemporal dementia and amyotrophic lateral sclerosis. J Neurol Neurosurg Psychiatry 2021; 92:485-493. [PMID: 33239440 PMCID: PMC8053348 DOI: 10.1136/jnnp-2020-324647] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/30/2020] [Accepted: 10/27/2020] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To identify potential biomarkers of preclinical and clinical progression in chromosome 9 open reading frame 72 gene (C9orf72)-associated disease by assessing the expression levels of plasma microRNAs (miRNAs) in C9orf72 patients and presymptomatic carriers. METHODS The PREV-DEMALS study is a prospective study including 22 C9orf72 patients, 45 presymptomatic C9orf72 mutation carriers and 43 controls. We assessed the expression levels of 2576 miRNAs, among which 589 were above noise level, in plasma samples of all participants using RNA sequencing. The expression levels of the differentially expressed miRNAs between patients, presymptomatic carriers and controls were further used to build logistic regression classifiers. RESULTS Four miRNAs were differentially expressed between patients and controls: miR-34a-5p and miR-345-5p were overexpressed, while miR-200c-3p and miR-10a-3p were underexpressed in patients. MiR-34a-5p was also overexpressed in presymptomatic carriers compared with healthy controls, suggesting that miR-34a-5p expression is deregulated in cases with C9orf72 mutation. Moreover, miR-345-5p was also overexpressed in patients compared with presymptomatic carriers, which supports the correlation of miR-345-5p expression with the progression of C9orf72-associated disease. Together, miR-200c-3p and miR-10a-3p underexpression might be associated with full-blown disease. Four presymptomatic subjects in transitional/prodromal stage, close to the disease conversion, exhibited a stronger similarity with the expression levels of patients. CONCLUSIONS We identified a signature of four miRNAs differentially expressed in plasma between clinical conditions that have potential to represent progression biomarkers for C9orf72-associated frontotemporal dementia and amyotrophic lateral sclerosis. This study suggests that dysregulation of miRNAs is dynamically altered throughout neurodegenerative diseases progression, and can be detectable even long before clinical onset. TRIAL REGISTRATION NUMBER NCT02590276.
Collapse
Affiliation(s)
- Virgilio Kmetzsch
- Inria, Aramis project-team, F-75013, Paris, France.,Sorbonne Université, Paris Brain Institute - Institut du Cerveau - ICM, Inserm U1127, CNRS UMR 7225, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France
| | - Vincent Anquetil
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau - ICM, Inserm U1127, CNRS UMR 7225, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France
| | - Dario Saracino
- Inria, Aramis project-team, F-75013, Paris, France.,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
| | - Daisy Rinaldi
- 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
| | - 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
| | - Thomas Gareau
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau - ICM, Inserm U1127, CNRS UMR 7225, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France
| | - Ludmila Jornea
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau - ICM, Inserm U1127, CNRS UMR 7225, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France
| | - Sylvie Forlani
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau - ICM, Inserm U1127, CNRS UMR 7225, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France
| | | | - David Wallon
- Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, Department of Neurology and CNR-MAJ, Normandy Center for Genomic and Personalized Medicine, Rouen, France
| | | | | | | | - Ivan Moszer
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau - ICM, Inserm U1127, CNRS UMR 7225, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France
| | - 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.,Paris Brain Institute - Institut du Cerveau - ICM, FrontLab, Paris, France
| | - Olivier Colliot
- Inria, Aramis project-team, F-75013, Paris, France.,Sorbonne Université, Paris Brain Institute - Institut du Cerveau - ICM, Inserm U1127, CNRS UMR 7225, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France
| | | | | |
Collapse
|
37
|
Solomon DA, Smikle R, Reid MJ, Mizielinska S. Altered Phase Separation and Cellular Impact in C9orf72-Linked ALS/FTD. Front Cell Neurosci 2021; 15:664151. [PMID: 33967699 PMCID: PMC8096919 DOI: 10.3389/fncel.2021.664151] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 03/19/2021] [Indexed: 12/21/2022] Open
Abstract
Since the discovery of the C9orf72 repeat expansion mutation as causative for chromosome 9-linked amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) in 2011, a multitude of cellular pathways have been implicated. However, evidence has also been accumulating for a key mechanism of cellular compartmentalization—phase separation. Liquid-liquid phase separation (LLPS) is fundamental for the formation of membraneless organelles including stress granules, the nucleolus, Cajal bodies, nuclear speckles and the central channel of the nuclear pore. Evidence has now accumulated showing that the formation and function of these membraneless organelles is impaired by both the toxic arginine rich dipeptide repeat proteins (DPRs), translated from the C9orf72 repeat RNA transcript, and the repeat RNA itself. Both the arginine rich DPRs and repeat RNA themselves undergo phase separation and disrupt the physiological phase separation of proteins involved in the formation of these liquid-like organelles. Hence abnormal phase separation may explain a number of pathological cellular phenomena associated with C9orf72-ALS/FTD. In this review article, we will discuss the principles of phase separation, phase separation of the DPRs and repeat RNA themselves and how they perturb LLPS associated with membraneless organelles and the functional consequences of this. We will then discuss how phase separation may impact the major pathological feature of C9orf72-ALS/FTD, TDP-43 proteinopathy, and how LLPS may be targeted therapeutically in disease.
Collapse
Affiliation(s)
- Daniel A Solomon
- UK Dementia Research Institute at King's College London, London, United Kingdom.,Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Rebekah Smikle
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Matthew J Reid
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Sarah Mizielinska
- UK Dementia Research Institute at King's College London, London, United Kingdom.,Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| |
Collapse
|
38
|
Barry RL, Babu S, Anteraper SA, Triantafyllou C, Keil B, Rowe OE, Rangaprakash D, Paganoni S, Lawson R, Dheel C, Cernasov PM, Rosen BR, Ratai EM, Atassi N. Ultra-high field (7T) functional magnetic resonance imaging in amyotrophic lateral sclerosis: a pilot study. NEUROIMAGE-CLINICAL 2021; 30:102648. [PMID: 33872993 PMCID: PMC8060594 DOI: 10.1016/j.nicl.2021.102648] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 03/17/2021] [Accepted: 03/23/2021] [Indexed: 12/24/2022]
Abstract
Participants with ALS exhibited impaired function between the cortex and cerebellum. The cerebellum is associated with complex motor and cognitive processing tasks. These findings add to the growing number of ALS reports implicating the cerebellum.
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease of the central nervous system that results in a progressive loss of motor function and ultimately death. It is critical, yet also challenging, to develop non-invasive biomarkers to identify, localize, measure and/or track biological mechanisms implicated in ALS. Such biomarkers may also provide clues to identify potential molecular targets for future therapeutic trials. Herein we report on a pilot study involving twelve participants with ALS and nine age-matched healthy controls who underwent high-resolution resting state functional magnetic resonance imaging at an ultra-high field of 7 Tesla. A group-level whole-brain analysis revealed a disruption in long-range functional connectivity between the superior sensorimotor cortex (in the precentral gyrus) and bilateral cerebellar lobule VI. Post hoc analyses using atlas-derived left and right cerebellar lobule VI revealed decreased functional connectivity in ALS participants that predominantly mapped to bilateral postcentral and precentral gyri. Cerebellar lobule VI is a transition zone between anterior motor networks and posterior non-motor networks in the cerebellum, and is associated with a wide range of key functions including complex motor and cognitive processing tasks. Our observation of the involvement of cerebellar lobule VI adds to the growing number of studies implicating the cerebellum in ALS. Future avenues of scientific investigation should consider how high-resolution imaging at 7T may be leveraged to visualize differences in functional connectivity disturbances in various genotypes and phenotypes of ALS along the ALS-frontotemporal dementia spectrum.
Collapse
Affiliation(s)
- Robert L Barry
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA; Department of Radiology, Harvard Medical School, Boston, MA, USA; Harvard-Massachusetts Institute of Technology Health Sciences & Technology, Cambridge, MA, USA.
| | - Suma Babu
- Sean M. Healey & AMG Center for ALS at Massachusetts General Hospital, Department of Neurology, Neurological Clinical Research Institute, Boston, MA, USA; Department of Neurology, Harvard Medical School, Boston, MA, USA.
| | - Sheeba Arnold Anteraper
- Department of Psychology, Northeastern University, Boston, MA, USA; Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Christina Triantafyllou
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA; Department of Radiology, Harvard Medical School, Boston, MA, USA; Siemens Healthineers, Erlangen, Germany
| | - Boris Keil
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA; Department of Radiology, Harvard Medical School, Boston, MA, USA; Mittelhessen University of Applied Sciences, Department of Life Science Engineering, Institute of Medical Physics and Radiation Protection, Giessen, Germany
| | - Olivia E Rowe
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - D Rangaprakash
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA; Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Sabrina Paganoni
- Sean M. Healey & AMG Center for ALS at Massachusetts General Hospital, Department of Neurology, Neurological Clinical Research Institute, Boston, MA, USA; Spaulding Rehabilitation Hospital, Charlestown, MA, USA; Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA, USA
| | - Robert Lawson
- Sean M. Healey & AMG Center for ALS at Massachusetts General Hospital, Department of Neurology, Neurological Clinical Research Institute, Boston, MA, USA
| | - Christina Dheel
- Sean M. Healey & AMG Center for ALS at Massachusetts General Hospital, Department of Neurology, Neurological Clinical Research Institute, Boston, MA, USA
| | - Paul M Cernasov
- Sean M. Healey & AMG Center for ALS at Massachusetts General Hospital, Department of Neurology, Neurological Clinical Research Institute, Boston, MA, USA
| | - Bruce R Rosen
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA; Department of Radiology, Harvard Medical School, Boston, MA, USA; Harvard-Massachusetts Institute of Technology Health Sciences & Technology, Cambridge, MA, USA
| | - Eva-Maria Ratai
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA; Department of Radiology, Harvard Medical School, Boston, MA, USA; Division of Neuroradiology, Massachusetts General Hospital, Boston, MA, USA
| | - Nazem Atassi
- Sean M. Healey & AMG Center for ALS at Massachusetts General Hospital, Department of Neurology, Neurological Clinical Research Institute, Boston, MA, USA; Department of Neurology, Harvard Medical School, Boston, MA, USA; Sanofi Genzyme, Cambridge, MA, USA
| |
Collapse
|
39
|
Robinson JL, Porta S, Garrett FG, Zhang P, Xie SX, Suh E, Van Deerlin VM, Abner EL, Jicha GA, Barber JM, Lee VMY, Lee EB, Trojanowski JQ, Nelson PT. Limbic-predominant age-related TDP-43 encephalopathy differs from frontotemporal lobar degeneration. Brain 2021; 143:2844-2857. [PMID: 32830216 DOI: 10.1093/brain/awaa219] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/01/2020] [Accepted: 05/21/2020] [Indexed: 12/12/2022] Open
Abstract
TAR-DNA binding protein-43 (TDP-43) proteinopathy is seen in multiple brain diseases. A standardized terminology was recommended recently for common age-related TDP-43 proteinopathy: limbic-predominant, age-related TDP-43 encephalopathy (LATE) and the underlying neuropathological changes, LATE-NC. LATE-NC may be co-morbid with Alzheimer's disease neuropathological changes (ADNC). However, there currently are ill-defined diagnostic classification issues among LATE-NC, ADNC, and frontotemporal lobar degeneration with TDP-43 (FTLD-TDP). A practical challenge is that different autopsy cohorts are composed of disparate groups of research volunteers: hospital- and clinic-based cohorts are enriched for FTLD-TDP cases, whereas community-based cohorts have more LATE-NC cases. Neuropathological methods also differ across laboratories. Here, we combined both cases and neuropathologists' diagnoses from two research centres-University of Pennsylvania and University of Kentucky. The study was designed to compare neuropathological findings between FTLD-TDP and pathologically severe LATE-NC. First, cases were selected from the University of Pennsylvania with pathological diagnoses of either FTLD-TDP (n = 33) or severe LATE-NC (mostly stage 3) with co-morbid ADNC (n = 30). Sections from these University of Pennsylvania cases were cut from amygdala, anterior cingulate, superior/mid-temporal, and middle frontal gyrus. These sections were stained for phospho-TDP-43 immunohistochemically and evaluated independently by two University of Kentucky neuropathologists blinded to case data. A simple set of criteria hypothesized to differentiate FTLD-TDP from LATE-NC was generated based on density of TDP-43 immunoreactive neuronal cytoplasmic inclusions in the neocortical regions. Criteria-based sensitivity and specificity of differentiating severe LATE-NC from FTLD-TDP cases with blind evaluation was ∼90%. Another proposed neuropathological feature related to TDP-43 proteinopathy in aged individuals is 'Alpha' versus 'Beta' in amygdala. Alpha and Beta status was diagnosed by neuropathologists from both universities (n = 5 raters). There was poor inter-rater reliability of Alpha/Beta classification (mean κ = 0.31). We next tested a separate cohort of cases from University of Kentucky with either FTLD-TDP (n = 8) or with relatively 'pure' severe LATE-NC (lacking intermediate or severe ADNC; n = 14). The simple criteria were applied by neuropathologists blinded to the prior diagnoses at University of Pennsylvania. Again, the criteria for differentiating LATE-NC from FTLD-TDP was effective, with sensitivity and specificity ∼90%. If more representative cases from each cohort (including less severe TDP-43 proteinopathy) had been included, the overall accuracy for identifying LATE-NC was estimated at >98% for both cohorts. Also across both cohorts, cases with FTLD-TDP died younger than those with LATE-NC (P < 0.0001). We conclude that in most cases, severe LATE-NC and FTLD-TDP can be differentiated by applying simple neuropathological criteria.
Collapse
Affiliation(s)
- John L Robinson
- Alzheimer's Disease Core Center, University of Pennsyvania, Philadelphia, PA, USA.,Center for Neurodegenerative Disease Research, University of Pennsyvania, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, University of Pennsyvania, Philadelphia, PA, USA
| | - Sílvia Porta
- Alzheimer's Disease Core Center, University of Pennsyvania, Philadelphia, PA, USA.,Center for Neurodegenerative Disease Research, University of Pennsyvania, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, University of Pennsyvania, Philadelphia, PA, USA
| | - Filip G Garrett
- Department of Pathology, University of Kentucky, Lexington, KY, USA
| | - Panpan Zhang
- Alzheimer's Disease Core Center, University of Pennsyvania, Philadelphia, PA, USA.,Department of Biostatistics, Epidemiology and Informatics, University of Pennsyvania, Philadelphia, PA, USA
| | - Sharon X Xie
- Alzheimer's Disease Core Center, University of Pennsyvania, Philadelphia, PA, USA.,Department of Biostatistics, Epidemiology and Informatics, University of Pennsyvania, Philadelphia, PA, USA
| | - EunRan Suh
- Alzheimer's Disease Core Center, University of Pennsyvania, Philadelphia, PA, USA.,Center for Neurodegenerative Disease Research, University of Pennsyvania, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, University of Pennsyvania, Philadelphia, PA, USA
| | - Vivianna M Van Deerlin
- Alzheimer's Disease Core Center, University of Pennsyvania, Philadelphia, PA, USA.,Center for Neurodegenerative Disease Research, University of Pennsyvania, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, University of Pennsyvania, Philadelphia, PA, USA
| | - Erin L Abner
- Department of Epidemiology, University of Kentucky, Lexington, KY, USA.,Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Gregory A Jicha
- Department of Neurology, University of Kentucky, Lexington, KY, USA.,Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Justin M Barber
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Virginia M-Y Lee
- Alzheimer's Disease Core Center, University of Pennsyvania, Philadelphia, PA, USA.,Center for Neurodegenerative Disease Research, University of Pennsyvania, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, University of Pennsyvania, Philadelphia, PA, USA
| | - Edward B Lee
- Alzheimer's Disease Core Center, University of Pennsyvania, Philadelphia, PA, USA.,Center for Neurodegenerative Disease Research, University of Pennsyvania, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, University of Pennsyvania, Philadelphia, PA, USA
| | - John Q Trojanowski
- Alzheimer's Disease Core Center, University of Pennsyvania, Philadelphia, PA, USA.,Center for Neurodegenerative Disease Research, University of Pennsyvania, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, University of Pennsyvania, Philadelphia, PA, USA
| | - Peter T Nelson
- Department of Pathology, University of Kentucky, Lexington, KY, USA.,Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
40
|
Schmahmann JD. Emotional disorders and the cerebellum: Neurobiological substrates, neuropsychiatry, and therapeutic implications. HANDBOOK OF CLINICAL NEUROLOGY 2021; 183:109-154. [PMID: 34389114 DOI: 10.1016/b978-0-12-822290-4.00016-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The notion that the cerebellum is devoted exclusively to motor control has been replaced by a more sophisticated understanding of its role in neurological function, one that includes cognition and emotion. Early clinical reports, as well as physiological and behavioral studies in animal models, raised the possibility of a nonmotor role for the cerebellum. Anatomical studies demonstrate cerebellar connectivity with the distributed neural circuits linked with autonomic, sensorimotor, vestibular, associative, and limbic/paralimbic brain areas. Identification of the cerebellar cognitive affective syndrome in adults and children underscored the clinical relevance of the role of the cerebellum in cognition and emotion. It opened new avenues of investigation into higher-order deficits that accompany the ataxias and other cerebellar diseases, as well as the contribution of cerebellar dysfunction to neuropsychiatric and neurocognitive disorders. Brain imaging studies have demonstrated the complexity of cerebellar functional topography, revealing a double representation of the sensorimotor cerebellum in the anterior lobe and lobule VIII and a triple cognitive representation in the cerebellar posterior lobe, as well as representation in the cerebellum of the intrinsic connectivity networks identified in the cerebral hemispheres. This paradigm shift in thinking about the cerebellum has been advanced by the theories of dysmetria of thought and the universal cerebellar transform, harmonizing the dual anatomic realities of homogeneously repeating cerebellar cortical microcircuitry set against the heterogeneous and topographically arranged cerebellar connections with extracerebellar structures. This new appreciation of cerebellar incorporation into circuits that subserve cognition and emotion mandates a deeper understanding of the cerebellum by practitioners in behavioral neurology and neuropsychiatry because it impacts the understanding and diagnosis of disorders of emotion and intellect and has potential for novel cerebellar-based approaches to therapy.
Collapse
Affiliation(s)
- Jeremy D Schmahmann
- Ataxia Center, Cognitive Behavioral Neurology Unit, Laboratory for Neuroanatomy and Cerebellar Neurobiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
41
|
Human ALS/FTD brain organoid slice cultures display distinct early astrocyte and targetable neuronal pathology. Nat Neurosci 2021; 24:1542-1554. [PMID: 34675437 PMCID: PMC8553627 DOI: 10.1038/s41593-021-00923-4] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 08/16/2021] [Indexed: 12/09/2022]
Abstract
Amyotrophic lateral sclerosis overlapping with frontotemporal dementia (ALS/FTD) is a fatal and currently untreatable disease characterized by rapid cognitive decline and paralysis. Elucidating initial cellular pathologies is central to therapeutic target development, but obtaining samples from presymptomatic patients is not feasible. Here, we report the development of a cerebral organoid slice model derived from human induced pluripotent stem cells (iPSCs) that recapitulates mature cortical architecture and displays early molecular pathology of C9ORF72 ALS/FTD. Using a combination of single-cell RNA sequencing and biological assays, we reveal distinct transcriptional, proteostasis and DNA repair disturbances in astroglia and neurons. We show that astroglia display increased levels of the autophagy signaling protein P62 and that deep layer neurons accumulate dipeptide repeat protein poly(GA), DNA damage and undergo nuclear pyknosis that could be pharmacologically rescued by GSK2606414. Thus, patient-specific iPSC-derived cortical organoid slice cultures are a reproducible translational platform to investigate preclinical ALS/FTD mechanisms as well as novel therapeutic approaches.
Collapse
|
42
|
Clinical Update on C9orf72: Frontotemporal Dementia, Amyotrophic Lateral Sclerosis, and Beyond. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1281:67-76. [PMID: 33433869 DOI: 10.1007/978-3-030-51140-1_5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The identification of C9orf72 gene has led to important scientific progresses and has considerably changed our clinical practice. However, a decade after C9orf72 discovery, some important clinical questions remain unsolved. The reliable cutoff for the pathogenic repeat number and the implication of intermediate alleles in frontotemporal dementia, amyotrophic lateral sclerosis, or in other diseases are still uncertain. The occurrence of an anticipation phenomenon - at the clinical and molecular levels - in C9orf72 kindreds is still debated as well, and the factors driving age at onset and phenotype variability are largely unknown. All these questions have a significant impact not only in clinical practice for diagnosis and genetic counseling but also in a research context for the initiation of therapeutic trials. In this chapter, we will address all those issues and summarize the recent updates about clinical aspects of C9orf72 disease, focusing on both the common and the less typical phenotypes.
Collapse
|
43
|
Cunningham KM, Maulding K, Ruan K, Senturk M, Grima JC, Sung H, Zuo Z, Song H, Gao J, Dubey S, Rothstein JD, Zhang K, Bellen HJ, Lloyd TE. TFEB/Mitf links impaired nuclear import to autophagolysosomal dysfunction in C9-ALS. eLife 2020; 9:59419. [PMID: 33300868 PMCID: PMC7758070 DOI: 10.7554/elife.59419] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 12/09/2020] [Indexed: 12/13/2022] Open
Abstract
Disrupted nucleocytoplasmic transport (NCT) has been implicated in neurodegenerative disease pathogenesis; however, the mechanisms by which disrupted NCT causes neurodegeneration remain unclear. In a Drosophila screen, we identified ref(2)P/p62, a key regulator of autophagy, as a potent suppressor of neurodegeneration caused by the GGGGCC hexanucleotide repeat expansion (G4C2 HRE) in C9orf72 that causes amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). We found that p62 is increased and forms ubiquitinated aggregates due to decreased autophagic cargo degradation. Immunofluorescence and electron microscopy of Drosophila tissues demonstrate an accumulation of lysosome-like organelles that precedes neurodegeneration. These phenotypes are partially caused by cytoplasmic mislocalization of Mitf/TFEB, a key transcriptional regulator of autophagolysosomal function. Additionally, TFEB is mislocalized and downregulated in human cells expressing GGGGCC repeats and in C9-ALS patient motor cortex. Our data suggest that the C9orf72-HRE impairs Mitf/TFEB nuclear import, thereby disrupting autophagy and exacerbating proteostasis defects in C9-ALS/FTD.
Collapse
Affiliation(s)
- Kathleen M Cunningham
- Cellular and Molecular Medicine Program, School of Medicine, Johns Hopkins University, Baltimore, United States
| | - Kirstin Maulding
- Cellular and Molecular Medicine Program, School of Medicine, Johns Hopkins University, Baltimore, United States
| | - Kai Ruan
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, United States
| | - Mumine Senturk
- Program in Developmental Biology, Baylor College of Medicine (BCM), Houston, United States
| | - Jonathan C Grima
- Brain Science Institute, School of Medicine, Johns Hopkins University, Baltimore, United States.,Solomon H. Snyder Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, United States
| | - Hyun Sung
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, United States
| | - Zhongyuan Zuo
- Department of Molecular and Human Genetics, BCM, Houston, United States
| | - Helen Song
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, United States
| | - Junli Gao
- Department of Neuroscience, Mayo Clinic, Jacksonville, United States
| | - Sandeep Dubey
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, United States
| | - Jeffrey D Rothstein
- Cellular and Molecular Medicine Program, School of Medicine, Johns Hopkins University, Baltimore, United States.,Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, United States.,Brain Science Institute, School of Medicine, Johns Hopkins University, Baltimore, United States.,Solomon H. Snyder Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, United States
| | - Ke Zhang
- Department of Neuroscience, Mayo Clinic, Jacksonville, United States
| | - Hugo J Bellen
- Program in Developmental Biology, Baylor College of Medicine (BCM), Houston, United States.,Department of Molecular and Human Genetics, BCM, Houston, United States.,Department of Neuroscience, BCM, Houston, United States.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, United States.,Howard Hughes Medical Institute, Houston, United States
| | - Thomas E Lloyd
- Cellular and Molecular Medicine Program, School of Medicine, Johns Hopkins University, Baltimore, United States.,Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, United States.,Solomon H. Snyder Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, United States
| |
Collapse
|
44
|
Klotz S, König T, Erdler M, Ulram A, Nguyen A, Ströbel T, Zimprich A, Stögmann E, Regelsberger G, Höftberger R, Budka H, Kovacs GG, Gelpi E. Co-incidental C9orf72 expansion mutation-related frontotemporal lobar degeneration pathology and sporadic Creutzfeldt-Jakob disease. Eur J Neurol 2020; 28:1009-1015. [PMID: 33131137 PMCID: PMC7898301 DOI: 10.1111/ene.14621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/26/2020] [Indexed: 11/29/2022]
Abstract
Background The C9orf72 hexanucleotide expansion mutation is the most common cause of genetic frontotemporal dementia (FTD), amyotrophic lateral sclerosis (ALS) and combined FTD‐ALS. Its underlying neuropathology combines TDP‐43 pathology and dipeptide repeat protein (DPR) deposits and may also associate with other neurodegeneration‐associated protein aggregates. Herein we present a unique combination of C9orf72 mutation with sporadic Creutzfeldt−Jakob disease (CJD) in a 74‐year‐old patient with rapidly progressive dementia. Methods Detailed neuropathological examination including immunohistochemistry for several proteinopathies. Genetic analysis was conducted by repeat primed polymerase chain reaction (PCR). Furthermore, we analyzed additional C9orf72 mutation carriers for prion−protein (PrP) deposits in brain tissue and screened the cerebellar cortex of other CJD cases for p62/DPR neuronal inclusions to assess the frequency of combined pathologies. Results Postmortem brain examination of a patient with a rapidly progressive neurological deterioration of 8 months’ duration confirmed the diagnosis of CJD. She harbored valine homozygosity at PRNP codon 129. In addition, a frontotemporal lobar degeneration (FTLD)‐pattern with TDP‐43 protein aggregates and p62+/C9RANT+ positive inclusions along with a high degree of Alzheimer‐related pathology (A3B3C3) were identified. The suspected C9orf72 expansion mutation was confirmed by repeat‐primed PCR. Screening of 13 C9orf72 cases showed no pathological PrP aggregates and screening of 100 CJD cases revealed no other C9orf72 expansion mutation carriers. Conclusion A combination of a C9orf72 expansion mutation‐related FTLD with sporadic CJD in the same patient is rare. While the rarity of both diseases makes this concurrence most likely to be coincidental, questions regarding a potential link between these two neurodegenerative pathologies deserve further studies.
Collapse
Affiliation(s)
- Sigrid Klotz
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria.,Austrian Reference Center for Human Prion Diseases (OERPE), Vienna, Austria
| | - Theresa König
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Marcus Erdler
- Department of Neurology, Klinik Donaustadt mit Ludwig-Boltzmann-Institut, Vienna, Austria
| | - Andreas Ulram
- Department of Neurosurgery, Krankenanstalt Rudolfstiftung, Vienna, Austria
| | - Anita Nguyen
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Thomas Ströbel
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria.,Austrian Reference Center for Human Prion Diseases (OERPE), Vienna, Austria
| | | | | | - Günther Regelsberger
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria.,Austrian Reference Center for Human Prion Diseases (OERPE), Vienna, Austria
| | - Romana Höftberger
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Herbert Budka
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria.,Austrian Reference Center for Human Prion Diseases (OERPE), Vienna, Austria
| | - Gabor G Kovacs
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology and Department of Medicine, University of Toronto, Toronto, ON, Canada.,Laboratory Medicine Program & Krembil Brain Institute, University Health Network, Toronto, ON, Canada
| | - Ellen Gelpi
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria.,Austrian Reference Center for Human Prion Diseases (OERPE), Vienna, Austria
| |
Collapse
|
45
|
Gagliardi D, Costamagna G, Taiana M, Andreoli L, Biella F, Bersani M, Bresolin N, Comi GP, Corti S. Insights into disease mechanisms and potential therapeutics for C9orf72-related amyotrophic lateral sclerosis/frontotemporal dementia. Ageing Res Rev 2020; 64:101172. [PMID: 32971256 DOI: 10.1016/j.arr.2020.101172] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 08/31/2020] [Indexed: 12/12/2022]
Abstract
In 2011, a hexanucleotide repeat expansion (HRE) in the noncoding region of C9orf72 was associated with the most frequent genetic cause of frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS). The main pathogenic mechanisms in C9-ALS/FTD are haploinsufficiency of the C9orf72 protein and gain of function toxicity from bidirectionally-transcribed repeat-containing RNAs and dipeptide repeat proteins (DPRs) resulting from non-canonical RNA translation. Additionally, abnormalities in different downstream cellular mechanisms, such as nucleocytoplasmic transport and autophagy, play a role in pathogenesis. Substantial research efforts using in vitro and in vivo models have provided valuable insights into the contribution of each mechanism in disease pathogenesis. However, conflicting evidence exists, and a unifying theory still lacks. Here, we provide an overview of the recently published literature on clinical, neuropathological and molecular features of C9-ALS/FTD. We highlight the supposed neuronal role of C9orf72 and the HRE pathogenic cascade, mainly focusing on the contribution of RNA foci and DPRs to neurodegeneration and discussing the several downstream mechanisms. We summarize the emerging biochemical and neuroimaging biomarkers, as well as the potential therapeutic approaches. Despite promising results, a specific disease-modifying treatment is still not available to date and greater insights into disease mechanisms may help in this direction.
Collapse
Affiliation(s)
- Delia Gagliardi
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Gianluca Costamagna
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Michela Taiana
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Luca Andreoli
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Fabio Biella
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Margherita Bersani
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Nereo Bresolin
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy; Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milan, Italy
| | - Giacomo Pietro Comi
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy; Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milan, Italy
| | - Stefania Corti
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy; Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milan, Italy.
| |
Collapse
|
46
|
Trejo-Lopez JA, Sorrentino ZA, Riffe CJ, Prokop S, Dickson DW, Yachnis AT, Giasson BI. Generation and Characterization of Novel Monoclonal Antibodies Targeting p62/sequestosome-1 Across Human Neurodegenerative Diseases. J Neuropathol Exp Neurol 2020; 79:407-418. [PMID: 32106300 DOI: 10.1093/jnen/nlaa007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 01/25/2020] [Indexed: 12/17/2022] Open
Abstract
Human neurodegenerative diseases can be characterized as disorders of protein aggregation. As a key player in cellular autophagy and the ubiquitin proteasome system, p62 may represent an effective immunohistochemical target, as well as mechanistic operator, across neurodegenerative proteinopathies. In this study, 2 novel mouse-derived monoclonal antibodies 5G3 and 2A5 raised against residues 360-380 of human p62/sequestosome-1 were characterized via immunohistochemical application upon human tissues derived from cases of C9orf72-expansion spectrum diseases, Alzheimer disease, progressive supranuclear palsy, Lewy body disease, and multiple system atrophy. 5G3 and 2A5 reliably highlighted neuronal dipeptide repeat, tau, and α-synuclein inclusions in a distribution similar to a polyclonal antibody to p62, phospho-tau antibodies 7F2 and AT8, and phospho-α-synuclein antibody 81A. However, antibodies 5G3 and 2A5 consistently stained less neuropil structures, such as tau neuropil threads and Lewy neurites, while 2A5 marked fewer glial inclusions in progressive supranuclear palsy. Both 5G3 and 2A5 revealed incidental astrocytic tau immunoreactivity in cases of Alzheimer disease and Lewy body disease with resolution superior to 7F2. Through their unique ability to highlight specific types of pathological deposits in neurodegenerative brain tissue, these novel monoclonal p62 antibodies may provide utility in both research and diagnostic efforts.
Collapse
Affiliation(s)
- Jorge A Trejo-Lopez
- Department of Pathology, Immunology, and Laboratory Medicine.,Center for Translational Research in Neurodegenerative Disease
| | - Zachary A Sorrentino
- Center for Translational Research in Neurodegenerative Disease.,Department of Neuroscience
| | - Cara J Riffe
- Center for Translational Research in Neurodegenerative Disease.,Department of Neuroscience
| | - Stefan Prokop
- Department of Pathology, Immunology, and Laboratory Medicine.,Center for Translational Research in Neurodegenerative Disease.,McKnight Brain Institute.,Fixel Institute for Neurological Diseases, University of Florida, Gainesville, Florida
| | | | | | - Benoit I Giasson
- Center for Translational Research in Neurodegenerative Disease.,Department of Neuroscience.,McKnight Brain Institute
| |
Collapse
|
47
|
Teyssou E, Muratet F, Amador MDM, Ferrien M, Lautrette G, Machat S, Boillée S, Larmonier T, Saker S, Leguern E, Cazeneuve C, Marie Y, Guegan J, Gyorgy B, Cintas P, Meininger V, Le Forestier N, Salachas F, Couratier P, Camu W, Seilhean D, Millecamps S. Genetic screening of ANXA11 revealed novel mutations linked to amyotrophic lateral sclerosis. Neurobiol Aging 2020; 99:102.e11-102.e20. [PMID: 33218681 DOI: 10.1016/j.neurobiolaging.2020.10.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 10/02/2020] [Accepted: 10/18/2020] [Indexed: 12/11/2022]
Abstract
ANXA11 mutations have previously been discovered in amyotrophic lateral sclerosis (ALS) motor neuron disease. To confirm the contribution of ANXA11 mutations to ALS, a large exome data set obtained from 330 French patients, including 150 familial ALS index cases and 180 sporadic ALS cases, was analyzed, leading to the identification of 3 rare ANXA11 variants in 5 patients. The novel p.L254V variant was associated with early onset sporadic ALS. The novel p.D40Y mutation and the p.G38R variant concerned patients with predominant pyramidal tract involvement and cognitive decline. Neuropathologic findings in a p.G38R carrier associated the presence of ALS typical inclusions within the spinal cord, massive degeneration of the lateral tracts, and type A frontotemporal lobar degeneration. This mutant form of annexin A11 accumulated in various brain regions and in spinal cord motor neurons, although its stability was decreased in patients' lymphoblasts. Because most ANXA11 inclusions were not colocalized with transactive response DNA-binding protein 43 or p62 deposits, ANXA11 aggregation does not seem mandatory to trigger neurodegeneration with additional participants/partner proteins that could intervene.
Collapse
Affiliation(s)
- Elisa Teyssou
- Institut du Cerveau et de la Moelle épinière, ICM, Inserm U1127, CNRS UMR7225, Sorbonne Université, UPMC Univ Paris 6 UMRS1127, Paris, France
| | - François Muratet
- Institut du Cerveau et de la Moelle épinière, ICM, Inserm U1127, CNRS UMR7225, Sorbonne Université, UPMC Univ Paris 6 UMRS1127, Paris, France
| | - Maria-Del-Mar Amador
- Institut du Cerveau et de la Moelle épinière, ICM, Inserm U1127, CNRS UMR7225, Sorbonne Université, UPMC Univ Paris 6 UMRS1127, Paris, France; Département de Neurologie, Assistance Publique Hôpitaux de Paris (APHP), Centre de référence SLA Ile de France, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Mélanie Ferrien
- Institut du Cerveau et de la Moelle épinière, ICM, Inserm U1127, CNRS UMR7225, Sorbonne Université, UPMC Univ Paris 6 UMRS1127, Paris, France
| | - Géraldine Lautrette
- Service de Neurologie, Centre de Référence SLA et autres maladies du neurone moteur, Limoges, France
| | - Selma Machat
- Service de Neurologie, Centre de Référence SLA et autres maladies du neurone moteur, Limoges, France
| | - Séverine Boillée
- Institut du Cerveau et de la Moelle épinière, ICM, Inserm U1127, CNRS UMR7225, Sorbonne Université, UPMC Univ Paris 6 UMRS1127, Paris, France
| | | | - Safaa Saker
- Banque d'ADN et de cellules du Généthon, Evry, France
| | - Eric Leguern
- Institut du Cerveau et de la Moelle épinière, ICM, Inserm U1127, CNRS UMR7225, Sorbonne Université, UPMC Univ Paris 6 UMRS1127, Paris, France; Département de Génétique et Cytogénétique, Unité Fonctionnelle de neurogénétique moléculaire et cellulaire, APHP, Hôpital Pitié-Salpêtrière, Paris, France
| | - Cécile Cazeneuve
- Département de Génétique et Cytogénétique, Unité Fonctionnelle de neurogénétique moléculaire et cellulaire, APHP, Hôpital Pitié-Salpêtrière, Paris, France
| | - Yannick Marie
- Institut du Cerveau et de la Moelle épinière, ICM, Inserm U1127, CNRS UMR7225, Sorbonne Université, UPMC Univ Paris 6 UMRS1127, Paris, France
| | - Justine Guegan
- Institut du Cerveau et de la Moelle épinière, ICM, Inserm U1127, CNRS UMR7225, Sorbonne Université, UPMC Univ Paris 6 UMRS1127, Paris, France
| | - Beata Gyorgy
- Institut du Cerveau et de la Moelle épinière, ICM, Inserm U1127, CNRS UMR7225, Sorbonne Université, UPMC Univ Paris 6 UMRS1127, Paris, France
| | - Pascal Cintas
- Département de Neurologie, Centre de référence SLA, CHU de Toulouse, Toulouse, France
| | | | - Nadine Le Forestier
- Département de Neurologie, Assistance Publique Hôpitaux de Paris (APHP), Centre de référence SLA Ile de France, Hôpital de la Pitié-Salpêtrière, Paris, France; Département de recherche en éthique, EA 1610, Etudes des sciences et techniques, Université Paris Sud/Paris Saclay, Orsay, France
| | - François Salachas
- Institut du Cerveau et de la Moelle épinière, ICM, Inserm U1127, CNRS UMR7225, Sorbonne Université, UPMC Univ Paris 6 UMRS1127, Paris, France; Département de Neurologie, Assistance Publique Hôpitaux de Paris (APHP), Centre de référence SLA Ile de France, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Philippe Couratier
- Service de Neurologie, Centre de Référence SLA et autres maladies du neurone moteur, Limoges, France
| | - William Camu
- Centre de référence SLA, Hôpital Gui de Chauliac, CHU et Université de Montpellier, Montpellier, France
| | - Danielle Seilhean
- Institut du Cerveau et de la Moelle épinière, ICM, Inserm U1127, CNRS UMR7225, Sorbonne Université, UPMC Univ Paris 6 UMRS1127, Paris, France; Département de Neuropathologie, APHP, Hôpital Pitié-Salpêtrière, Paris, France
| | - Stéphanie Millecamps
- Institut du Cerveau et de la Moelle épinière, ICM, Inserm U1127, CNRS UMR7225, Sorbonne Université, UPMC Univ Paris 6 UMRS1127, Paris, France.
| |
Collapse
|
48
|
Modelling frontotemporal dementia using patient-derived induced pluripotent stem cells. Mol Cell Neurosci 2020; 109:103553. [PMID: 32956830 DOI: 10.1016/j.mcn.2020.103553] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 08/27/2020] [Accepted: 09/12/2020] [Indexed: 12/12/2022] Open
Abstract
Frontotemporal dementia (FTD) describes a group of clinically heterogeneous conditions that frequently affect people under the age of 65 (Le Ber et al., 2013). There are multiple genetic causes of FTD, including coding or splice-site mutations in MAPT, GRN mutations that lead to haploinsufficiency of progranulin protein, and a hexanucleotide GGGGCC repeat expansion in C9ORF72. Pathologically, FTD is characterised by abnormal protein accumulations in neurons and glia. These aggregates can be composed of the microtubule-associated protein tau (observed in FTD with MAPT mutations), the DNA/RNA-binding protein TDP-43 (seen in FTD with mutations in GRN or C9ORF72 repeat expansions) or dipeptide proteins generated by repeat associated non-ATG translation of the C9ORF72 repeat expansion. There are currently no disease-modifying therapies for FTD and the availability of in vitro models that recapitulate pathologies in a disease-relevant cell type would accelerate the development of novel therapeutics. It is now possible to generate patient-specific stem cells through the reprogramming of somatic cells from a patient with a genotype/phenotype of interest into induced pluripotent stem cells (iPSCs). iPSCs can subsequently be differentiated into a plethora of cell types including neurons, astrocytes and microglia. Using this approach has allowed researchers to generate in vitro models of genetic FTD in human cell types that are largely inaccessible during life. In this review we explore the recent progress in the use of iPSCs to model FTD, and consider the merits, limitations and future prospects of this approach.
Collapse
|
49
|
Vasques JF, Mendez-Otero R, Gubert F. Modeling ALS using iPSCs: is it possible to reproduce the phenotypic variations observed in patients in vitro? Regen Med 2020; 15:1919-1933. [PMID: 32795164 DOI: 10.2217/rme-2020-0067] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal disease that leads to progressive degeneration of motoneurons. Mutations in the C9ORF72, SOD1, TARDBP and FUS genes, among others, have been associated with ALS. Although motoneuron degeneration is the common outcome of ALS, different pathological mechanisms seem to be involved in this process, depending on the genotypic background of the patient. The advent of induced pluripotent stem cell (iPSC) technology enabled the development of patient-specific cell lines, from which it is possible to generate different cell types and search for phenotypic alterations. In this review, we summarize the pathophysiological markers detected in cells differentiated from iPSCs of ALS patients. In a translational perspective, iPSCs from ALS patients could be useful for drug screening, through stratifying patients according to their genetic background.
Collapse
Affiliation(s)
- Juliana Ferreira Vasques
- Instituto de Biofísica Carlos Chagas Filho, UFRJ, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa
| | - Rosalia Mendez-Otero
- Instituto de Biofísica Carlos Chagas Filho, UFRJ, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa
| | - Fernanda Gubert
- Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa.,Instituto de Ciências Biomédicas, UFRJ, Rio de Janeiro, Brazil
| |
Collapse
|
50
|
Tang X, Toro A, T G S, Gao J, Chalk J, Oskarsson B, Zhang K. Divergence, Convergence, and Therapeutic Implications: A Cell Biology Perspective of C9ORF72-ALS/FTD. Mol Neurodegener 2020; 15:34. [PMID: 32513219 PMCID: PMC7282082 DOI: 10.1186/s13024-020-00383-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 05/26/2020] [Indexed: 12/12/2022] Open
Abstract
Ever since a GGGGCC hexanucleotide repeat expansion mutation in C9ORF72 was identified as the most common cause of familial amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), three competing but nonexclusive hypotheses to explain how this mutation causes diseases have been proposed and are still under debate. Recent studies in the field have tried to understand how the repeat expansion disrupts cellular physiology, which has suggested interesting convergence of these hypotheses on downstream, functional defects in cells, such as nucleocytoplasmic transport disruption, membrane-less organelle defects, and DNA damage. These studies have not only provided an integrated view of the disease mechanism but also revealed novel cell biology implicated in neurodegeneration. Furthermore, some of the discoveries have given rise to new ideas for therapeutic development. Here, we review the research progress on cellular pathophysiology of C9ORF72-mediated ALS and FTD and its therapeutic implication. We suggest that the repeat expansion drives pathogenesis through a combination of downstream defects, of which some can be therapeutic targets.
Collapse
Affiliation(s)
- Xiaoqiang Tang
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Arturo Toro
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Sahana T G
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Junli Gao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Jessica Chalk
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Ke Zhang
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA. .,Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, USA.
| |
Collapse
|