1
|
Krishnan D, Talbot DL, Ashhurst JF, Park SB, Vucic S, Timmins HC, Kiernan MC. Longitudinal assessment of cortical motor function in amyotrophic lateral sclerosis. Sci Rep 2025; 15:16978. [PMID: 40374755 PMCID: PMC12081609 DOI: 10.1038/s41598-025-01570-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 05/07/2025] [Indexed: 05/18/2025] Open
Abstract
Background Short interval intracortical inhibition (SICI) remains the most sensitive parameter to assess motor cortical function in amyotrophic lateral sclerosis (ALS). While an initial value of SICI has been utilised to support a diagnosis of ALS, less is known about progression of change. Methods Motor cortex function was prospectively assessed in ALS patients, through serial threshold tracking transcranial magnetic stimulation (TMS) assessment over more than 12 months. Motor cortical potentials were recorded from the abductor pollicis brevis (APB). Demographic information and clinical variables were analysed. Results A cohort of 52 ALS patients (69.2% limb-onset disease; 47.2% right-side) were assigned to undergo longitudinal assessment of cortical motor function. Mean ALSFRS-R score at baseline was 39.5 ± 1.0 denoting relatively milder clinical deficits at study commencement. Cortical motor dysfunction was evident at baseline, with reduction in averaged SICI (p = 0.004) when compared to healthy controls. In terms of disease trajectory, ALS patients experienced a significant decline in averaged SICI overtime. When compared to initial assessment, averaged SICI was significantly reduced after 12 months (p = 0.004). There was no significant main effect of site of onset on averaged SICI (p = 0.78). The progressive change in averaged SICI was more robust in the dominant hemisphere, with the proportion of ALS patients who demonstrated a clinically abnormal averaged SICI value (< 5.5%) increasing by 50%, compared to 15.4% for the non-dominant hemisphere. Conclusion ALS patients demonstrate progressive cortical motor abnormalities, evident through longitudinal assessment. While SICI represents a diagnostic biomarker, the rate of decline in the present series is consistent with disease progression, suggesting a potential role to monitor the efficacy of therapeutic intervention.
Collapse
Affiliation(s)
- Dhayalen Krishnan
- Neuroscience Research Australia, Barker St, Randwick, Sydney, NSW, 2031, Australia
| | - Dayna-Lee Talbot
- Neuroscience Research Australia, Barker St, Randwick, Sydney, NSW, 2031, Australia
| | - Jasmine F Ashhurst
- Neuroscience Research Australia, Barker St, Randwick, Sydney, NSW, 2031, Australia
- University of New South Wales and South Eastern Sydney Local Health District, Sydney, Australia
| | - Susanna B Park
- School of Medical Sciences, University of Sydney, Camperdown, Australia
| | - Steve Vucic
- Brain and Nerve Centre, Concord Hospital, Sydney, Australia
| | - Hannah C Timmins
- Neuroscience Research Australia, Barker St, Randwick, Sydney, NSW, 2031, Australia
| | - Matthew C Kiernan
- Neuroscience Research Australia, Barker St, Randwick, Sydney, NSW, 2031, Australia.
- University of New South Wales and South Eastern Sydney Local Health District, Sydney, Australia.
| |
Collapse
|
2
|
Calma AD, Pavey N, Silva CS, van den Bos MAJ, Yiannikas C, Farrar MA, Kiernan MC, Menon P, Vucic S. Diagnostic utility of threshold tracking TMS paradigms in early amyotrophic lateral sclerosis. Clin Neurophysiol 2025; 174:105-113. [PMID: 40250284 DOI: 10.1016/j.clinph.2025.03.044] [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: 12/17/2024] [Revised: 03/19/2025] [Accepted: 03/28/2025] [Indexed: 04/20/2025]
Abstract
OBJECTIVE Threshold tracking transcranial magnetic stimulation (TMS) has exhibited utility as a diagnostic technique in Amyotrophic Lateral Sclerosis (ALS). Different threshold tracking paradigms have recently been proposed. The present study assessed the diagnostic utility of serial ascending and parallel threshold tracking TMS in ALS. METHODS Threshold tracking TMS was undertaken on 90 prospectively recruited participants suspected of ALS. Short interval intracortical inhibition (SICI) was recorded with serial ascending and parallel threshold tracking paradigms between Interstimulus Interval (ISI) 1-to-7 ms. The primary outcome measure was differences in diagnostic utility of the paradigms in differentiating ALS from ALS mimicking disorders using receiver operating characteristic (ROC) analysis (DeLong statistical method). RESULTS Reduction in SICI reliably differentiated ALS from mimic disorders, irrespective of the threshold tracking paradigm. Comparison of area under the curve (AUC) established a significantly higher value for mean SICI (1-7 ms) with the serial ascending SICI paradigm (0.81, 95 % confidence interval 0.72-0.91) compared to the parallel paradigm (SICI 0.72, 95 % confidence interval 0.61-0.83, p = 0.0065). The better diagnostic utility of serial ascending paradigm was evident for SICI recorded between 1-to-5 ms, and was maintained irrespective of disease onset site, degree of functional impairment, and the degree of lower motor neuron dysfunction. A comparable diagnostic utility across threshold tracking paradigms was evident in ALS participants who presented with a relative paucity of upper motor neuron signs. CONCLUSION While threshold tracking TMS reliably differentiated ALS from mimic disorders, the present study established better diagnostic utility with the serial ascending threshold tracking TMS paradigm. SIGNIFICANCE The serial ascending threshold tracking TMS should be used in a clinical setting as a diagnostic tool for ALS.
Collapse
Affiliation(s)
- Aicee Dawn Calma
- Brain and Nerve Research Centre, Concord Clinical School, University of Sydney, Concord Hospital, Sydney, Australia; Department of Neurology, Concord Repatriation General Hospital, Sydney, Australia
| | - Nathan Pavey
- Brain and Nerve Research Centre, Concord Clinical School, University of Sydney, Concord Hospital, Sydney, Australia; Department of Neurology, Concord Repatriation General Hospital, Sydney, Australia
| | - Cláudia Santos Silva
- Brain and Nerve Research Centre, Concord Clinical School, University of Sydney, Concord Hospital, Sydney, Australia; Department of Neurosciences and Mental Health, Unidade Local de Saúde de Santa Maria, Lisbon, Portugal; Faculdade de Medicina-Instituto de Medicina Molecular, Centro de Estudos Egas Moniz, Universidade de Lisboa, Lisbon, Portugal
| | - Mehdi A J van den Bos
- Brain and Nerve Research Centre, Concord Clinical School, University of Sydney, Concord Hospital, Sydney, Australia
| | - Con Yiannikas
- Department of Neurology, Concord Repatriation General Hospital, Sydney, Australia
| | - Michelle A Farrar
- Department of Neurology, Sydney Children's Hospital Network, Sydney, NSW, Australia; Discipline of Paediatrics and Child Health, School of Clinical Medicine, UNSW Medicine and Health, The University of New South Wales, Sydney, Australia
| | - Matthew C Kiernan
- Neuroscience Research Australia, Sydney, Australia; Scientia Professor of Neuroscience, University of NSW; and Department of Neurology, SouthEastern Sydney Local Health District, Sydney, Australia
| | - Parvathi Menon
- Brain and Nerve Research Centre, Concord Clinical School, University of Sydney, Concord Hospital, Sydney, Australia; Department of Neurology, Concord Repatriation General Hospital, Sydney, Australia
| | - Steve Vucic
- Brain and Nerve Research Centre, Concord Clinical School, University of Sydney, Concord Hospital, Sydney, Australia; Department of Neurology, Concord Repatriation General Hospital, Sydney, Australia.
| |
Collapse
|
3
|
van den Bos MAJ, Menon P, Pavey N, Higashihara M, Kiernan MC, Vucic S. Direct interrogation of cortical interneuron circuits in amyotrophic lateral sclerosis. Brain 2025; 148:1169-1179. [PMID: 39385724 DOI: 10.1093/brain/awae317] [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: 04/29/2024] [Revised: 07/30/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024] Open
Abstract
Cortical hyperexcitability is a key pathogenic feature of amyotrophic lateral sclerosis (ALS), believed to be mediated through complex interplay of cortical interneurons. To date, there has been no technological approach to facilitate the direct capture of cortical interneuron function. Through combination of transcranial magnetic stimulation (TMS) with advanced EEG, the present study examined GABAergic dysfunction in ALS by recording focused cortical output whilst applying TMS over the primary motor cortex contralateral to the site of symptom onset. Using both a single-pulse and a novel inhibitory paired-pulse paradigm, TMS-EEG studies were undertaken on 21 ALS patients and results compared with healthy controls. TMS responses captured by EEG form a discrete waveform known as the transcranial evoked potential (TEP), with positive (P) or upward deflections occurring at 30 (P30), 60 (P60) and 190 ms (P190) after TMS stimulus. Negative (N) or downward deflections occur at 44 (N44), 100 (N100) and 280 ms (N280) after TMS stimulus. The single-pulse TEPs recorded in ALS patients demonstrated novel differences suggestive of cortical GABAergic dysfunction. When compared with controls, the N100 component was significantly reduced (P < 0.05), whereas the P190 component increased (P < 0.05) in ALS patients. Additionally, the N44 component was correlated with muscle weakness (r = -0.501, P < 0.05). These findings were supported by reduced paired-pulse inhibition of TEP components in ALS patients (P60, P < 0.01; N100, P < 0.005), consistent with dysfunction of cortical interneuronal GABAA-ergic circuits. Furthermore, the reduction in short-interval intracortical inhibition, as reflected by changes in paired-pulse inhibition of the N100 component, was associated with longer disease duration in ALS patients (r = -0.698, P < 0.001). In conclusion, intensive and focused interrogation of the motor cortex using novel TMS-EEG combined technologies has established localized dysfunction of GABAergic circuits, supporting the notion that cortical hyperexcitability is mediated by cortical disinhibition in ALS. Dysfunction of GABAergic circuits was correlated with greater clinical disability and disease duration, implying pathophysiological significance.
Collapse
Affiliation(s)
- Mehdi A J van den Bos
- Brain and Nerve Research Centre, Concord Clinical School, The University of Sydney, Sydney, NSW 2139, Australia
- Concord Repatriation General Hospital, Sydney, NSW 2139, Australia
| | - Parvathi Menon
- Brain and Nerve Research Centre, Concord Clinical School, The University of Sydney, Sydney, NSW 2139, Australia
- Concord Repatriation General Hospital, Sydney, NSW 2139, Australia
| | - Nathan Pavey
- Brain and Nerve Research Centre, Concord Clinical School, The University of Sydney, Sydney, NSW 2139, Australia
- Concord Repatriation General Hospital, Sydney, NSW 2139, Australia
| | - Mana Higashihara
- Department of Neurology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo 173-0015, Japan
| | - Matthew C Kiernan
- Neuroscience Research Australia, Randwick, Sydney, NSW 2031, Australia
- Institute of Neurological Sciences, Eastern Sydney Local Health District, Randwick, Sydney, NSW 2031, Australia
| | - Steve Vucic
- Brain and Nerve Research Centre, Concord Clinical School, The University of Sydney, Sydney, NSW 2139, Australia
- Concord Repatriation General Hospital, Sydney, NSW 2139, Australia
| |
Collapse
|
4
|
Kollstrøm AM, Christiansen N, Sandvig A, Sandvig I. Dysregulation of synaptic transcripts underlies network abnormalities in ALS patient-derived motor neurons. Am J Physiol Cell Physiol 2025; 328:C1029-C1044. [PMID: 39726289 DOI: 10.1152/ajpcell.00725.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is characterized by dysfunction and loss of upper and lower motor neurons. Several studies have identified structural and functional alterations in the motor neurons before the manifestation of symptoms, yet the underlying cause of such alterations and how they contribute to the progressive degeneration of affected motor neuron networks remain unclear. Importantly, the short- and long-term spatiotemporal dynamics of neuronal network activity make it challenging to discern how ALS-related network reconfigurations emerge and evolve. To address this, we systematically monitored the structural and functional dynamics of motor neuron networks with a confirmed endogenous C9orf72 mutation. We show that ALS patient-derived motor neurons display time-dependent neural network dysfunction, specifically reduced firing rate and spike amplitude, impaired bursting, but higher overall synchrony in network activity. These changes coincided with altered neurite outgrowth and branching within the networks. Moreover, transcriptional analyses revealed dysregulation of molecular pathways involved in synaptic development and maintenance, neurite outgrowth, and cell adhesion, suggesting impaired synaptic stabilization. This study identifies early synaptic dysfunction as a contributing mechanism resulting in network-wide structural and functional compensation, which may over time render the networks vulnerable to neurodegeneration.NEW & NOTEWORTHY RNA-sequencing of ALS patient-derived motor neurons revealed altered expression of genes involved in cell adhesion, neurite outgrowth, synaptic development and maintenance, and synaptic plasticity. These alterations were accompanied by time-dependent structural impairments and disrupted neuronal activity, suggesting that early synaptic changes and network-wide structural and functional compensations contribute to motor neuron vulnerability in ALS.
Collapse
Affiliation(s)
- Anna M Kollstrøm
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Nicholas Christiansen
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Axel Sandvig
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Clinical Neurosciences, Division of Neuro, Head and Neck, Umeå University Hospital, Umeå, Sweden
- Department of Community Medicine and Rehabilitation, Umeå University Hospital, Umeå, Sweden
| | - Ioanna Sandvig
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| |
Collapse
|
5
|
Djukic S, Zhao Z, Jørgensen LMH, Bak AN, Jensen DB, Meehan CF. TDP-43 pathology is sufficient to drive axon initial segment plasticity and hyperexcitability of spinal motoneurones in vivo in the TDP43-ΔNLS model of Amyotrophic Lateral Sclerosis. Acta Neuropathol Commun 2025; 13:42. [PMID: 39994742 PMCID: PMC11849383 DOI: 10.1186/s40478-025-01934-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 01/24/2025] [Indexed: 02/26/2025] Open
Abstract
A hyperexcitability of the motor system is consistently observed in Amyotrophic Lateral Sclerosis (ALS) and has been implicated in the disease pathogenesis. What drives this hyperexcitability in the vast majority of patients is unknown. This is important to know as existing treatments simply reduce all neuronal excitability and fail to distinguish between pathological changes and important homeostatic changes. Understanding what drives the initial pathological changes could therefore provide better treatments. One challenge is that patients represent a heterogeneous population and the vast majority of cases are sporadic. One pathological feature that almost all (~97%) cases (familial and sporadic) have in common are cytoplasmic aggregates of the protein TDP-43 which is normally located in the nucleus. In our experiments we investigated whether this pathology was sufficient to increase neuronal excitability and the mechanisms by which this occurs. We used the TDP-43(ΔNLS) mouse model which successfully recapitulates this pathology in a controllable way. We used in vivo intracellular recordings in this model to demonstrate that TDP-43 pathology is sufficient to drive a severe hyper-excitability of spinal motoneurones. Reductions in soma size and a lengthening and constriction of axon initial segments were observed, which would contribute to enhanced excitability. Resuppression of the transgene resulted in a return to normal excitability parameters by 6-8 weeks. We therefore conclude that TDP-43 pathology itself is sufficient to drive a severe but reversible hyperexcitability of spinal motoneurones.
Collapse
Affiliation(s)
- Svetlana Djukic
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Zhenxiang Zhao
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | | | - Anna Normann Bak
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Dennis Bo Jensen
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Claire Francesca Meehan
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark.
| |
Collapse
|
6
|
Vucic S, Pavey N, Menon P, Babayev M, Maslyukova A, Muraviev A, Kiernan MC. Neurophysiological assessment of cortical motor function: A direct comparison of methodologies. Clin Neurophysiol 2025; 170:14-21. [PMID: 39647177 DOI: 10.1016/j.clinph.2024.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/30/2024] [Accepted: 12/01/2024] [Indexed: 12/10/2024]
Abstract
OBJECTIVE Assessment of cortical function with threshold tracking transcranial magnetic stimulation (TT-TMS) has developed as a biomarker to inform disease pathophysiology, particularly in neurodegenerative disease and dementia. At present, a fully integrated testing system does not exist. To advance clinical utility, and to streamline software design to integrate with diagnostic approaches in an outpatient setting, the present series of studies assessed the effects of altering diagnostic paradigms to measure interstimulus interval (ISI) including serial ascending [T-SICIs] and parallel [T-SICIp] methodologies as measures of cortical motor function (the MagXite software). METHODS Cortical excitability was assessed in 30 healthy controls with a figure-of-eight coil, using an integrated approach compared to previously established experimental paradigms. Motor evoked responses were recorded over the contralateral abductor pollicis brevis muscle. Short interval intracortical inhibition (SICI) was recorded with each testing paradigm and validated in a healthy control cohort. RESULTS The integrated system determined a robust measure of T-SICIs between ISI 1-to-7 ms (16.6 ± 2.2 %) that was comparable to previously established testing paradigms (P = 0.34), but greater than T-SICIp (MagXite 10.7 ± 1.5 %, P = 0.016; Sydney TT-TMS 8.7 ± 1.4 %, P = 0.03). SICI peaks at ISI 1 and 2.5-to-3 ms were evident with both protocols. Significant correlations were evident between mean T-SICIs-MagXite and T-SICIp-MagXite (R = 0.599, P < 0.001). CONCLUSION The present series validates a fully integrated motor cortical functional assessment to provide reproducible measures of SICI, with data obtained for intracortical inhibition that is more prominent when assessed using the method of serial ascending order. SIGNIFICANCE An integrated system for transcranial magnetic stimulation of the human motor system has been validated for clinical practice, suitable for the assessment of cortical function in neurological disease in an outpatient clinic setting.
Collapse
Affiliation(s)
- Steve Vucic
- Brain and Nerve Research Center, Concord Clinical School, University of Sydney, Hospital Rd, Concord West, 2139, Sydney, Australia.
| | - Nathan Pavey
- Brain and Nerve Research Center, Concord Clinical School, University of Sydney, Hospital Rd, Concord West, 2139, Sydney, Australia
| | - Parvathi Menon
- Brain and Nerve Research Center, Concord Clinical School, University of Sydney, Hospital Rd, Concord West, 2139, Sydney, Australia
| | | | | | | | - Matthew C Kiernan
- Neuroscience Research Australia, 139 Barker Street, Randwick, 2031, Sydney, Australia; University of NSW and Department of Neurology, Prince of Wales Hospital, South Eastern Sydney Area Health Service, Sydney, Australia
| |
Collapse
|
7
|
Benatar M, Heiman-Patterson TD, Cooper-Knock J, Brickman D, Casaletto KB, Goutman SA, Vinceti M, Dratch L, Arias JJ, Swidler J, Turner MR, Shefner J, Westeneng HJ, van den Berg LH, Al-Chalabi A. Guidance for clinical management of pathogenic variant carriers at elevated genetic risk for ALS/FTD. J Neurol Neurosurg Psychiatry 2025; 96:jnnp-2024-334339. [PMID: 39572211 PMCID: PMC12015018 DOI: 10.1136/jnnp-2024-334339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 08/19/2024] [Indexed: 02/02/2025]
Abstract
There is a growing understanding of the presymptomatic stages of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) and nascent efforts aiming to prevent these devastating neurodegenerative diseases have emerged. This progress is attributable, in no small part, to the altruism of people living with pathogenic variants at elevated genetic risk for ALS/FTD via their willingness to participate in natural history studies and disease prevention trials. Increasingly, this community has also highlighted the urgent need to develop paradigms for providing appropriate clinical care for those at elevated risk for ALS and FTD. This manuscript summarises recommendations emanating from a multi-stakeholder Workshop (Malvern, Pennsylvania, 2023) that aimed to develop guidance for at-risk carriers and their treating physicians. Clinical care recommendations span genetic testing (including counselling and sociolegal implications); monitoring for the emergence of early motor, cognitive and behavioural signs of disease; and the use of Food and Drug Administration-approved small molecule drugs and gene-targeting therapies. Lifestyle recommendations focus on exercise, smoking, statin use, supplement use, caffeine intake and head trauma, as well as occupational and environmental exposures. While the evidence base to inform clinical and lifestyle recommendations is limited, this guidance document aims to appraise carriers and clinicians of the issues and best available evidence, and also to define the research agenda that could yield more evidence-informed guidelines.
Collapse
Affiliation(s)
- Michael Benatar
- Neurology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Terry D Heiman-Patterson
- Department of Neurology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | | | - Daniel Brickman
- Genetic ALS & FTD: End the Legacy, Philadelphia, Pennsylvania, USA
| | - Kaitlin B Casaletto
- Department of Neurology, UCSF Memory and Aging Center, San Francisco, California, USA
| | - Stephen A Goutman
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Marco Vinceti
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Laynie Dratch
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jalayne J Arias
- Department of Health Policy & Behavioral Sciences, Georgia State University School of Public Health, Atlanta, Georgia, USA
| | - Jean Swidler
- Genetic ALS & FTD: End the Legacy, Philadelphia, Pennsylvania, USA
| | - Martin R Turner
- Nuffield Department of Clinical Neurosciences, Oxford University, Oxford, UK
| | - Jeremy Shefner
- Department of Neurology, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Henk-Jan Westeneng
- Department of Neurology, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, The Netherlands
| | - Leonard H van den Berg
- Department of Neurology, University Medical Centre Utrecht Brain Centre, Utrecht, The Netherlands
| | - Ammar Al-Chalabi
- Department of Basic and Clinical Neuroscience, King's College London, London, UK
| |
Collapse
|
8
|
Dykstra MM, Weskamp K, Gómez NB, Waksmacki J, Tank E, Glineburg MR, Snyder A, Pinarbasi E, Bekier M, Li X, Miller MR, Bai J, Shahzad S, Nedumaran N, Wieland C, Stewart C, Willey S, Grotewold N, McBride J, Moran JJ, Suryakumar AV, Lucas M, Tessier PM, Ward M, Todd PK, Barmada SJ. TDP43 autoregulation gives rise to dominant negative isoforms that are tightly controlled by transcriptional and post-translational mechanisms. Cell Rep 2025; 44:115113. [PMID: 39792557 PMCID: PMC11848802 DOI: 10.1016/j.celrep.2024.115113] [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: 07/09/2024] [Revised: 11/06/2024] [Accepted: 12/04/2024] [Indexed: 01/12/2025] Open
Abstract
The nuclear RNA-binding protein TDP43 is integrally involved in the pathogenesis of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). Previous studies uncovered N-terminal TDP43 isoforms that are predominantly cytosolic in localization, prone to aggregation, and enriched in susceptible spinal motor neurons. In healthy cells, however, these shortened (s)TDP43 isoforms are difficult to detect in comparison to full-length (fl)TDP43, raising questions regarding their origin and selective regulation. Here, we show that sTDP43 is created as a by-product of TDP43 autoregulation and cleared by nonsense-mediated RNA decay (NMD). sTDP43-encoding transcripts that escape NMD are rapidly degraded post-translationally via the proteasome and macroautophagy. Circumventing these regulatory mechanisms by overexpressing sTDP43 results in neurodegeneration via N-terminal oligomerization and impairment of flTDP43 splicing activity, in addition to RNA-binding-dependent gain-of-function toxicity. Collectively, these studies highlight endogenous mechanisms that tightly regulate sTDP43 expression and underscore the consequences of aberrant sTDP43 accumulation in disease.
Collapse
Affiliation(s)
- Megan M Dykstra
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Kaitlin Weskamp
- Chemistry Department, Nebraska Wesleyan University, Lincoln, NE, USA
| | - Nicolás B Gómez
- Graduate Program in Cell and Molecular Biology, University of Michigan, Ann Arbor, MI, USA; Medical Scientist Training Program, University of Michigan, Ann Arbor, MI, USA
| | - Jacob Waksmacki
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Elizabeth Tank
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - M Rebecca Glineburg
- Biological Sciences, Schmid College of Science and Technology, Chapman University, Orange, CA, USA
| | | | - Emile Pinarbasi
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA; Neuropathology, Department of Pathology, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Michael Bekier
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Xingli Li
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Morgan R Miller
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Jen Bai
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Shameena Shahzad
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Neha Nedumaran
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Clare Wieland
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA; Medical Scientist Training Program, University of Michigan, Ann Arbor, MI, USA
| | - Corey Stewart
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Sydney Willey
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Nikolas Grotewold
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI, USA
| | - Jonathon McBride
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
| | - John J Moran
- Atlanta Pediatric Research Alliance, Emory University, Atlanta, GA, USA
| | | | - Michael Lucas
- Departments of Chemical Engineering and Pharmaceutical Sciences, Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Peter M Tessier
- Departments of Chemical Engineering and Pharmaceutical Sciences, Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Michael Ward
- Neurogenetics Branch, NINDS, NIH, Bethesda, MD, USA
| | - Peter K Todd
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA; Graduate Program in Cell and Molecular Biology, University of Michigan, Ann Arbor, MI, USA; Medical Scientist Training Program, University of Michigan, Ann Arbor, MI, USA; Department of Neurology, University of Michigan, Ann Arbor, MI, USA; Veterans Affairs Medical Center, Ann Arbor, MI, USA
| | - Sami J Barmada
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA; Graduate Program in Cell and Molecular Biology, University of Michigan, Ann Arbor, MI, USA; Medical Scientist Training Program, University of Michigan, Ann Arbor, MI, USA; Department of Neurology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
9
|
Ghaffari LT, Welebob E, Boehringer A, Cyliax K, Pasinelli P, Trotti D, Haeusler AR. Neuronal Activity-Dependent Gene Dysregulation in C9orf72 i 3Neuronal Models of ALS/FTD Pathogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.27.632228. [PMID: 39975241 PMCID: PMC11838197 DOI: 10.1101/2025.01.27.632228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The GGGGCC nucleotide repeat expansion (NRE) mutation in the C9orf72 (C9) gene is the most common cause of ALS and FTD. Neuronal activity plays an essential role in shaping biological processes within both healthy and neurodegenerative disease scenarios. Here, we show that at baseline conditions, C9-NRE iPSC-cortical neurons display aberrations in several pathways, including synaptic signaling and transcriptional machinery, potentially priming diseased neurons for an altered response to neuronal stimulation. Indeed, exposure to two pathophysiologically relevant stimulation modes, prolonged membrane depolarization, or a blockade of K+ channels, followed by RNA sequencing, induces a temporally divergent activity-dependent transcriptome of C9-NRE cortical neurons compared to healthy controls. This study provides new insights into how neuronal activity influences the ALS/FTD-associated transcriptome, offering a dataset that enables further exploration of pathways necessary for conferring neuronal resilience or degeneration.
Collapse
Affiliation(s)
- Layla T. Ghaffari
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Emily Welebob
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Ashley Boehringer
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Kelly Cyliax
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Piera Pasinelli
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Davide Trotti
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Aaron R. Haeusler
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
10
|
Quintanilla CA, Fitzgerald Z, Kashow O, Radojicic MS, Ulupinar E, Bitlis D, Genc B, Andjus P, van Drongelen W, Ozdinler PH. High-density multielectrode arrays bring cellular resolution to neuronal activity and network analyses of corticospinal motor neurons. Sci Rep 2025; 15:732. [PMID: 39753665 PMCID: PMC11699118 DOI: 10.1038/s41598-024-83883-6] [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: 05/30/2024] [Accepted: 12/18/2024] [Indexed: 01/06/2025] Open
Abstract
Corticospinal motor neurons (CSMN), located in the motor cortex of the brain, are one of the key components of the motor neuron circuitry. They are in part responsible for the initiation and modulation of voluntary movement, and their degeneration is the hallmark for numerous diseases, such as amyotrophic lateral sclerosis (ALS), hereditary spastic paraplegia, and primary lateral sclerosis. Cortical hyperexcitation followed by in-excitability suggests the early involvement of cortical dysfunction in ALS pathology. However, a high-spatiotemporal resolution on our understanding of their functional health and connectivity is lacking. Here, we combine optical imaging with high-density microelectrode array (HD-MEA) system enabling single cell resolution and utilize UCHL1-eGFP mice to bring cell-type specificity to our understanding of the electrophysiological features of healthy CSMN, as they mature and form network connections with other cortical neurons, in vitro. This novel approach lays the foundation for future cell-type specific analyses of CSMN that are diseased due to different underlying causes with cellular precision, and it will allow the assessment of their functional response to compound treatment, especially for drug discovery efforts in upper motor neuron diseases.
Collapse
Affiliation(s)
- Christopher A Quintanilla
- Department of Neurology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Ave, Chicago, IL, 60611, USA
| | - Zachary Fitzgerald
- Department of Neurology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Ave, Chicago, IL, 60611, USA
| | - Omar Kashow
- Department of Neurology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Ave, Chicago, IL, 60611, USA
| | - Mihailo S Radojicic
- Institute for Physiology and Biochemistry "Jean Giaja", Faculty of Biology, University of Belgrade, Studentski trg 3, Belgrade, 11000, Serbia
| | - Emel Ulupinar
- Department of Neurology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Ave, Chicago, IL, 60611, USA
| | - Dila Bitlis
- Department of Neurology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Ave, Chicago, IL, 60611, USA
| | - Baris Genc
- Department of Neurology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Ave, Chicago, IL, 60611, USA
| | - Pavle Andjus
- Institute for Physiology and Biochemistry "Jean Giaja", Faculty of Biology, University of Belgrade, Studentski trg 3, Belgrade, 11000, Serbia
| | - Wim van Drongelen
- Pediatric Neurology, The University of Chicago, 5841 S Maryland Ave, Chicago, IL, 60637, USA
| | - P Hande Ozdinler
- Department of Neurology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Ave, Chicago, IL, 60611, USA.
- Les Turner ALS Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA.
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
11
|
Ziser L, van Eijk RPA, Kiernan MC, McRae A, Henderson RD, Schultz D, Needham M, Mathers S, McCombe P, Talman P, Vucic S. Amyotrophic lateral sclerosis established as a multistep process across phenotypes. Eur J Neurol 2025; 32:e16532. [PMID: 39475283 PMCID: PMC11622508 DOI: 10.1111/ene.16532] [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: 08/06/2024] [Revised: 09/21/2024] [Accepted: 10/07/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND AND PURPOSE Given the accepted multistep process of disease causation in amyotrophic lateral sclerosis (ALS), the present study was undertaken to determine the number of steps required for disease onset across each of the ALS phenotypes. METHODS Clinical and demographic data were prospectively accumulated using the Australian Motor Neurone Disease Registry (2005-2016), and age-specific incidence rates were calculated. Poisson regression was utilized to assess the relationship between log age-specific incidence and log age of onset, with McFadden's R2 used to assess the goodness of fit of the model. RESULTS In total, 2647 ALS patients were included, with mean disease-onset age being 62.2 ± 12.1 years. A linear relationship between log incidence and log age was established across ALS phenotypes, with variable slope estimates: bulbar 5.1 (95% confidence interval [CI] 4.6-5.6); cervical 2.7 (95% CI 2.3-3.0); lumbar 3.5 (95% CI 3.2-3.9); flail arm 4.7 (95% CI 3.9-5.5); flail leg 3.6 (95% CI 2.6-4.5); primary lateral sclerosis 2.7 (95% CI 1.8-3.7). Slope estimates were significantly higher in the bulbar compared to the cervical, lumbar and primary lateral sclerosis phenotypes. McFadden's R2 values were >0.4 for all phenotypes indicating excellent model fit. DISCUSSION A multistep process has been established across all ALS phenotypes with variable slope estimates, suggesting that the number of steps to develop disease is different across clinical presentations. Identification of mechanisms underlying slope estimate variability could exert pathophysiological significance.
Collapse
Affiliation(s)
- Laura Ziser
- Institute for Molecular Bioscience, University of QueenslandBrisbaneQueenslandAustralia
| | - Ruben P. A. van Eijk
- Department of Neurology, UMC Utrecht Brain CentreUniversity Medical Center UtrechtUtrechtThe Netherlands
- Biostatistics and Research Support, Julius Centre for Health Sciences and Primary CareUniversity Medical Center UtrechtUtrechtThe Netherlands
| | | | - Allan McRae
- Institute for Molecular Bioscience, University of QueenslandBrisbaneQueenslandAustralia
| | - Robert D. Henderson
- Department of NeurologyRoyal Brisbane and Women's HospitalBrisbaneQueenslandAustralia
| | - David Schultz
- Department of NeurologyFlinders University of South Australia, and Flinders Medical CentreBedford ParkSouth AustraliaAustralia
| | - Merrilee Needham
- Department of NeurologyFiona Stanley HospitalMurdochWestern AustraliaAustralia
- Centre for Molecular, MedicineMurdoch UniversityPerthWestern AustraliaAustralia
- Department of NeurologyPerron Institute for Neurological and Translational ScienceNedlandsWestern AustraliaAustralia
- Department of NeurologyUniversity of Notre DameFremantleWestern AustraliaAustralia
| | - Susan Mathers
- Department of NeurologyCalvary Health Care BethlehemMelbourneVictoriaAustralia
- School of Clinical Sciences, Monash UniversityMelbourneVictoriaAustralia
| | - Pam McCombe
- Department of NeurologyRoyal Brisbane and Women's HospitalBrisbaneQueenslandAustralia
| | - Paul Talman
- Deakin University, University Hospital GeelongGeelongVictoriaAustralia
| | - Steve Vucic
- Brain and Nerve Research CentreThe University of Sydney, Concord HospitalSydneyNew South WalesAustralia
| |
Collapse
|
12
|
Donaghy R, Pioro EP. Neurophysiologic Innovations in ALS: Enhancing Diagnosis, Monitoring, and Treatment Evaluation. Brain Sci 2024; 14:1251. [PMID: 39766450 PMCID: PMC11674262 DOI: 10.3390/brainsci14121251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive disease of both upper motor neurons (UMNs) and lower motor neurons (LMNs) leading invariably to decline in motor function. The clinical exam is foundational to the diagnosis of the disease, and ordinal severity scales are used to track its progression. However, the lack of objective biomarkers of disease classification and progression delay clinical trial enrollment, muddle inclusion criteria, and limit accurate assessment of drug efficacy. Ultimately, biomarker evidence of therapeutic target engagement will support, and perhaps supplant, more traditional clinical trial outcome measures. Electrophysiology tools including nerve conduction study and electromyography (EMG) have already been established as diagnostic biomarkers of LMN degeneration in ALS. Additional understanding of the motor manifestations of disease is provided by motor unit number estimation, electrical impedance myography, and single-fiber EMG techniques. Dysfunction of UMN and non-motor brain areas is being increasingly assessed with transcranial magnetic stimulation, high-density electroencephalography, and magnetoencephalography; less common autonomic and sensory nervous system dysfunction in ALS can also be characterized. Although most of these techniques are used to explore the underlying disease mechanisms of ALS in research settings, they have the potential on a broader scale to noninvasively identify disease subtypes, predict progression rates, and assess physiologic engagement of experimental therapies.
Collapse
Affiliation(s)
- Ryan Donaghy
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Erik P. Pioro
- Djavad Mowafaghian Centre for Brain Health, Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
13
|
Tirassa P, Rosso P, Fico E, Marenco M, Mallone F, Gharbiya M, Lambiase A, Severini C. Perspective role of Substance P in Amyotrophic Lateral Sclerosis: From neuronal vulnerability to neuroprotection. Neurosci Biobehav Rev 2024; 167:105914. [PMID: 39374680 DOI: 10.1016/j.neubiorev.2024.105914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/18/2024] [Accepted: 09/29/2024] [Indexed: 10/09/2024]
Abstract
The neuropeptide Substance P (SP) and its preferred Neurokinin1 Receptor (NK1R) are known to participate in the physiopathology of neurodegenerative diseases and mainly exert a neuroprotective role. In the present work, we have described the involvement of SP and NK1R in Amyotrophic Lateral Sclerosis (ALS). This was demonstrated by the detection of altered levels of SP in the brain, spinal cord and cerebrospinal fluid (CSF) of patients and preclinical models of ALS, and by its ability to inhibit excitotoxicity-induced neurodegeneration in ALS animal models. These data are supported by results indicating an excitatory effect of SP at the motor neuron (MN) level, which promotes locomotor activity. ALS patients are characterized by a differential susceptibility to MNs degeneration, since sphincters and extraocular muscles are classically spared. It is hypothesized that SP may play a role in the maintenance of the ocular system and the innervation of the pelvic floor by contributing directly or indirectly to the selective resistance of this subset of MNs.
Collapse
Affiliation(s)
- Paola Tirassa
- Institute of Biochemistry and Cell Biology, National Research Council (CNR), International Campus A. Buzzati-Traverso, Monterotondo Scalo, Rome 00015, Italy.
| | - Pamela Rosso
- Institute of Biochemistry and Cell Biology, National Research Council (CNR), International Campus A. Buzzati-Traverso, Monterotondo Scalo, Rome 00015, Italy.
| | - Elena Fico
- Institute of Biochemistry and Cell Biology, National Research Council (CNR), International Campus A. Buzzati-Traverso, Monterotondo Scalo, Rome 00015, Italy.
| | - Marco Marenco
- Department of Sense Organs, Sapienza University of Rome, Viale del Policlinico, 155, Rome 00161, Italy.
| | - Fabiana Mallone
- Department of Sense Organs, Sapienza University of Rome, Viale del Policlinico, 155, Rome 00161, Italy.
| | - Magda Gharbiya
- Department of Sense Organs, Sapienza University of Rome, Viale del Policlinico, 155, Rome 00161, Italy.
| | - Alessandro Lambiase
- Department of Sense Organs, Sapienza University of Rome, Viale del Policlinico, 155, Rome 00161, Italy.
| | - Cinzia Severini
- Institute of Biochemistry and Cell Biology, National Research Council (CNR), International Campus A. Buzzati-Traverso, Monterotondo Scalo, Rome 00015, Italy.
| |
Collapse
|
14
|
Lorenc F, Dupuis L, Cassel R. Impairments of inhibitory neurons in amyotrophic lateral sclerosis and frontotemporal dementia. Neurobiol Dis 2024; 203:106748. [PMID: 39592063 DOI: 10.1016/j.nbd.2024.106748] [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: 10/03/2024] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 11/28/2024] Open
Abstract
Amyotrophic lateral sclerosis and frontotemporal dementia are two fatal neurodegenerative disorders. They are part of a pathophysiological continuum, displaying clinical, neuropathological, and genetic overlaps. There is compelling evidence that neuronal circuit dysfunction is an early feature of both diseases. Impaired neuronal excitability, imbalanced excitatory and inhibitory influences, and altered functional connectivity have been reported. These phenomena are likely due to combined alterations in the various cellular components involved in the functioning of neuronal networks. This review focuses on one of these cellular components: inhibitory neurons. We assess the evidence for inhibitory neuron impairments in amyotrophic lateral sclerosis and frontotemporal dementia, as well as the mechanisms leading to the loss of inhibition. We also discuss the contributions of these alterations to symptoms, and the potential therapeutic strategies for targeting inhibitory neuron deficits.
Collapse
Affiliation(s)
- Félicie Lorenc
- Université de Strasbourg, INSERM, UMR-S 1329, Strasbourg Translational Neuroscience and Psychiatry, CRBS, Strasbourg, France.
| | - Luc Dupuis
- Université de Strasbourg, INSERM, UMR-S 1329, Strasbourg Translational Neuroscience and Psychiatry, CRBS, Strasbourg, France.
| | - Raphaelle Cassel
- Université de Strasbourg, INSERM, UMR-S 1329, Strasbourg Translational Neuroscience and Psychiatry, CRBS, Strasbourg, France.
| |
Collapse
|
15
|
Lopes M, Swash M, de Carvalho M. F-waves responses derived from low-intensity electrical stimulation: A method to explore split-hand pathogenesis. Neurophysiol Clin 2024; 54:103018. [PMID: 39488863 DOI: 10.1016/j.neucli.2024.103018] [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: 08/14/2024] [Revised: 10/05/2024] [Accepted: 10/07/2024] [Indexed: 11/05/2024] Open
Abstract
OBJECTIVES The "split-hand syndrome" is a common clinical sign in amyotrophic lateral sclerosis (ALS), being characterized by more severe atrophy of the hand muscles on the radial side of the hand compared to the ulnar side. We aimed to investigate possible physiological differences between relevant hand muscles using low-intensity F-wave stimulation to assess spinal motoneuron excitability. METHODS We recruited 36 healthy volunteers. F-waves were recorded from the abductor pollicis brevis (APB), first dorsal interosseous (FDI) and abductor digiti minimi (ADM), using 20 supramaximal stimuli followed by 20 stimuli at a low-intensity required to obtain M-waves with 10 % amplitude of maximal CMAP. We evaluated the following F-wave parameters: F-M latency, chronodispersion, persistence, amplitude, F/CMAP amplitude ratio and number of F-wave repeaters (with low-intensity). In 10 subjects, low-intensity stimulation F-waves were compared after 20 and 50 stimuli in each muscle. RESULTS Low-intensity stimulation resulted in lower F-wave amplitude and persistence and higher F/CMAP amplitude ratios. There were no significant differences in F-wave latencies and chronodispersion. When comparing the three muscles, we found higher F-wave persistence and F/CMAP amplitude ratios when recording over the ADM and APB compared to the FDI. We also found a higher number of F-wave repeaters in the ADM with low-intensity stimulation. Results from 20 to 50 low-intensity stimuli were similar. DISCUSSION A small number of low-intensity stimuli is appropriate to study F-wave latencies and chronodispersion. We found differences in some physiological properties of the ADM spinal motoneuron pool compared to other hand muscles.
Collapse
Affiliation(s)
- Miguel Lopes
- Institute of Physiology, Instituto de Medicina Molecular João Lobo Antunes, Centro de Estudos Egas Moniz, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Michael Swash
- Institute of Physiology, Instituto de Medicina Molecular João Lobo Antunes, Centro de Estudos Egas Moniz, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal; Departments of Neurology and Neuroscience, Barts and the London School of Medicine, Queen Mary University of London and Royal London Hospital, UK
| | - Mamede de Carvalho
- Institute of Physiology, Instituto de Medicina Molecular João Lobo Antunes, Centro de Estudos Egas Moniz, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal; Department of Neurosciences and Mental Health, Hospital de Santa Maria, Centro Hospitalar Universitário de Lisboa Norte, Lisbon, Portugal.
| |
Collapse
|
16
|
Theme 7 Pre-Clinical Therapeutic Strategies. Amyotroph Lateral Scler Frontotemporal Degener 2024; 25:197-217. [PMID: 39508670 DOI: 10.1080/21678421.2024.2403304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
|
17
|
Yang M, You D, Liu G, Lu Y, Yang G, O'Brien T, Henshall DC, Hardiman O, Cai L, Liu M, Shen S. Polyethyleneimine facilitates the growth and electrophysiological characterization of iPSC-derived motor neurons. Sci Rep 2024; 14:26106. [PMID: 39478194 PMCID: PMC11525838 DOI: 10.1038/s41598-024-77710-1] [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: 07/13/2024] [Accepted: 10/24/2024] [Indexed: 11/02/2024] Open
Abstract
Induced pluripotent stem cell (iPSC) technology, in combination with electrophysiological characterization via multielectrode array (MEA), has facilitated the utilization of iPSC-derived motor neurons (iPSC-MNs) as highly valuable models for underpinning pathogenic mechanisms and developing novel therapeutic interventions for motor neuron diseases (MNDs). However, the challenge of MN adherence to the MEA plate and the heterogeneity presented in iPSC-derived cultures raise concerns about the reproducibility of the findings obtained from these cellular models. We discovered that one novel factor modulating the electrophysiological activity of iPSC-MNs is the extracellular matrix (ECM) used in the coating to support in vitro growth, differentiation and maturation of iPSC-MNs. The current study showed that two coating conditions, namely, Poly-L-ornithine/Matrigel (POM) and Polyethyleneimine (PEI) strongly promoted attachment of iPSC-MNs on MEA culture dishes compared to three other coating conditions, and both facilitated the maturation of iPSC-MNs as characterized by the detection of extensive electrophysiological activities from the MEA plates. POM coating accelerated the maturation of the iPSC-MNs for up to 5 weeks, which suits modeling of neurodevelopmental disorders. However, the application of PEI resulted in more even distribution of the MNs on the culture dish and reduced variability of electrophysiological signals from the iPSC-MNs in 7-week cultures, which permitted the detection of enhanced excitability in iPSC-MNs from patients with amyotrophic lateral sclerosis (ALS). This study provides a comprehensive comparison of five coating conditions and offers POM and PEI as favorable coatings for in vitro modeling of neurodevelopmental and neurodegenerative disorders, respectively.
Collapse
Affiliation(s)
- Meimei Yang
- Key Laboratory of Measurement and Evaluation in Exercise Bioinformation of Hebei Province, School of Physical Education, Hebei Normal University, Shijiazhuang, 050024, China.
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, H91 W2TY, Ireland.
- FutureNeuro SFI Research Centre for Chronic and Rare Neurological Diseases and Department of Physiology and Medical Physics, RCSI University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland.
| | - Daofeng You
- Emergency Department, The First Hospital of Hebei Medical University, No. 89, Donggang Road, Shijiazhuang, China
| | - Gang Liu
- Department of Cardiology, Hebei Key Laboratory of Cardiac Injury Repair Mechanism Study; Hebei Key Laboratory of Heart and Metabolism, Hebei Engineering Research Center of Intelligent Medical Clinical Application, Hebei International Joint Research Center for Structural Heart Disease, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yin Lu
- College of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Guangming Yang
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
- Confucius Institute of Chinese and Regenerative Medicine, University of Galway, Galway, H91 W2TY, Ireland
| | - Timothy O'Brien
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, H91 W2TY, Ireland
- Confucius Institute of Chinese and Regenerative Medicine, University of Galway, Galway, H91 W2TY, Ireland
| | - David C Henshall
- FutureNeuro SFI Research Centre for Chronic and Rare Neurological Diseases and Department of Physiology and Medical Physics, RCSI University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
- Department of Physiology and Medical Physics, RCSI University of Medicine & Health Sciences, Dublin, D02 YN77, Ireland
| | - Orla Hardiman
- FutureNeuro SFI Research Centre for Chronic and Rare Neurological Diseases and Department of Physiology and Medical Physics, RCSI University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
- Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland
| | - Li Cai
- Department of Ophthalmology, Shenzhen University General Hospital, Xueyuan Road 1098, Shenzhen, 518000, China.
| | - Min Liu
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Molecular and Cellular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China.
| | - Sanbing Shen
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, H91 W2TY, Ireland.
- FutureNeuro SFI Research Centre for Chronic and Rare Neurological Diseases and Department of Physiology and Medical Physics, RCSI University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland.
- Confucius Institute of Chinese and Regenerative Medicine, University of Galway, Galway, H91 W2TY, Ireland.
| |
Collapse
|
18
|
Le NT, Chu N, Joshi G, Higgins NR, Nebie O, Adelakun N, Butts M, Monteiro MJ. Prion protein pathology in Ubiquilin 2 models of ALS. Neurobiol Dis 2024; 201:106674. [PMID: 39299489 DOI: 10.1016/j.nbd.2024.106674] [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: 06/10/2024] [Revised: 09/16/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024] Open
Abstract
Mutations in UBQLN2 cause ALS and frontotemporal dementia (FTD). The pathological signature in UBQLN2 cases is deposition of highly unusual types of inclusions in the brain and spinal cord that stain positive for UBQLN2. However, what role these inclusions play in pathogenesis remains unclear. Here we show cellular prion protein (PrPC) is found in UBQLN2 inclusions in both mouse and human neuronal induced pluripotent (IPSC) models of UBQLN2 mutations, evidenced by the presence of aggregated forms of PrPC with UBQLN2 inclusions. Turnover studies indicated that the P497H UBQLN2 mutation slows PrPC protein degradation and leads to mislocalization of PrPC in the cytoplasm. Immunoprecipitation studies indicated UBQLN2 and PrPC bind together in a complex. The abnormalities in PrPC caused by UBQLN2 mutations may be relevant in disease pathogenesis.
Collapse
Affiliation(s)
- Nhat T Le
- Department of Cancer Biology and Genetics, Ohio State University College of Medicine, Columbus, OH, United States of America
| | - Nam Chu
- Department of Cancer Biology and Genetics, Ohio State University College of Medicine, Columbus, OH, United States of America
| | - Gunjan Joshi
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, United States of America; Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Nicole R Higgins
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, United States of America; Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Ouada Nebie
- Department of Cancer Biology and Genetics, Ohio State University College of Medicine, Columbus, OH, United States of America
| | - Niyi Adelakun
- Department of Cancer Biology and Genetics, Ohio State University College of Medicine, Columbus, OH, United States of America
| | - Marie Butts
- Department of Cancer Biology and Genetics, Ohio State University College of Medicine, Columbus, OH, United States of America
| | - Mervyn J Monteiro
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, United States of America; Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| |
Collapse
|
19
|
Mazurie Z, Branchereau P, Cattaert D, Henkous N, Savona-Baron C, Vouimba RM. Acute stress differently modulates interneurons excitability and synaptic plasticity in the primary motor cortex of wild-type and SOD1 G93A mouse model of ALS. J Physiol 2024; 602:4987-5015. [PMID: 39216080 DOI: 10.1113/jp285210] [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: 06/28/2023] [Accepted: 07/12/2024] [Indexed: 09/04/2024] Open
Abstract
Primary motor cortex (M1) network stability depends on activity of inhibitory interneurons, for which susceptibility to stress was previously demonstrated in limbic regions. Hyperexcitability in M1 following changes in the excitatory/inhibitory balance is a key pathological hallmark of amyotrophic lateral sclerosis (ALS). Using electrophysiological approaches, we assessed the impact of acute restraint stress on inhibitory interneurons excitability and global synaptic plasticity in M1 of the SOD1G93A ALS mouse model at a late pre-symptomatic stage (10-12.5 weeks). Based on their firing type (continuous, discontinuous, with accommodation or not) and electrophysiological characteristics (resting potential, rheobase, firing frequency), interneurons from M1 slices were separated into four clusters, labelled from 1 to 4. Among them, only interneurons from the first cluster, presenting continuous firing with few accommodations, tended to show increased excitability in wild-type (WT) and decreased excitability in SOD1G93A animals following stress. In vivo analyses of evoked field potentials showed that stress suppressed the theta burst-induced plasticity of an excitatory component (N1) recorded in the superficial layers of M1 in WT, with no impact on an inhibitory complex (N2-P1) from the deeper layers. In SOD1G93A mice, stress did not affect N1 but suppressed the N2-P1 plasticity. These data suggest that stress can alter M1 network functioning in a different manner in WT and SOD1G93A mice, possibly through changes of inhibitory interneurons excitability and synaptic plasticity. This suggests that stress-induced activity changes in M1 may therefore influence ALS outcomes. KEY POINTS: Disruption of the excitatory/inhibitory balance in the primary motor cortex (M1) has been linked to cortical hyperexcitability development, a key pathological hallmark of amyotrophic lateral sclerosis (ALS). Psychological stress was reported to influence excitatory/inhibitory balance in limbic regions, but very little is known about its influence on the M1 functioning under physiological or pathological conditions. Our study revealed that acute stress influences the excitatory/inhibitory balance within the M1, through changes in interneurons excitability along with network plasticity. Such changes were different in pathological (SOD1G93A ALS mouse model) vs. physiological (wild-type) conditions. The results of our study help us to better understand how stress modulates the M1 and highlight the need to further characterize stress-induced motor cortex changes because it may be of importance when evaluating ALS outcomes.
Collapse
Affiliation(s)
- Zoé Mazurie
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), CNRS, UMR 5287, University of Bordeaux, Bordeaux, France
| | - Pascal Branchereau
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), CNRS, UMR 5287, University of Bordeaux, Bordeaux, France
| | - Daniel Cattaert
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), CNRS, UMR 5287, University of Bordeaux, Bordeaux, France
| | - Nadia Henkous
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), CNRS, UMR 5287, University of Bordeaux, Bordeaux, France
| | - Catherine Savona-Baron
- Present address: BoRdeaux Institute of onCology (BRIC), INSERM U1312, University of Bordeaux, Bordeaux, France
| | - Rose-Marie Vouimba
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), CNRS, UMR 5287, University of Bordeaux, Bordeaux, France
| |
Collapse
|
20
|
Tu S, Vucic S, Kiernan MC. Pathological insights derived from neuroimaging in amyotrophic lateral sclerosis: emerging clinical applications. Curr Opin Neurol 2024; 37:577-584. [PMID: 38958573 DOI: 10.1097/wco.0000000000001295] [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: 07/04/2024]
Abstract
PURPOSE OF REVIEW Neuroimaging has been instrumental in shaping current understanding of the pathoanatomical signature of amyotrophic lateral sclerosis (ALS) across clinically well defined patient cohorts. The potential utility of imaging as an objective disease marker, however, remains poorly defined. RECENT FINDINGS Increasingly advanced quantitative and computational imaging studies have highlighted emerging clinical applications for neuroimaging as a complementary clinical modality for diagnosis, monitoring, and modelling disease propagation. Multimodal neuroimaging has demonstrated novel approaches for capturing primary motor disease. Extra-motor subcortical dysfunction is increasingly recognized as key modulators of disease propagation. SUMMARY The neural signature of cortical and subcortical dysfunction in ALS has been well defined at the population level. Objective metrics of focal primary motor dysfunction are increasingly sensitive and translatable to the individual patient level. Integrity of extra-motor subcortical abnormalities are recognized to represent critical pathways of the ALS disease 'connectome', predicting pathological spread. Neuroimaging plays a pivotal role in capturing upper motor neuron pathology in ALS. Their potential clinical role as objective disease markers for disease classification, longitudinal monitoring, and prognosis in ALS have become increasingly well defined.
Collapse
Affiliation(s)
- Sicong Tu
- Brain and Mind Centre
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney
- Neuroscience Research Australia
| | - Steve Vucic
- Brain and Nerve Research Center, The University of Sydney, Sydney, New South Wales, Australia
| | - Matthew C Kiernan
- Brain and Mind Centre
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney
- Neuroscience Research Australia
| |
Collapse
|
21
|
Tu S, Li T, Carroll AS, Mahoney CJ, Huynh W, Park SB, Henderson R, Vucic S, Kiernan MC, Lin CSY. Central neurodegeneration in Kennedy's disease accompanies peripheral motor dysfunction. Sci Rep 2024; 14:18331. [PMID: 39112530 PMCID: PMC11306389 DOI: 10.1038/s41598-024-69393-5] [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: 11/23/2023] [Accepted: 08/05/2024] [Indexed: 08/10/2024] Open
Abstract
Spinal and bulbar muscular atrophy (SBMA), or Kennedy's disease (KD), is a rare hereditary neuromuscular disorder demonstrating commonalities with amyotrophic lateral sclerosis (ALS). The current study aimed to define functional and central nervous system abnormalities associated with SBMA pathology, their interaction, and to identify novel clinical markers for quantifying disease activity. 27 study participants (12 SBMA; 8 ALS; 7 Control) were recruited. SBMA patients underwent comprehensive motor and sensory functional assessments, and neurophysiological testing. All participants underwent whole-brain structural and diffusion MRI. SBMA patients demonstrated marked peripheral motor and sensory abnormalities across clinical assessments. Increased abnormalities on neurological examination were significantly associated with increased disease duration in SBMA patients (R2 = 0.85, p < 0.01). Widespread juxtacortical axonal degeneration of corticospinal white matter tracts were detected in SBMA patients (premotor; motor; somatosensory; p < 0.05), relative to controls. Increased axial diffusivity was significantly correlated with total neuropathy score in SBMA patients across left premotor (R2 = 0.59, p < 0.01), motor (R2 = 0.63, p < 0.01), and somatosensory (R2 = 0.61, p < 0.01) tracts. The present series has identified involvement of motor and sensory brain regions in SBMA, associated with disease duration and increasing severity of peripheral neuropathy. Quantification of annualized brain MRI together with Total Neuropathy Score may represent a novel approach for clinical monitoring.
Collapse
Affiliation(s)
- Sicong Tu
- Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, 2050, Australia.
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, 2050, Australia.
| | - Tiffany Li
- Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, 2050, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, 2050, Australia
| | - Antonia S Carroll
- Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, 2050, Australia
- Department of Neurology and Neurophysiology, St Vincent's Hospital, Sydney, 2010, Australia
| | - Colin J Mahoney
- Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, 2050, Australia
| | - William Huynh
- Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, 2050, Australia
- Prince of Wales Clinical School, The University of New South Wales, Sydney, 2052, Australia
| | - Susanna B Park
- Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, 2050, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, 2050, Australia
| | - Robert Henderson
- Royal Brisbane and Women's Hospital, University of Queensland, Brisbane, 4029, Australia
| | - Steve Vucic
- Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, 2050, Australia
- Brain and Nerve Research Centre, Concord Clinical School, The University of Sydney, Sydney, 2137, Australia
| | - Matthew C Kiernan
- Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, 2050, Australia
- Department of Neurology, Royal Prince Alfred Hospital, Sydney, 2050, Australia
| | - Cindy S-Y Lin
- Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, 2050, Australia.
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, 2050, Australia.
| |
Collapse
|
22
|
Pavey NA, Menon P, Peterchev AV, Kiernan MC, Vucic S. Abnormalities of cortical stimulation strength-duration time constant in amyotrophic lateral sclerosis. Clin Neurophysiol 2024; 164:161-167. [PMID: 38901111 PMCID: PMC11345808 DOI: 10.1016/j.clinph.2024.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 04/23/2024] [Accepted: 05/26/2024] [Indexed: 06/22/2024]
Abstract
OBJECTIVES Strength-duration time constant (SDTC) may now be determined for cortical motor neurones, with activity mediated by transient Na+ conductances. The present study determined whether cortical SDTC is abnormal and linked to the pathogenesis of amyotrophic lateral sclerosis. METHODS Cortical SDTC and rheobase were estimated from 17 ALS patients using a controllable pulse parameter transcranial magnetic stimulation (cTMS) device. Resting motor thresholds (RMTs) were determined at pulse widths (PW) of 30, 45, 60, 90 and 120 µs and M-ratio of 0.1, using a figure-of-eight coil applied to the primary motor cortex. RESULTS SDTC was significantly reduced in ALS patients (150.58 ± 9.98 µs; controls 205.94 ± 13.7 µs, P < 0.01). The reduced SDTC correlated with a rate of disease progression (Rho = -0.440, P < 0.05), ALS functional rating score (ALSFRS-R) score (Rho = 0.446, P < 0.05), and disease duration (R = 0.428, P < 0.05). The degree of change in SDTC was greater in patients with cognitive abnormalities as manifested by an abnormal total Edinburgh Cognitive ALS Screen score (140.5 ± 28.7 µs, P < 0.001) and ALS-specific subscore (141.7 ± 33.2 µs, P = 0.003). CONCLUSIONS Cortical SDTC reduction was associated with a more aggressive ALS phenotype, or with more prominent cognitive impairment. SIGNIFICANCE An increase in transient Na+ conductances may account for the reduction in SDTC, linked to the pathogenesis of ALS.
Collapse
Affiliation(s)
- Nathan A Pavey
- Brain and Nerve Research Centre, Concord Clinical School, University of Sydney, Concord Hospital, Sydney, Australia
| | - Parvathi Menon
- Brain and Nerve Research Centre, Concord Clinical School, University of Sydney, Concord Hospital, Sydney, Australia
| | - Angel V Peterchev
- Department of Biological Sciences, Dartmouth College, Hanover, NH, USA; Department of Biomedical Engineering, Duke University, Durham, NC, USA; Department of Psychiatry and Behavioural Sciences, Duke University, Durham, NC, USA; Department of Electrical and Computer Engineering, Duke University, Durham, NC, USA; Department of Neurosurgery, Duke University, Durham, NC, USA
| | | | - Steve Vucic
- Brain and Nerve Research Centre, Concord Clinical School, University of Sydney, Concord Hospital, Sydney, Australia.
| |
Collapse
|
23
|
Calma AD, van den Bos M, Pavey N, Santos Silva C, Menon P, Vucic S. Physiological Biomarkers of Upper Motor Neuron Dysfunction in ALS. Brain Sci 2024; 14:760. [PMID: 39199454 PMCID: PMC11352893 DOI: 10.3390/brainsci14080760] [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: 06/03/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 09/01/2024] Open
Abstract
Upper motor neuron (UMN) dysfunction is an important feature of amyotrophic lateral sclerosis (ALS) for the diagnosis and understanding of pathogenesis. The identification of UMN signs forms the basis of ALS diagnosis, although may be difficult to discern, especially in the setting of severe muscle weakness. Transcranial magnetic stimulation (TMS) techniques have yielded objective physiological biomarkers of UMN dysfunction in ALS, enabling the interrogation of cortical and subcortical neuronal networks with diagnostic, pathophysiological, and prognostic implications. Transcranial magnetic stimulation techniques have provided pertinent pathogenic insights and yielded novel diagnostic and prognostic biomarkers. Cortical hyperexcitability, as heralded by a reduction in short interval intracortical inhibition (SICI) and an increase in short interval intracortical facilitation (SICF), has been associated with lower motor neuron degeneration, patterns of disease evolution, as well as the development of specific ALS clinical features including the split hand phenomenon. Reduction in SICI has also emerged as a potential diagnostic aid in ALS. More recently, physiological distinct inhibitory and facilitatory cortical interneuronal circuits have been identified, which have been shown to contribute to ALS pathogenesis. The triple stimulation technique (TST) was shown to enhance the diagnostic utility of conventional TMS measures in detecting UMN dysfunction. Resting-state EEG is a novel neurophysiological technique developed for directly interrogating cortical neuronal networks in ALS, that have yielded potentially useful physiological biomarkers of UMN dysfunction. The present review discusses physiological biomarkers of UMN dysfunction in ALS, encompassing conventional and novel TMS techniques developed to interrogate the functional integrity of the corticomotoneuronal system, focusing on pathogenic, diagnostic, and prognostic utility.
Collapse
Affiliation(s)
- Aicee Dawn Calma
- Brain and Nerve Research Center, The University of Sydney, Sydney 2139, Australia (C.S.S.)
| | - Mehdi van den Bos
- Brain and Nerve Research Center, The University of Sydney, Sydney 2139, Australia (C.S.S.)
| | - Nathan Pavey
- Brain and Nerve Research Center, The University of Sydney, Sydney 2139, Australia (C.S.S.)
| | - Cláudia Santos Silva
- Brain and Nerve Research Center, The University of Sydney, Sydney 2139, Australia (C.S.S.)
- Department of Neurosciences and Mental Health, Unidade Local de Saúde de Santa Maria, 1649-028 Lisbon, Portugal
- Faculdade de Medicina-Instituto de Medicina Molecular, Centro de Estudos Egas Moniz, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Parvathi Menon
- Brain and Nerve Research Center, The University of Sydney, Sydney 2139, Australia (C.S.S.)
| | - Steve Vucic
- Brain and Nerve Research Center, The University of Sydney, Sydney 2139, Australia (C.S.S.)
| |
Collapse
|
24
|
Otani R, Shibuya K, Suzuki YI, Suichi T, Morooka M, Aotsuka Y, Ogushi M, Kuwabara S. Effects of motor cortical and peripheral axonal hyperexcitability on survival in amyotrophic lateral sclerosis. J Neurol Neurosurg Psychiatry 2024; 95:730-736. [PMID: 38418214 DOI: 10.1136/jnnp-2023-333039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 02/13/2024] [Indexed: 03/01/2024]
Abstract
BACKGROUND Increased 'cortical' and 'peripheral' excitability are reportedly associated with shorter survival in amyotrophic lateral sclerosis (ALS) patients, suggesting that hyperexcitability contributes to motor neuron death. However, whether upper or lower motor function has a greater impact on survival is unclear. We aimed to investigate the component that strongly impacts the prognosis of ALS. METHODS A total of 103 consecutive patients with ALS who underwent cortical (threshold tracking transcranial magnetic stimulation (TMS)) and motor nerve excitability tests were included. Motor cortical excitability was evaluated using short-interval intracortical inhibition (SICI) during TMS. Motor axonal excitability was assessed using the strength-duration time constant (SDTC). Survival time was defined as the time from examination to death or tracheostomy. RESULTS Compared with healthy subjects, patients with ALS had lower SICI and longer SDTC (p<0.05), indicating increased excitability of cortical motor neurons and motor axons. According to the SICI and SDTC findings, patients were divided into the following four groups: 'cortical high and peripheral high (high-high)', 'high-low', 'low-high' and 'low-low' groups. In Kaplan-Meier curves, the 'high-high' and 'low-high' groups showed significantly shorter survival than the other groups. Multivariate analysis revealed that increased cortical (HR=5.3, p<0.05) and peripheral (HR=20.0, p<0.001) excitability were significantly associated with shorter survival. CONCLUSIONS In patients with ALS, both motor cortical and peripheral hyperexcitability independently affected survival time, with peripheral hyperexcitability having a greater impact on shorter survival. The modulation of neuronal/axonal excitability is a potential therapeutic target for ALS.
Collapse
Affiliation(s)
- Ryo Otani
- Neurology, Chiba University Graduate School of Medicine School of Medicine, Chiba, Japan
| | - Kazumoto Shibuya
- Neurology, Chiba University Graduate School of Medicine School of Medicine, Chiba, Japan
| | - Yo-Ichi Suzuki
- Neurology, Chiba University Graduate School of Medicine School of Medicine, Chiba, Japan
| | - Tomoki Suichi
- Neurology, Chiba University Graduate School of Medicine School of Medicine, Chiba, Japan
| | - Marie Morooka
- Neurology, Chiba University Graduate School of Medicine School of Medicine, Chiba, Japan
| | - Yuya Aotsuka
- Neurology, Chiba University Graduate School of Medicine School of Medicine, Chiba, Japan
| | - Moeko Ogushi
- Neurology, Chiba University Graduate School of Medicine School of Medicine, Chiba, Japan
| | - Satoshi Kuwabara
- Neurology, Chiba University Graduate School of Medicine School of Medicine, Chiba, Japan
| |
Collapse
|
25
|
Pavey N, Hannaford A, van den Bos M, Kiernan MC, Menon P, Vucic S. Distinct neuronal circuits mediate cortical hyperexcitability in amyotrophic lateral sclerosis. Brain 2024; 147:2344-2356. [PMID: 38374770 DOI: 10.1093/brain/awae049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/16/2024] [Accepted: 01/27/2024] [Indexed: 02/21/2024] Open
Abstract
Cortical hyperexcitability is an important pathophysiological mechanism in amyotrophic lateral sclerosis (ALS), reflecting a complex interaction of inhibitory and facilitatory interneuronal processes that evolves in the degenerating brain. The advances in physiological techniques have made it possible to interrogate progressive changes in the motor cortex. Specifically, the direction of transcranial magnetic stimulation (TMS) stimulus within the primary motor cortex can be utilized to influence descending corticospinal volleys and to thereby provide information about distinct interneuronal circuits. Cortical motor function and cognition was assessed in 29 ALS patients with results compared to healthy volunteers. Cortical dysfunction was assessed using threshold-tracking TMS to explore alterations in short interval intracortical inhibition (SICI), short interval intracortical facilitation (SICF), the index of excitation and stimulus response curves using a figure-of-eight coil with the coil oriented relative to the primary motor cortex in a posterior-anterior, lateral-medial and anterior-posterior direction. Mean SICI, between interstimulus interval of 1-7 ms, was significantly reduced in ALS patients compared to healthy controls when assessed with the coil oriented in posterior-anterior (P = 0.044) and lateral-medial (P = 0.005) but not the anterior-posterior (P = 0.08) directions. A significant correlation between mean SICI oriented in a posterior-anterior direction and the total Edinburgh Cognitive and Behavioural ALS Screen score (Rho = 0.389, P = 0.037) was evident. In addition, the mean SICF, between interstimulus interval 1-5 ms, was significantly increased in ALS patients when recorded with TMS coil oriented in posterior-anterior (P = 0.035) and lateral-medial (P < 0.001) directions. In contrast, SICF recorded with TMS coil oriented in the anterior-posterior direction was comparable between ALS and controls (P = 0.482). The index of excitation was significantly increased in ALS patients when recorded with the TMS coil oriented in posterior-anterior (P = 0.041) and lateral-medial (P = 0.003) directions. In ALS patients, a significant increase in the stimulus response curve gradient was evident compared to controls when recorded with TMS coil oriented in posterior-anterior (P < 0.001), lateral-medial (P < 0.001) and anterior-posterior (P = 0.002) directions. The present study has established that dysfunction of distinct interneuronal circuits mediates the development of cortical hyperexcitability in ALS. Specifically, complex interplay between inhibitory circuits and facilitatory interneuronal populations, that are preferentially activated by stimulation in posterior-to-anterior or lateral-to-medial directions, promotes cortical hyperexcitability in ALS. Mechanisms that underlie dysfunction of these specific cortical neuronal circuits will enhance understanding of the pathophysiological processes in ALS, with the potential to uncover focussed therapeutic targets.
Collapse
Affiliation(s)
- Nathan Pavey
- Brain and Nerve Research Centre, Concord Clinical School, The University of Sydney, Concord Hospital, Sydney, NSW 2139, Australia
| | - Andrew Hannaford
- Brain and Nerve Research Centre, Concord Clinical School, The University of Sydney, Concord Hospital, Sydney, NSW 2139, Australia
| | - Mehdi van den Bos
- Brain and Nerve Research Centre, Concord Clinical School, The University of Sydney, Concord Hospital, Sydney, NSW 2139, Australia
| | - Matthew C Kiernan
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2139, Australia
- Department of Neurology, Royal Prince Alfred Hospital, Sydney, NSW 2139, Australia
| | - Parvathi Menon
- Brain and Nerve Research Centre, Concord Clinical School, The University of Sydney, Concord Hospital, Sydney, NSW 2139, Australia
| | - Steve Vucic
- Brain and Nerve Research Centre, Concord Clinical School, The University of Sydney, Concord Hospital, Sydney, NSW 2139, Australia
| |
Collapse
|
26
|
Dykstra MM, Weskamp K, Gómez NB, Waksmacki J, Tank E, Glineburg MR, Snyder A, Pinarbasi E, Bekier M, Li X, Bai J, Shahzad S, Nedumaran J, Wieland C, Stewart C, Willey S, Grotewold N, McBride J, Moran JJ, Suryakumar AV, Lucas M, Tessier P, Ward M, Todd P, Barmada SJ. TDP43 autoregulation gives rise to shortened isoforms that are tightly controlled by both transcriptional and post-translational mechanisms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.02.601776. [PMID: 39005384 PMCID: PMC11244999 DOI: 10.1101/2024.07.02.601776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
The nuclear RNA-binding protein TDP43 is integrally involved in the pathogenesis of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). Previous studies uncovered N-terminal TDP43 isoforms that are predominantly cytosolic in localization, highly prone to aggregation, and enriched in susceptible spinal motor neurons. In healthy cells, however, these shortened (s)TDP43 isoforms are difficult to detect in comparison to full-length (fl)TDP43, raising questions regarding their origin and selective regulation. Here, we show that sTDP43 is created as a byproduct of TDP43 autoregulation and cleared by nonsense mediated RNA decay (NMD). The sTDP43-encoding transcripts that escape NMD can lead to toxicity but are rapidly degraded post-translationally. Circumventing these regulatory mechanisms by overexpressing sTDP43 results in neurodegeneration in vitro and in vivo via N-terminal oligomerization and impairment of flTDP43 splicing activity, in addition to RNA binding-dependent gain-of-function toxicity. Collectively, these studies highlight endogenous mechanisms that tightly regulate sTDP43 expression and provide insight into the consequences of aberrant sTDP43 accumulation in disease.
Collapse
Affiliation(s)
- Megan M. Dykstra
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI
| | - Kaitlin Weskamp
- Chemistry Department, Nebraska Wesleyan University, Lincoln, NE
| | - Nicolás B. Gómez
- Graduate Program in Cell and Molecular Biology, University of Michigan, Ann Arbor, MI
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI
| | - Jacob Waksmacki
- Department of Neurology, University of Michigan, Ann Arbor, MI
| | - Elizabeth Tank
- Department of Neurology, University of Michigan, Ann Arbor, MI
| | - M. Rebecca Glineburg
- Biological Sciences, Schmid College of Science and Technology, Chapman University, Orange, CA
| | | | - Emile Pinarbasi
- Department of Neurology, University of Michigan, Ann Arbor, MI
- Neuropathology, Department of Pathology, Michigan Medicine, University of Michigan, Ann Arbor, MI
| | - Michael Bekier
- Department of Neurology, University of Michigan, Ann Arbor, MI
| | - Xingli Li
- Department of Neurology, University of Michigan, Ann Arbor, MI
| | - Jen Bai
- Department of Neurology, University of Michigan, Ann Arbor, MI
| | | | - Juno Nedumaran
- Department of Neurology, University of Michigan, Ann Arbor, MI
| | - Clare Wieland
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI
| | - Corey Stewart
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI
| | - Sydney Willey
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI
| | - Nikolas Grotewold
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI
| | - Jonathon McBride
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI
| | - John J. Moran
- Atlanta Pediatric Research Alliance, Emory University, Atlanta, GA
| | | | - Michael Lucas
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI
| | - Peter Tessier
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI
| | | | - Peter Todd
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI
- Graduate Program in Cell and Molecular Biology, University of Michigan, Ann Arbor, MI
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI
- Department of Neurology, University of Michigan, Ann Arbor, MI
- Veterans Affairs Medical Center, Ann Arbor, MI
| | - Sami J. Barmada
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI
- Graduate Program in Cell and Molecular Biology, University of Michigan, Ann Arbor, MI
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI
- Department of Neurology, University of Michigan, Ann Arbor, MI
| |
Collapse
|
27
|
Dharmadasa T, Pavey N, Tu S, Menon P, Huynh W, Mahoney CJ, Timmins HC, Higashihara M, van den Bos M, Shibuya K, Kuwabara S, Grosskreutz J, Kiernan MC, Vucic S. Novel approaches to assessing upper motor neuron dysfunction in motor neuron disease/amyotrophic lateral sclerosis: IFCN handbook chapter. Clin Neurophysiol 2024; 163:68-89. [PMID: 38705104 DOI: 10.1016/j.clinph.2024.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 02/08/2024] [Accepted: 04/14/2024] [Indexed: 05/07/2024]
Abstract
Identifying upper motor neuron (UMN) dysfunction is fundamental to the diagnosis and understanding of disease pathogenesis in motor neuron disease (MND). The clinical assessment of UMN dysfunction may be difficult, particularly in the setting of severe muscle weakness. From a physiological perspective, transcranial magnetic stimulation (TMS) techniques provide objective biomarkers of UMN dysfunction in MND and may also be useful to interrogate cortical and network function. Single, paired- and triple pulse TMS techniques have yielded novel diagnostic and prognostic biomarkers in MND, and have provided important pathogenic insights, particularly pertaining to site of disease onset. Cortical hyperexcitability, as heralded by reduced short interval intracortical inhibition (SICI) and increased short interval intracortical facilitation, has been associated with the onset of lower motor neuron degeneration, along with patterns of disease spread, development of specific clinical features such as the split hand phenomenon, and may provide an indication about the rate of disease progression. Additionally, reduction of SICI has emerged as a potential diagnostic aid in MND. The triple stimulation technique (TST) was shown to enhance the diagnostic utility of conventional TMS measures in detecting UMN dysfunction in MND. Separately, sophisticated brain imaging techniques have uncovered novel biomarkers of neurodegeneration that have bene associated with progression. The present review will discuss the utility of TMS and brain neuroimaging derived biomarkers of UMN dysfunction in MND, focusing on recently developed TMS techniques and advanced neuroimaging modalities that interrogate structural and functional integrity of the corticomotoneuronal system, with an emphasis on pathogenic, diagnostic, and prognostic utility.
Collapse
Affiliation(s)
- Thanuja Dharmadasa
- Department of Neurology, The Royal Melbourne Hospital City Campus, Parkville, Victoria, Australia
| | - Nathan Pavey
- Brain and Nerve Research Center, The University of Sydney, Sydney, Australia
| | - Sicong Tu
- Brain and Mind Centre, The University of Sydney, and Department of Neurology, Royal Prince Alfred Hospital, Australia
| | - Parvathi Menon
- Brain and Nerve Research Center, The University of Sydney, Sydney, Australia
| | - William Huynh
- Brain and Mind Centre, The University of Sydney, and Department of Neurology, Royal Prince Alfred Hospital, Australia
| | - Colin J Mahoney
- Brain and Mind Centre, The University of Sydney, and Department of Neurology, Royal Prince Alfred Hospital, Australia
| | - Hannah C Timmins
- Brain and Mind Centre, The University of Sydney, and Department of Neurology, Royal Prince Alfred Hospital, Australia
| | - Mana Higashihara
- Department of Neurology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Mehdi van den Bos
- Brain and Nerve Research Center, The University of Sydney, Sydney, Australia
| | - Kazumoto Shibuya
- Neurology, Chiba University, Graduate School of Medicine, Chiba, Japan
| | - Satoshi Kuwabara
- Neurology, Chiba University, Graduate School of Medicine, Chiba, Japan
| | - Julian Grosskreutz
- Precision Neurology, Excellence Cluster Precision Medicine in Inflammation, University of Lübeck, University Hospital Schleswig-Holstein Campus, Lübeck, Germany
| | - Matthew C Kiernan
- Brain and Mind Centre, The University of Sydney, and Department of Neurology, Royal Prince Alfred Hospital, Australia
| | - Steve Vucic
- Brain and Nerve Research Center, The University of Sydney, Sydney, Australia.
| |
Collapse
|
28
|
Koopman M, Güngördü L, Janssen L, Seinstra RI, Richmond JE, Okerlund N, Wardenaar R, Islam P, Hogewerf W, Brown AEX, Jorgensen EM, Nollen EAA. Rebalancing the motor circuit restores movement in a Caenorhabditis elegans model for TDP-43 toxicity. Cell Rep 2024; 43:114204. [PMID: 38748878 DOI: 10.1016/j.celrep.2024.114204] [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: 07/04/2023] [Revised: 02/29/2024] [Accepted: 04/23/2024] [Indexed: 06/01/2024] Open
Abstract
Amyotrophic lateral sclerosis can be caused by abnormal accumulation of TAR DNA-binding protein 43 (TDP-43) in the cytoplasm of neurons. Here, we use a C. elegans model for TDP-43-induced toxicity to identify the biological mechanisms that lead to disease-related phenotypes. By applying deep behavioral phenotyping and subsequent dissection of the neuromuscular circuit, we show that TDP-43 worms have profound defects in GABA neurons. Moreover, acetylcholine neurons appear functionally silenced. Enhancing functional output of repressed acetylcholine neurons at the level of, among others, G-protein-coupled receptors restores neurotransmission, but inefficiently rescues locomotion. Rebalancing the excitatory-to-inhibitory ratio in the neuromuscular system by simultaneous stimulation of the affected GABA- and acetylcholine neurons, however, not only synergizes the effects of boosting individual neurotransmitter systems, but instantaneously improves movement. Our results suggest that interventions accounting for the altered connectome may be more efficient in restoring motor function than those solely focusing on diseased neuron populations.
Collapse
Affiliation(s)
- Mandy Koopman
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Lale Güngördü
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Leen Janssen
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Renée I Seinstra
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Janet E Richmond
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Nathan Okerlund
- Howard Hughes Medical Institute and School of Biological Science, The University of Utah, Salt Lake City, UT, USA
| | - René Wardenaar
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Priota Islam
- MRC London Institute of Medical Sciences, London, UK; Institute of Clinical Sciences, Imperial College London, London, UK
| | - Wytse Hogewerf
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Andre E X Brown
- MRC London Institute of Medical Sciences, London, UK; Institute of Clinical Sciences, Imperial College London, London, UK
| | - Erik M Jorgensen
- Howard Hughes Medical Institute and School of Biological Science, The University of Utah, Salt Lake City, UT, USA
| | - Ellen A A Nollen
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
29
|
Salzinger A, Ramesh V, Das Sharma S, Chandran S, Thangaraj Selvaraj B. Neuronal Circuit Dysfunction in Amyotrophic Lateral Sclerosis. Cells 2024; 13:792. [PMID: 38786016 PMCID: PMC11120636 DOI: 10.3390/cells13100792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/27/2024] [Accepted: 04/30/2024] [Indexed: 05/25/2024] Open
Abstract
The primary neural circuit affected in Amyotrophic Lateral Sclerosis (ALS) patients is the corticospinal motor circuit, originating in upper motor neurons (UMNs) in the cerebral motor cortex which descend to synapse with the lower motor neurons (LMNs) in the spinal cord to ultimately innervate the skeletal muscle. Perturbation of these neural circuits and consequent loss of both UMNs and LMNs, leading to muscle wastage and impaired movement, is the key pathophysiology observed. Despite decades of research, we are still lacking in ALS disease-modifying treatments. In this review, we document the current research from patient studies, rodent models, and human stem cell models in understanding the mechanisms of corticomotor circuit dysfunction and its implication in ALS. We summarize the current knowledge about cortical UMN dysfunction and degeneration, altered excitability in LMNs, neuromuscular junction degeneration, and the non-cell autonomous role of glial cells in motor circuit dysfunction in relation to ALS. We further highlight the advances in human stem cell technology to model the complex neural circuitry and how these can aid in future studies to better understand the mechanisms of neural circuit dysfunction underpinning ALS.
Collapse
Affiliation(s)
- Andrea Salzinger
- UK Dementia Research Institute, University of Edinburgh, Edinburgh EH16 4SB, UK; (A.S.); (V.R.); (S.D.S.); (S.C.)
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Vidya Ramesh
- UK Dementia Research Institute, University of Edinburgh, Edinburgh EH16 4SB, UK; (A.S.); (V.R.); (S.D.S.); (S.C.)
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Shreya Das Sharma
- UK Dementia Research Institute, University of Edinburgh, Edinburgh EH16 4SB, UK; (A.S.); (V.R.); (S.D.S.); (S.C.)
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Siddharthan Chandran
- UK Dementia Research Institute, University of Edinburgh, Edinburgh EH16 4SB, UK; (A.S.); (V.R.); (S.D.S.); (S.C.)
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Anne Rowling Regenerative Neurology Clinic (ARRNC), University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Bhuvaneish Thangaraj Selvaraj
- UK Dementia Research Institute, University of Edinburgh, Edinburgh EH16 4SB, UK; (A.S.); (V.R.); (S.D.S.); (S.C.)
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Anne Rowling Regenerative Neurology Clinic (ARRNC), University of Edinburgh, Edinburgh EH16 4SB, UK
| |
Collapse
|
30
|
Odierna GL, Vucic S, Dyer M, Dickson T, Woodhouse A, Blizzard C. How do we get from hyperexcitability to excitotoxicity in amyotrophic lateral sclerosis? Brain 2024; 147:1610-1621. [PMID: 38408864 PMCID: PMC11068114 DOI: 10.1093/brain/awae039] [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: 07/17/2023] [Revised: 11/15/2023] [Accepted: 12/10/2023] [Indexed: 02/28/2024] Open
Abstract
Amyotrophic lateral sclerosis is a devastating neurodegenerative disease that, at present, has no effective cure. Evidence of increased circulating glutamate and hyperexcitability of the motor cortex in patients with amyotrophic lateral sclerosis have provided an empirical support base for the 'dying forward' excitotoxicity hypothesis. The hypothesis postulates that increased activation of upper motor neurons spreads pathology to lower motor neurons in the spinal cord in the form of excessive glutamate release, which triggers excitotoxic processes. Many clinical trials have focused on therapies that target excitotoxicity via dampening neuronal activation, but not all are effective. As such, there is a growing tension between the rising tide of evidence for the 'dying forward' excitotoxicity hypothesis and the failure of therapies that target neuronal activation. One possible solution to these contradictory outcomes is that our interpretation of the current evidence requires revision in the context of appreciating the complexity of the nervous system and the limitations of the neurobiological assays we use to study it. In this review we provide an evaluation of evidence relevant to the 'dying forward' excitotoxicity hypothesis and by doing so, identify key gaps in our knowledge that need to be addressed. We hope to provide a road map from hyperexcitability to excitotoxicity so that we can better develop therapies for patients suffering from amyotrophic lateral sclerosis. We conclude that studies of upper motor neuron activity and their synaptic output will play a decisive role in the future of amyotrophic lateral sclerosis therapy.
Collapse
Affiliation(s)
- G Lorenzo Odierna
- Tasmanian School of Medicine, University of Tasmania, Hobart, TAS 7000, Australia
| | - Steve Vucic
- Brain and Nerve Research Center, The University of Sydney, Sydney 2050, Australia
| | - Marcus Dyer
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
- Department of Pharmaceutical and Pharmacological Sciences, Center for Neurosciences, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| | - Tracey Dickson
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Adele Woodhouse
- The Wicking Dementia Centre, University of Tasmania, Hobart, TAS 7000, Australia
| | - Catherine Blizzard
- Tasmanian School of Medicine, University of Tasmania, Hobart, TAS 7000, Australia
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| |
Collapse
|
31
|
Vucic S, de Carvalho M, Bashford J, Alix JJP. Contribution of neurophysiology to the diagnosis and monitoring of ALS. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 176:87-118. [PMID: 38802184 DOI: 10.1016/bs.irn.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
This chapter describes the role of neurophysiological techniques in diagnosing and monitoring amyotrophic lateral sclerosis (ALS). Despite many advances, electromyography (EMG) remains a keystone investigation from which to build support for a diagnosis of ALS, demonstrating the pathophysiological processes of motor unit hyperexcitability, denervation and reinnervation. We consider development of the different diagnostic criteria and the role of EMG therein. While not formally recognised by established diagnostic criteria, we discuss the pioneering studies that have demonstrated the diagnostic potential of transcranial magnetic stimulation (TMS) of the motor cortex and highlight the growing evidence for TMS in the diagnostic process. Finally, accurately monitoring disease progression is crucial for the successful implementation of clinical trials. Neurophysiological measures of disease state have been incorporated into clinical trials for over 20 years and we review prominent techniques for assessing disease progression.
Collapse
Affiliation(s)
- Steve Vucic
- Brain and Nerve Research Centre, Concord Clinical School and Department of Neurology, Concord Repatriation General Hospital, The University of Sydney, Sydney, NSW, Australia
| | - Mamede de Carvalho
- Instituto de Medicina Molecular João Lobo Antunes, Centro de Estudos Egas Moniz, Faculty of Medicine, Universidade de Lisboa, Lisboa, Portugal; Department of Neurosciences, CHULN, Centro Académico de Medicina de Lisboa, Lisboa, Portugal
| | - James Bashford
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - James J P Alix
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kingdom.
| |
Collapse
|
32
|
Costa-Pinto S, Gonçalves-Ribeiro J, Tedim-Moreira J, Socodato R, Relvas JB, Sebastião AM, Vaz SH. Communication defects with astroglia contribute to early impairments in the motor cortex plasticity of SOD1 G93A mice. Neurobiol Dis 2024; 193:106435. [PMID: 38336279 DOI: 10.1016/j.nbd.2024.106435] [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: 10/13/2023] [Revised: 02/06/2024] [Accepted: 02/06/2024] [Indexed: 02/12/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease, involving the selective degeneration of cortical upper synapses in the primary motor cortex (M1). Excitotoxicity in ALS occurs due to an imbalance between excitation and inhibition, closely linked to the loss/gain of astrocytic function. Using the ALS SOD1G93A mice, we investigated the astrocytic contribution for the electrophysiological alterations observed in the M1 of SOD1G93A mice, throughout disease progression. Results showed that astrocytes are involved in synaptic dysfunction observed in presymptomatic SOD1G93A mice, since astrocytic glutamate transport currents are diminished and pharmacological inhibition of astrocytes only impaired long-term potentiation and basal transmission in wild-type mice. Proteomic analysis revealed major differences in neuronal transmission, metabolism, and immune system in upper synapses, confirming early communication deficits between neurons and astroglia. These results provide valuable insights into the early impact of upper synapses in ALS and the lack of supportive functions of cortical astrocytes, highlighting the possibility of manipulating astrocytes to improve synaptic function.
Collapse
Affiliation(s)
- Sara Costa-Pinto
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon 1649-028, Portugal
| | - Joana Gonçalves-Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon 1649-028, Portugal
| | - Joana Tedim-Moreira
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC), University of Porto, Porto 4200-135, Portugal; Department of Biomedicine, Faculty of Medicine, University of Porto, Porto 4200-135, Portugal
| | - Renato Socodato
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC), University of Porto, Porto 4200-135, Portugal
| | - João B Relvas
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC), University of Porto, Porto 4200-135, Portugal; Department of Biomedicine, Faculty of Medicine, University of Porto, Porto 4200-135, Portugal
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon 1649-028, Portugal
| | - Sandra H Vaz
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon 1649-028, Portugal.
| |
Collapse
|
33
|
Lépine S, Nauleau-Javaudin A, Deneault E, Chen CXQ, Abdian N, Franco-Flores AK, Haghi G, Castellanos-Montiel MJ, Maussion G, Chaineau M, Durcan TM. Homozygous ALS-linked mutations in TARDBP/TDP-43 lead to hypoactivity and synaptic abnormalities in human iPSC-derived motor neurons. iScience 2024; 27:109166. [PMID: 38433895 PMCID: PMC10905001 DOI: 10.1016/j.isci.2024.109166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/21/2023] [Accepted: 02/05/2024] [Indexed: 03/05/2024] Open
Abstract
Cytoplasmic mislocalization and aggregation of the RNA-binding protein TDP-43 is a pathological hallmark of the motor neuron (MN) disease amyotrophic lateral sclerosis (ALS). Furthermore, while mutations in TARDBP (encoding TDP-43) have been associated with ALS, the pathogenic consequences of these mutations remain poorly understood. Using CRISPR-Cas9, we engineered two homozygous knock-in induced pluripotent stem cell lines carrying mutations in TARDBP encoding TDP-43A382T and TDP-43G348C, two common yet understudied ALS TDP-43 variants. Motor neurons (MNs) differentiated from knock-in iPSCs had normal viability and displayed no significant changes in TDP-43 subcellular localization, phosphorylation, solubility, or aggregation compared with isogenic control MNs. However, our results highlight synaptic impairments in both TDP-43A382T and TDP-43G348C MN cultures, as reflected in synapse abnormalities and alterations in spontaneous neuronal activity. Collectively, our findings suggest that MN dysfunction may precede the occurrence of TDP-43 pathology and neurodegeneration in ALS and further implicate synaptic and excitability defects in the pathobiology of this disease.
Collapse
Affiliation(s)
- Sarah Lépine
- Early Drug Discovery Unit (EDDU), The Neuro-Montreal Neurological Institute and Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
- Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 2M1, Canada
| | - Angela Nauleau-Javaudin
- Early Drug Discovery Unit (EDDU), The Neuro-Montreal Neurological Institute and Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
- Faculty of Medicine, Université de Montréal, Montreal, QC H3C 3J7, Canada
| | - Eric Deneault
- Centre for Oncology, Radiopharmaceuticals and Research; Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada, Ottawa, ON K1A 0K9, Canada
| | - Carol X.-Q. Chen
- Early Drug Discovery Unit (EDDU), The Neuro-Montreal Neurological Institute and Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Narges Abdian
- Early Drug Discovery Unit (EDDU), The Neuro-Montreal Neurological Institute and Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Anna Krystina Franco-Flores
- Early Drug Discovery Unit (EDDU), The Neuro-Montreal Neurological Institute and Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Ghazal Haghi
- Early Drug Discovery Unit (EDDU), The Neuro-Montreal Neurological Institute and Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - María José Castellanos-Montiel
- Early Drug Discovery Unit (EDDU), The Neuro-Montreal Neurological Institute and Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Gilles Maussion
- Early Drug Discovery Unit (EDDU), The Neuro-Montreal Neurological Institute and Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Mathilde Chaineau
- Early Drug Discovery Unit (EDDU), The Neuro-Montreal Neurological Institute and Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Thomas Martin Durcan
- Early Drug Discovery Unit (EDDU), The Neuro-Montreal Neurological Institute and Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| |
Collapse
|
34
|
Di Lazzaro V, Ranieri F, Bączyk M, de Carvalho M, Dileone M, Dubbioso R, Fernandes S, Kozak G, Motolese F, Ziemann U. Novel approaches to motoneuron disease/ALS treatment using non-invasive brain and spinal stimulation: IFCN handbook chapter. Clin Neurophysiol 2024; 158:114-136. [PMID: 38218077 DOI: 10.1016/j.clinph.2023.12.012] [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: 10/13/2023] [Revised: 11/28/2023] [Accepted: 12/17/2023] [Indexed: 01/15/2024]
Abstract
Non-invasive brain stimulation techniques have been exploited in motor neuron disease (MND) with multifold objectives: to support the diagnosis, to get insights in the pathophysiology of these disorders and, more recently, to slow down disease progression. In this review, we consider how neuromodulation can now be employed to treat MND, with specific attention to amyotrophic lateral sclerosis (ALS), the most common form with upper motoneuron (UMN) involvement, taking into account electrophysiological abnormalities revealed by human and animal studies that can be targeted by neuromodulation techniques. This review article encompasses repetitive transcranial magnetic stimulation methods (including low-frequency, high-frequency, and pattern stimulation paradigms), transcranial direct current stimulation as well as experimental findings with the newer approach of trans-spinal direct current stimulation. We also survey and discuss the trials that have been performed, and future perspectives.
Collapse
Affiliation(s)
- Vincenzo Di Lazzaro
- Department of Medicine and Surgery, Unit of Neurology, Neurophysiology, Neurobiology and Psychiatry, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy; Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy.
| | - Federico Ranieri
- Neurology Unit, Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, P.Le L.A. Scuro 10, 37134 Verona, Italy
| | - Marcin Bączyk
- Department of Neurobiology, Poznań University of Physical Education, Królowej Jadwigi Street 27/39, 61-871 Poznań, Poland
| | - Mamede de Carvalho
- Institute of Physiology, Institute of Molecular Medicine-JLA, Egas Moniz Study Centre, Faculty of Medicine, University of Lisbon, Lisbon 1649-028, Portugal; Department of Neurosciences and Mental Health, CHULN, Lisbon, Portugal
| | - Michele Dileone
- Faculty of Health Sciences, UCLM Talavera de la Reina, Toledo, Spain; Neurology Department, Hospital Nuestra Señora del Prado, Talavera de la Reina, Toledo, Spain
| | - Raffaele Dubbioso
- Neurophysiology Unit, Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Napoli, Italy
| | - Sofia Fernandes
- Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016-Lisboa, Portugal
| | - Gabor Kozak
- Department of Neurology and Stroke, University of Tübingen, Tübingen, Germany; Hertie-Institute of Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Francesco Motolese
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
| | - Ulf Ziemann
- Department of Neurology and Stroke, University of Tübingen, Tübingen, Germany; Hertie-Institute of Clinical Brain Research, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
35
|
Moreno-Roco J, del Valle L, Jiménez D, Acosta I, Castillo JL, Dharmadasa T, Kiernan MC, Matamala JM. Diagnostic utility of transcranial magnetic stimulation for neurodegenerative disease: a critical review. Dement Neuropsychol 2024; 17:e20230048. [PMID: 38189033 PMCID: PMC10768644 DOI: 10.1590/1980-5764-dn-2023-0048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/13/2023] [Accepted: 09/24/2023] [Indexed: 01/09/2024] Open
Abstract
Neurodegenerative diseases pose significant challenges due to their impact on brain structure, function, and cognition. As life expectancy rises, the prevalence of these disorders is rapidly increasing, resulting in substantial personal, familial, and societal burdens. Efforts have been made to optimize the diagnostic and therapeutic processes, primarily focusing on clinical, cognitive, and imaging characterization. However, the emergence of non-invasive brain stimulation techniques, specifically transcranial magnetic stimulation (TMS), offers unique functional insights and diagnostic potential. TMS allows direct evaluation of brain function, providing valuable information inaccessible through other methods. This review aims to summarize the current and potential diagnostic utility of TMS in investigating neurodegenerative diseases, highlighting its relevance to the field of cognitive neuroscience. The findings presented herein contribute to the growing body of research focused on improving our understanding and management of these debilitating conditions, particularly in regions with limited resources and a pressing need for innovative approaches.
Collapse
Affiliation(s)
- Javier Moreno-Roco
- Universidad de Chile, Facultad de Medicina, Laboratorio de Neurología y Neurofisiología Traslacional, Santiago, Chile
- Universidad de Chile, Facultad de Medicina, Centro de Investigación Clínica Avanzado (CICA) Oriente, Santiago, Chile
- Universidad de Chile, Facultad de Medicina, Departamento de Ciencias Neurológicas Oriente, Santiago, Chile
| | - Lucía del Valle
- Universidad de Chile, Facultad de Medicina, Laboratorio de Neurología y Neurofisiología Traslacional, Santiago, Chile
- Universidad de Chile, Facultad de Medicina, Centro de Investigación Clínica Avanzado (CICA) Oriente, Santiago, Chile
- Universidad de Chile, Facultad de Medicina, Departamento de Ciencias Neurológicas Oriente, Santiago, Chile
| | - Daniel Jiménez
- Universidad de Chile, Facultad de Medicina, Laboratorio de Neurología y Neurofisiología Traslacional, Santiago, Chile
- Universidad de Chile, Facultad de Medicina, Centro de Investigación Clínica Avanzado (CICA) Oriente, Santiago, Chile
- Universidad de Chile, Facultad de Medicina, Departamento de Ciencias Neurológicas Oriente, Santiago, Chile
- Hospital del Salvador, Servicio de Neurología, Santiago, Chile
| | - Ignacio Acosta
- Universidad de Chile, Facultad de Medicina, Laboratorio de Neurología y Neurofisiología Traslacional, Santiago, Chile
- Universidad de Chile, Facultad de Medicina, Centro de Investigación Clínica Avanzado (CICA) Oriente, Santiago, Chile
- Universidad de Chile, Facultad de Medicina, Departamento de Ciencias Neurológicas Oriente, Santiago, Chile
- Hospital del Salvador, Servicio de Neurología, Santiago, Chile
| | - José Luis Castillo
- Universidad de Chile, Facultad de Medicina, Laboratorio de Neurología y Neurofisiología Traslacional, Santiago, Chile
- Universidad de Chile, Facultad de Medicina, Departamento de Ciencias Neurológicas Oriente, Santiago, Chile
| | - Thanuja Dharmadasa
- University of Melbourne, The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
- The Royal Melbourne Hospital, Department of Neurology, Parkville, Victoria, Australia
- University of Sydney, Brain and Mind Centre, Sydney, Australia
| | - Matthew C. Kiernan
- University of Sydney, Brain and Mind Centre, Sydney, Australia
- Royal Prince Alfred Hospital, Department of Neurology, Sydney, AustraliaArgento
| | - José Manuel Matamala
- Universidad de Chile, Facultad de Medicina, Laboratorio de Neurología y Neurofisiología Traslacional, Santiago, Chile
- Universidad de Chile, Facultad de Medicina, Centro de Investigación Clínica Avanzado (CICA) Oriente, Santiago, Chile
- Universidad de Chile, Facultad de Medicina, Departamento de Ciencias Neurológicas Oriente, Santiago, Chile
- Universidad de Chile, Facultad de Medicina, Departamento de Neurociencias, Santiago, Chile
- Universidad de Chile, Facultad de Medicina, Instituto de Neurociencia Biomédica (BNI), Santiago, Chile
| |
Collapse
|
36
|
Nolan M, Scott C, Hof PR, Ansorge O. Betz cells of the primary motor cortex. J Comp Neurol 2024; 532:e25567. [PMID: 38289193 PMCID: PMC10952528 DOI: 10.1002/cne.25567] [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: 08/09/2023] [Revised: 11/11/2023] [Accepted: 11/17/2023] [Indexed: 02/01/2024]
Abstract
Betz cells, named in honor of Volodymyr Betz (1834-1894), who described them as "giant pyramids" in the primary motor cortex of primates and other mammalian species, are layer V extratelencephalic projection (ETP) neurons that directly innervate α-motoneurons of the brainstem and spinal cord. Despite their large volume and circumferential dendritic architecture, to date, no single molecular criterion has been established that unequivocally distinguishes adult Betz cells from other layer V ETP neurons. In primates, transcriptional signatures suggest the presence of at least two ETP neuron clusters that contain mature Betz cells; these are characterized by an abundance of axon guidance and oxidative phosphorylation transcripts. How neurodevelopmental programs drive the distinct positional and morphological features of Betz cells in humans remains unknown. Betz cells display a distinct biphasic firing pattern involving early cessation of firing followed by delayed sustained acceleration in spike frequency and magnitude. Few cell type-specific transcripts and electrophysiological characteristics are conserved between rodent layer V ETP neurons of the motor cortex and primate Betz cells. This has implications for the modeling of disorders that affect the motor cortex in humans, such as amyotrophic lateral sclerosis (ALS). Perhaps vulnerability to ALS is linked to the evolution of neural networks for fine motor control reflected in the distinct morphomolecular architecture of the human motor cortex, including Betz cells. Here, we discuss histological, molecular, and functional data concerning the position of Betz cells in the emerging taxonomy of neurons across diverse species and their role in neurological disorders.
Collapse
Affiliation(s)
- Matthew Nolan
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
- Department of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Connor Scott
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Patrick. R. Hof
- Nash Family Department of Neuroscience and Friedman Brain InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Olaf Ansorge
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| |
Collapse
|
37
|
de Carvalho M, Swash M. Diagnosis and differential diagnosis of MND/ALS: IFCN handbook chapter. Clin Neurophysiol Pract 2023; 9:27-38. [PMID: 38249779 PMCID: PMC10796809 DOI: 10.1016/j.cnp.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 11/01/2023] [Accepted: 12/07/2023] [Indexed: 01/23/2024] Open
Abstract
•Accurate and rapid diagnosis of amyotrophic lateral sclerosis (ALS) is important to prevent erroneous interventions. •The recent Gold Coast criteria are easily applicable and have high sensitivity and specificity. •Future developments will help to distinguish ALS as a specific clinical-pathologic entity. Accurate and rapid diagnosis of amyotrophic lateral sclerosis (ALS) is essential in order to provide accurate information for patient and family, to avoid time-consuming investigations and to permit an appropriate management plan. ALS is variable regarding presentation, disease progression, genetic profile and patient reaction to the diagnosis. It is obviously important to exclude treatable conditions but, in most patients, for experienced neurologists the diagnosis is clear-cut, depending on the presence of progressive upper and lower motor neuron signs. Patients with signs of restricted lower motor neuron (LMN) or upper motor neuron (UMN) dysfunction may present diagnostic difficulty, but electromyography (EMG) is often a determinant diagnostic test since it may exclude other disorders. Transcranial magnetic stimulation may aid detection of UMN dysfunction, and brain and spinal cord MRI, ultrasound and blood neurofilament measurements, have begun to have clinical impact, although none are themselves diagnostic tests. Several sets of diagnostic criteria have been proposed in the past; all rely on clinical LMN and UMN signs in different anatomic territories, EMG changes, exclusion of other disorders, and disease progression, in particular evidence of spreading to other anatomic territories. Fasciculations are a characteristic clinical feature and increased importance is now attached to fasciculation potentials detected by EMG, when associated with classical signs of denervation and reinnervation. The Gold Coast diagnostic criteria rely on the presence of UMN and LMN signs in one (or more) anatomic territory, or LMN signs in two (or more) anatomic territories, recognizing the fundamental clinical requirements of disease progression and exclusion of other diseases. Recent studies confirm a high sensitivity without loss of specificity using these Gold Coast criteria. In considering the diagnosis of ALS a critical question for future understanding is whether ALS should be considered a syndrome or a specific clinico-pathologic entity; this can only be addressed in the light of more complete knowledge.
Collapse
Affiliation(s)
- Mamede de Carvalho
- Faculdade de Medicina- Instituto de Medicina Molecular, Centro de Estudos Egas Moniz, Universidade de Lisboa, Lisbon, Portugal
- Department of Neurosciences and Mental Health, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa-Norte, Lisbon, Portugal
| | - Michael Swash
- Faculdade de Medicina- Instituto de Medicina Molecular, Centro de Estudos Egas Moniz, Universidade de Lisboa, Lisbon, Portugal
- Departments of Neurology and Neurosciences, Barts and the London School of Medicine, Queen Mary University of London and Royal London Hospital, UK
| |
Collapse
|
38
|
Stringer RN, Weiss N. Pathophysiology of ion channels in amyotrophic lateral sclerosis. Mol Brain 2023; 16:82. [PMID: 38102715 PMCID: PMC10722804 DOI: 10.1186/s13041-023-01070-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 12/11/2023] [Indexed: 12/17/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) stands as the most prevalent and severe form of motor neuron disease, affecting an estimated 2 in 100,000 individuals worldwide. It is characterized by the progressive loss of cortical, brainstem, and spinal motor neurons, ultimately resulting in muscle weakness and death. Although the etiology of ALS remains poorly understood in most cases, the remodelling of ion channels and alteration in neuronal excitability represent a hallmark of the disease, manifesting not only during the symptomatic period but also in the early pre-symptomatic stages. In this review, we delve into these alterations observed in ALS patients and preclinical disease models, and explore their consequences on neuronal activities. Furthermore, we discuss the potential of ion channels as therapeutic targets in the context of ALS.
Collapse
Affiliation(s)
- Robin N Stringer
- Department of Pathophysiology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Norbert Weiss
- Department of Pathophysiology, Third Faculty of Medicine, Charles University, Prague, Czech Republic.
- Center of Biosciences, Institute of Molecular Physiology and Genetics, Slovak Academy of Sciences, Bratislava, Slovakia.
| |
Collapse
|
39
|
Pavey N, Hannaford A, Higashihara M, van den Bos M, Kiernan MC, Menon P, Vucic S. Utility of split hand index with different motor unit number estimation techniques in ALS. Clin Neurophysiol 2023; 156:175-182. [PMID: 37967511 DOI: 10.1016/j.clinph.2023.09.018] [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: 08/07/2023] [Revised: 09/04/2023] [Accepted: 09/27/2023] [Indexed: 11/17/2023]
Abstract
OBJECTIVE Utility of the split hand index (SI) in amyotrophic lateral sclerosis (ALS) has been reported when using the compound muscle action potential (CMAP) amplitude method (SICMAP amp). A motor unit number index (MUNIX) based SI method (SIMUNIX) was purported to exhibit higher sensitivity. The present study assessed the clinical utility of SI, derived by CMAP amplitude, MUNIX and MScan-MUNE (SIMScanFit-MUNE) methods, in ALS. METHODS Sixty-two consecutive patients with neuromuscular symptoms (36 ALS and 26 ALS-mimics) were prospectively recruited. The SI was derived by dividing the product of the CMAP amplitude, MUNIX and MScan-MUNE values recorded over first dorsal interosseous and abductor pollicis brevis by values recorded over abductor digit minimi. RESULTS SICMAP amp, SIMUNIX and SIMScanFit-MUNE were significantly reduced in ALS, with SICMAP amp (area under curve (AUC) = 0.801) and SIMScanFit-MUNE (AUC = 0.805) exhibiting greater diagnostic utility than SIMUNIX (AUC = 0.713). SICMAP amp and SIMScanFit-MUNE exhibited significant correlations with clinical measures of functional disability and weakness of intrinsic hand muscles. CONCLUSIONS SI differentiated ALS from mimic disorders, with SICMAP amp and SIMScanFit-MUNE exhibiting greater utility. SIGNIFICANCE The split hand index represents could serve as a potential diagnostic biomarker in ALS.
Collapse
Affiliation(s)
- Nathan Pavey
- Brain and Nerve Research Centre, Concord Clinical School, The University of Sydney, Concord Hospital, Sydney, NSW Australia
| | - Andrew Hannaford
- Brain and Nerve Research Centre, Concord Clinical School, The University of Sydney, Concord Hospital, Sydney, NSW Australia
| | - Mana Higashihara
- Department of Neurology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Mehdi van den Bos
- Brain and Nerve Research Centre, Concord Clinical School, The University of Sydney, Concord Hospital, Sydney, NSW Australia
| | - Matthew C Kiernan
- Brain and Mind Centre, The University of Sydney; and Department of Neurology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Parvathi Menon
- Brain and Nerve Research Centre, Concord Clinical School, The University of Sydney, Concord Hospital, Sydney, NSW Australia
| | - Steve Vucic
- Brain and Nerve Research Centre, Concord Clinical School, The University of Sydney, Concord Hospital, Sydney, NSW Australia.
| |
Collapse
|
40
|
McMackin R, Bede P, Ingre C, Malaspina A, Hardiman O. Biomarkers in amyotrophic lateral sclerosis: current status and future prospects. Nat Rev Neurol 2023; 19:754-768. [PMID: 37949994 DOI: 10.1038/s41582-023-00891-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2023] [Indexed: 11/12/2023]
Abstract
Disease heterogeneity in amyotrophic lateral sclerosis poses a substantial challenge in drug development. Categorization based on clinical features alone can help us predict the disease course and survival, but quantitative measures are also needed that can enhance the sensitivity of the clinical categorization. In this Review, we describe the emerging landscape of diagnostic, categorical and pharmacodynamic biomarkers in amyotrophic lateral sclerosis and their place in the rapidly evolving landscape of new therapeutics. Fluid-based markers from cerebrospinal fluid, blood and urine are emerging as useful diagnostic, pharmacodynamic and predictive biomarkers. Combinations of imaging measures have the potential to provide important diagnostic and prognostic information, and neurophysiological methods, including various electromyography-based measures and quantitative EEG-magnetoencephalography-evoked responses and corticomuscular coherence, are generating useful diagnostic, categorical and prognostic markers. Although none of these biomarker technologies has been fully incorporated into clinical practice or clinical trials as a primary outcome measure, strong evidence is accumulating to support their clinical utility.
Collapse
Affiliation(s)
- Roisin McMackin
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, The University of Dublin, Dublin, Ireland
- Academic Unit of Neurology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, The University of Dublin, Dublin, Ireland
| | - Peter Bede
- Academic Unit of Neurology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, The University of Dublin, Dublin, Ireland
- Computational Neuroimaging Group, School of Medicine, Trinity College Dublin, The University of Dublin, Dublin, Ireland
- Department of Neurology, St James's Hospital, Dublin, Ireland
| | - Caroline Ingre
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Andrea Malaspina
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Orla Hardiman
- Academic Unit of Neurology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, The University of Dublin, Dublin, Ireland.
- Department of Neurology, Beaumont Hospital, Dublin, Ireland.
| |
Collapse
|
41
|
Clark RM, Clark CM, Lewis KE, Dyer MS, Chuckowree JA, Hoyle JA, Blizzard CA, Dickson TC. Intranasal neuropeptide Y1 receptor antagonism improves motor deficits in symptomatic SOD1 ALS mice. Ann Clin Transl Neurol 2023; 10:1985-1999. [PMID: 37644692 PMCID: PMC10647012 DOI: 10.1002/acn3.51885] [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: 07/31/2023] [Accepted: 08/09/2023] [Indexed: 08/31/2023] Open
Abstract
OBJECTIVE Neuropeptide Y (NPY) is a 36 amino acid peptide widely considered to provide neuroprotection in a range of neurodegenerative diseases. In the fatal motor neuron disease amyotrophic lateral sclerosis (ALS), recent evidence supports a link between NPY and ALS disease processes. The goal of this study was to determine the therapeutic potential and role of NPY in ALS, harnessing the brain-targeted intranasal delivery of the peptide, previously utilised to correct motor and cognitive phenotypes in other neurological conditions. METHODS To confirm the association with clinical disease characteristics, NPY expression was quantified in post-mortem motor cortex tissue of ALS patients and age-matched controls. The effect of NPY on ALS cortical pathophysiology was investigated using slice electrophysiology and multi-electrode array recordings of SOD1G93A cortical cultures in vitro. The impact of NPY on ALS disease trajectory was investigated by treating SOD1G93A mice intranasally with NPY and selective NPY receptor agonists and antagonists from pre-symptomatic and symptomatic phases of disease. RESULTS In the human post-mortem ALS motor cortex, we observe a significant increase in NPY expression, which is not present in the somatosensory cortex. In vitro, we demonstrate that NPY can ameliorate ALS hyperexcitability, while brain-targeted nasal delivery of NPY and a selective NPY Y1 receptor antagonist modified survival and motor deficits specifically within the symptomatic phase of the disease in the ALS SOD1G93A mouse. INTERPRETATION Taken together, these findings highlight the capacity for non-invasive brain-targeted interventions in ALS and support antagonism of NPY Y1Rs as a novel strategy to improve ALS motor function.
Collapse
Affiliation(s)
- Rosemary M. Clark
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmania7000Australia
| | - Courtney M. Clark
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmania7000Australia
| | - Katherine E.A. Lewis
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmania7000Australia
| | - Marcus S. Dyer
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmania7000Australia
| | - Jyoti A. Chuckowree
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmania7000Australia
| | - Joshua A. Hoyle
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmania7000Australia
| | - Catherine A. Blizzard
- Tasmanian School of Medicine, College of Health and MedicineUniversity of TasmaniaHobartTasmania7000Australia
| | - Tracey C. Dickson
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmania7000Australia
| |
Collapse
|
42
|
Benussi A, Cantoni V, Grassi M, Libri I, Cotelli MS, Tarantino B, Datta A, Thomas C, Huber N, Kärkkäinen S, Herukka SK, Haapasalo A, Filosto M, Padovani A, Borroni B. Cortico-spinal tDCS in amyotrophic lateral sclerosis: A randomized, double-blind, sham-controlled trial followed by an open-label phase. Brain Stimul 2023; 16:1666-1676. [PMID: 37977335 DOI: 10.1016/j.brs.2023.11.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/07/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a progressive disease for which no curative treatment is currently available. OBJECTIVE This study aimed to investigate whether cortico-spinal transcranial direct current stimulation (tDCS) could mitigate symptoms in ALS patients via a randomized, double-blind, sham-controlled trial, followed by an open-label phase. METHODS Thirty-one participants were randomized into two groups for the initial controlled phase. At baseline (T0), Group 1 received placebo stimulation (sham tDCS), while Group 2 received cortico-spinal stimulation (real tDCS) for five days/week for two weeks (T1), with an 8-week (T2) follow-up (randomized, double-blind, sham-controlled phase). At the 24-week follow-up (T3), all participants (Groups 1 and 2) received a second treatment of anodal bilateral motor cortex and cathodal spinal stimulation (real tDCS) for five days/week for two weeks (T4). Follow-up evaluations were performed at 32-weeks (T5) and 48-weeks (T6) (open-label phase). At each time point, clinical assessment, blood sampling, and intracortical connectivity measures using transcranial magnetic stimulation (TMS) were evaluated. Additionally, we evaluated survival rates. RESULTS Compared to sham stimulation, cortico-spinal tDCS significantly improved global strength, caregiver burden, and quality of life scores, which correlated with the restoration of intracortical connectivity measures. Serum neurofilament light levels decreased among patients who underwent real tDCS but not in those receiving sham tDCS. The number of completed 2-week tDCS treatments significantly influenced patient survival. CONCLUSIONS Cortico-spinal tDCS may represent a promising therapeutic and rehabilitative approach for patients with ALS. Further larger-scale studies are necessary to evaluate whether tDCS could potentially impact patient survival. CLINICAL TRIAL REGISTRATION NCT04293484.
Collapse
Affiliation(s)
- Alberto Benussi
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy; Neurology Unit, Department of Neurological and Vision Sciences, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Valentina Cantoni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Mario Grassi
- Department of Brain and Behavioural Sciences, Medical and Genomic Statistics Unit, University of Pavia, Pavia, Italy
| | - Ilenia Libri
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | | | - Barbara Tarantino
- Department of Brain and Behavioural Sciences, Medical and Genomic Statistics Unit, University of Pavia, Pavia, Italy
| | - Abhishek Datta
- Research & Development, Soterix Medical, Inc., New York, USA
| | - Chris Thomas
- Research & Development, Soterix Medical, Inc., New York, USA
| | - Nadine Huber
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Sari Kärkkäinen
- Institute of Clinical Medicine, Neurology, University of Eastern Finland, Kuopio, Finland
| | - Sanna-Kaisa Herukka
- Institute of Clinical Medicine, Neurology, University of Eastern Finland, Kuopio, Finland; Department of Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Annakaisa Haapasalo
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Massimiliano Filosto
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy; NeMo-Brescia Clinical Center for Neuromuscular Diseases, Gussago, Brescia, Italy
| | - Alessandro Padovani
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy; Neurology Unit, Department of Neurological and Vision Sciences, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Barbara Borroni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy; Neurology Unit, Department of Neurological and Vision Sciences, ASST Spedali Civili di Brescia, Brescia, Italy.
| |
Collapse
|
43
|
Dyer MS, Odierna GL, Clark RM, Woodhouse A, Blizzard CA. Synaptic remodeling follows upper motor neuron hyperexcitability in a rodent model of TDP-43. Front Cell Neurosci 2023; 17:1274979. [PMID: 37941604 PMCID: PMC10628445 DOI: 10.3389/fncel.2023.1274979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 10/05/2023] [Indexed: 11/10/2023] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is an incurable disease characterized by relentlessly progressive degeneration of the corticomotor system. Cortical hyperexcitability has been identified as an early pre-symptomatic biomarker of ALS. This suggests that hyperexcitability occurs upstream in the ALS pathological cascade and may even be part of the mechanism that drives development of symptoms or loss of motor neurons in the spinal cord. However, many studies also indicate a loss to the synaptic machinery that mediates synaptic input which raises the question of which is the driver of disease, and which is a homeostatic response. Herein, we used an inducible mouse model of TDP-43 mediated ALS that permits for the construction of detailed phenotypic timelines. Our work comprehensively describes the relationship between intrinsic hyperexcitability and altered synaptic input onto motor cortical layer 5 pyramidal neurons over time. As a result, we have constructed the most complete timeline of electrophysiological changes following induction of TDP-43 dysfunction in the motor cortex. We report that intrinsic hyperexcitability of layer 5 pyramidal neurons precedes changes to excitatory synaptic connections, which manifest as an overall loss of inputs onto layer 5 pyramidal neurons. This finding highlights the importance of hyperexcitability as a primary mechanism of ALS and re-contextualizes synaptic changes as possibly representing secondary adaptive responses. Recognition of the relationship between intrinsic hyperexcitability and reduced excitatory synaptic input has important implications for the development of useful therapies against ALS. Novel strategies will need to be developed that target neuronal output by managing excitability against synapses separately.
Collapse
Affiliation(s)
- Marcus S. Dyer
- Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
- Department of Pharmaceutical and Pharmacological Sciences, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - G. Lorenzo Odierna
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Rosemary M. Clark
- Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Adele Woodhouse
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Catherine A. Blizzard
- Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
| |
Collapse
|
44
|
Xie M, Pallegar PN, Parusel S, Nguyen AT, Wu LJ. Regulation of cortical hyperexcitability in amyotrophic lateral sclerosis: focusing on glial mechanisms. Mol Neurodegener 2023; 18:75. [PMID: 37858176 PMCID: PMC10585818 DOI: 10.1186/s13024-023-00665-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 10/05/2023] [Indexed: 10/21/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder characterized by the loss of both upper and lower motor neurons, resulting in muscle weakness, atrophy, paralysis, and eventually death. Motor cortical hyperexcitability is a common phenomenon observed at the presymptomatic stage of ALS. Both cell-autonomous (the intrinsic properties of motor neurons) and non-cell-autonomous mechanisms (cells other than motor neurons) are believed to contribute to cortical hyperexcitability. Decoding the pathological relevance of these dynamic changes in motor neurons and glial cells has remained a major challenge. This review summarizes the evidence of cortical hyperexcitability from both clinical and preclinical research, as well as the underlying mechanisms. We discuss the potential role of glial cells, particularly microglia, in regulating abnormal neuronal activity during the disease progression. Identifying early changes such as neuronal hyperexcitability in the motor system may provide new insights for earlier diagnosis of ALS and reveal novel targets to halt the disease progression.
Collapse
Affiliation(s)
- Manling Xie
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Praveen N Pallegar
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Sebastian Parusel
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Aivi T Nguyen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
- Department of Immunology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
45
|
Yang M, Liu M, Sánchez YF, Avazzadeh S, Quinlan LR, Liu G, Lu Y, Yang G, O'Brien T, Henshall DC, Hardiman O, Shen S. A novel protocol to derive cervical motor neurons from induced pluripotent stem cells for amyotrophic lateral sclerosis. Stem Cell Reports 2023; 18:1870-1883. [PMID: 37595581 PMCID: PMC10545486 DOI: 10.1016/j.stemcr.2023.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 08/20/2023] Open
Abstract
Sporadic amyotrophic lateral sclerosis (sALS) is the majority of ALS, and the lack of appropriate disease models has hindered its research. Induced pluripotent stem cell (iPSC) technology now permits derivation of iPSCs from somatic cells of sALS patients to investigate disease phenotypes and mechanisms. Most existing differentiation protocols are time-consuming or low efficient in generating motor neurons (MNs). Here we report a rapid and simple protocol to differentiate MNs in monolayer culture using small molecules, which led to nearly pure neural stem cells in 6 days, robust OLIG2+ pMNs (73%-91%) in 12 days, enriched CHAT+ cervical spinal MNs (sMNs) (88%-97%) in 18 days, and functionally mature sMNs in 28 days. This simple and reproducible protocol permitted the identification of hyperexcitability phenotypes in our sALS iPSC-derived sMNs, and its application in neurodegenerative diseases should facilitate in vitro disease modeling, drug screening, and the development of cell therapy.
Collapse
Affiliation(s)
- Meimei Yang
- Regenerative Medicine Institute, School of Medicine, University of Galway, H91 W2TY Galway, Ireland; FutureNeuro SFI Research Centre for Chronic and Rare Neurological Diseases and Department of Physiology & Medical Physics, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland
| | - Min Liu
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Molecular and Cellular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, China
| | - Yajaira Feller Sánchez
- Cellular Physiology Research Laboratory and CÚRAM SFI Centre for Research in Medical Devices, School of Medicine, University of Galway, H91 TK33 Galway, Ireland
| | - Sahar Avazzadeh
- Cellular Physiology Research Laboratory and CÚRAM SFI Centre for Research in Medical Devices, School of Medicine, University of Galway, H91 TK33 Galway, Ireland
| | - Leo R Quinlan
- Cellular Physiology Research Laboratory and CÚRAM SFI Centre for Research in Medical Devices, School of Medicine, University of Galway, H91 TK33 Galway, Ireland
| | - Gang Liu
- Department of Cardiology, The First Hospital of Hebei Medical University, Hebei Key Laboratory of Cardiac Injury Repair Mechanism Study, Hebei Key Laboratory of Heart and Metabolism, Hebei Engineering Research Center of Intelligent Medical Clinical Application, Hebei International Joint Research Center for Structural Heart Disease, Shijiazhuang, Hebei, China
| | - Yin Lu
- College of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Guangming Yang
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Timothy O'Brien
- Regenerative Medicine Institute, School of Medicine, University of Galway, H91 W2TY Galway, Ireland
| | - David C Henshall
- FutureNeuro SFI Research Centre for Chronic and Rare Neurological Diseases and Department of Physiology & Medical Physics, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland; Department of Physiology and Medical Physics, RCSI University of Medicine & Health Sciences, D02 YN77 Dublin, Ireland.
| | - Orla Hardiman
- FutureNeuro SFI Research Centre for Chronic and Rare Neurological Diseases and Department of Physiology & Medical Physics, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland; Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland.
| | - Sanbing Shen
- Regenerative Medicine Institute, School of Medicine, University of Galway, H91 W2TY Galway, Ireland; FutureNeuro SFI Research Centre for Chronic and Rare Neurological Diseases and Department of Physiology & Medical Physics, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland.
| |
Collapse
|
46
|
Aly A, Laszlo ZI, Rajkumar S, Demir T, Hindley N, Lamont DJ, Lehmann J, Seidel M, Sommer D, Franz-Wachtel M, Barletta F, Heumos S, Czemmel S, Kabashi E, Ludolph A, Boeckers TM, Henstridge CM, Catanese A. Integrative proteomics highlight presynaptic alterations and c-Jun misactivation as convergent pathomechanisms in ALS. Acta Neuropathol 2023; 146:451-475. [PMID: 37488208 PMCID: PMC10412488 DOI: 10.1007/s00401-023-02611-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 07/05/2023] [Accepted: 07/06/2023] [Indexed: 07/26/2023]
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a fatal neurodegenerative disease mainly affecting upper and lower motoneurons. Several functionally heterogeneous genes have been associated with the familial form of this disorder (fALS), depicting an extremely complex pathogenic landscape. This heterogeneity has limited the identification of an effective therapy, and this bleak prognosis will only improve with a greater understanding of convergent disease mechanisms. Recent evidence from human post-mortem material and diverse model systems has highlighted the synapse as a crucial structure actively involved in disease progression, suggesting that synaptic aberrations might represent a shared pathological feature across the ALS spectrum. To test this hypothesis, we performed the first comprehensive analysis of the synaptic proteome from post-mortem spinal cord and human iPSC-derived motoneurons carrying mutations in the major ALS genes. This integrated approach highlighted perturbations in the molecular machinery controlling vesicle release as a shared pathomechanism in ALS. Mechanistically, phosphoproteomic analysis linked the presynaptic vesicular phenotype to an accumulation of cytotoxic protein aggregates and to the pro-apoptotic activation of the transcription factor c-Jun, providing detailed insights into the shared pathobiochemistry in ALS. Notably, sub-chronic treatment of our iPSC-derived motoneurons with the fatty acid docosahexaenoic acid exerted a neuroprotective effect by efficiently rescuing the alterations revealed by our multidisciplinary approach. Together, this study provides strong evidence for the central and convergent role played by the synaptic microenvironment within the ALS spinal cord and highlights a potential therapeutic target that counteracts degeneration in a heterogeneous cohort of human motoneuron cultures.
Collapse
Affiliation(s)
- Amr Aly
- Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Zsofia I Laszlo
- Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, Scotland, UK
| | - Sandeep Rajkumar
- Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Tugba Demir
- Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Nicole Hindley
- Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, Scotland, UK
| | - Douglas J Lamont
- FingerPrints Proteomics Facility, Discovery Centre, School of Life Sciences, University of Dundee, Dundee, Scotland, UK
| | - Johannes Lehmann
- Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Mira Seidel
- Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Daniel Sommer
- Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | | | - Francesca Barletta
- Quantitative Biology Center (QBiC), University of Tübingen, 72076, Tübingen, Germany
| | - Simon Heumos
- Quantitative Biology Center (QBiC), University of Tübingen, 72076, Tübingen, Germany
- Biomedical Data Science, Department of Computer Science, University of Tübingen, 72076, Tübingen, Germany
| | - Stefan Czemmel
- Quantitative Biology Center (QBiC), University of Tübingen, 72076, Tübingen, Germany
| | - Edor Kabashi
- Laboratory of Translational Research for Neurological Disorders, Imagine Institute, Université de Paris, INSERM, UMR 1163, 75015, Paris, France
| | - Albert Ludolph
- Department of Neurology, Ulm University School of Medicine, Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE), Ulm Site, Germany
| | - Tobias M Boeckers
- Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE), Ulm Site, Germany
| | - Christopher M Henstridge
- Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, Scotland, UK.
| | - Alberto Catanese
- Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Ulm Site, Germany.
| |
Collapse
|
47
|
Acosta-Galeana I, Hernández-Martínez R, Reyes-Cruz T, Chiquete E, Aceves-Buendia JDJ. RNA-binding proteins as a common ground for neurodegeneration and inflammation in amyotrophic lateral sclerosis and multiple sclerosis. Front Mol Neurosci 2023; 16:1193636. [PMID: 37475885 PMCID: PMC10355071 DOI: 10.3389/fnmol.2023.1193636] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 06/14/2023] [Indexed: 07/22/2023] Open
Abstract
The neurodegenerative and inflammatory illnesses of amyotrophic lateral sclerosis and multiple sclerosis were once thought to be completely distinct entities that did not share any remarkable features, but new research is beginning to reveal more information about their similarities and differences. Here, we review some of the pathophysiological features of both diseases and their experimental models: RNA-binding proteins, energy balance, protein transportation, and protein degradation at the molecular level. We make a thorough analysis on TDP-43 and hnRNP A1 dysfunction, as a possible common ground in both pathologies, establishing a potential link between neurodegeneration and pathological immunity. Furthermore, we highlight the putative variations that diverge from a common ground in an atemporal course that proposes three phases for all relevant molecular events.
Collapse
Affiliation(s)
| | | | - Tania Reyes-Cruz
- Laboratorio de Biología Molecular, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Mexico City, Mexico
| | - Erwin Chiquete
- Departamento de Neurología y Psiquiatría, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Jose de Jesus Aceves-Buendia
- Departamento de Neurología y Psiquiatría, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
48
|
Spampinato DA, Ibanez J, Rocchi L, Rothwell J. Motor potentials evoked by transcranial magnetic stimulation: interpreting a simple measure of a complex system. J Physiol 2023; 601:2827-2851. [PMID: 37254441 PMCID: PMC10952180 DOI: 10.1113/jp281885] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 05/18/2023] [Indexed: 06/01/2023] Open
Abstract
Transcranial magnetic stimulation (TMS) is a non-invasive technique that is increasingly used to study the human brain. One of the principal outcome measures is the motor-evoked potential (MEP) elicited in a muscle following TMS over the primary motor cortex (M1), where it is used to estimate changes in corticospinal excitability. However, multiple elements play a role in MEP generation, so even apparently simple measures such as peak-to-peak amplitude have a complex interpretation. Here, we summarize what is currently known regarding the neural pathways and circuits that contribute to the MEP and discuss the factors that should be considered when interpreting MEP amplitude measured at rest in the context of motor processing and patients with neurological conditions. In the last part of this work, we also discuss how emerging technological approaches can be combined with TMS to improve our understanding of neural substrates that can influence MEPs. Overall, this review aims to highlight the capabilities and limitations of TMS that are important to recognize when attempting to disentangle sources that contribute to the physiological state-related changes in corticomotor excitability.
Collapse
Affiliation(s)
- Danny Adrian Spampinato
- Department of Clinical and Movement NeurosciencesUniversity College LondonLondonUK
- Department of Human NeurosciencesSapienza University of RomeRomeItaly
- Department of Clinical and Behavioral NeurologyIRCCS Santa Lucia FoundationRomeItaly
| | - Jaime Ibanez
- Department of Clinical and Movement NeurosciencesUniversity College LondonLondonUK
- BSICoS group, I3A Institute and IIS AragónUniversity of ZaragozaZaragozaSpain
- Department of Bioengineering, Centre for NeurotechnologiesImperial College LondonLondonUK
| | - Lorenzo Rocchi
- Department of Clinical and Movement NeurosciencesUniversity College LondonLondonUK
- Department of Medical Sciences and Public HealthUniversity of CagliariCagliariItaly
| | - John Rothwell
- Department of Clinical and Movement NeurosciencesUniversity College LondonLondonUK
| |
Collapse
|
49
|
Fogarty MJ. Loss of larger hypoglossal motor neurons in aged Fischer 344 rats. Respir Physiol Neurobiol 2023:104092. [PMID: 37331418 DOI: 10.1016/j.resp.2023.104092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/06/2023] [Accepted: 06/15/2023] [Indexed: 06/20/2023]
Abstract
The intrinsic (longitudinal, transversalis and verticalis) and extrinsic (genioglossus, styloglossus, hyoglossus and geniohyoid) tongue muscles are innervated by hypoglossal motor neurons (MNs). Tongue muscle activations occur during many behaviors: maintaining upper airway patency, chewing, swallowing, vocalization, vomiting, coughing, sneezing and grooming/sexual activities. In the tongues of the elderly, reduced oral motor function and strength contribute to increased risk of obstructive sleep apnoea. Tongue muscle atrophy and weakness is also described in rats, yet hypoglossal MN numbers are unknown. In young (6-months, n=10) and old (24-months, n=8) female and male Fischer 344 (F344) rats, stereological assessment of hypoglossal MN numbers and surface areas were performed on 16µm Nissl-stained brainstem cryosections. We observed a robust loss of ~15% of hypoglossal MNs and a modest ~8% reduction in their surface areas with age. In the larger size tertile of hypoglossal MNs, age-associated loss of hypoglossal MNs approached ~30% These findings uncover a potential neurogenic locus of pathology for age-associated tongue dysfunctions.
Collapse
Affiliation(s)
- Matthew J Fogarty
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905.
| |
Collapse
|
50
|
Ghaffari LT, Trotti D, Haeusler AR. Differential response of C9orf72 transcripts following neuronal depolarization. iScience 2023; 26:106959. [PMID: 37332610 PMCID: PMC10272498 DOI: 10.1016/j.isci.2023.106959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 04/19/2023] [Accepted: 05/22/2023] [Indexed: 06/20/2023] Open
Abstract
The (G4C2)n nucleotide repeat expansion (NRE) mutation in C9orf72 is the most common genetic cause of ALS and FTD. The biological functions of C9orf72 are becoming understood, but it is unclear if this gene is regulated in a neural-specific manner. Neuronal activity is a crucial modifier of biological processes in health and neurodegenerative disease contexts. Here, we show that prolonged membrane depolarization in healthy human iPSC-cortical neurons leads to a significant downregulation of a transcript variant 3 (V3) of C9orf72, with a concomitant increase in variant 2 (V2), which leads to total C9orf72 RNA transcript levels remaining unchanged. However, the same response is not observed in cortical neurons derived from patients with the C9-NRE mutation. These findings reveal the impact of depolarization on C9orf72 transcripts, and how this response diverges in C9-NRE-carriers, which may have important implications in the underlying unique clinical associations of C9-NRE transcripts and disease pathogenesis.
Collapse
Affiliation(s)
- Layla T. Ghaffari
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Davide Trotti
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Aaron R. Haeusler
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|