1
|
Qiu Q, Yang M, Gong D, Liang H, Chen T. Potassium and calcium channels in different nerve cells act as therapeutic targets in neurological disorders. Neural Regen Res 2025; 20:1258-1276. [PMID: 38845230 PMCID: PMC11624876 DOI: 10.4103/nrr.nrr-d-23-01766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 02/18/2024] [Accepted: 04/07/2024] [Indexed: 07/31/2024] Open
Abstract
The central nervous system, information integration center of the body, is mainly composed of neurons and glial cells. The neuron is one of the most basic and important structural and functional units of the central nervous system, with sensory stimulation and excitation conduction functions. Astrocytes and microglia belong to the glial cell family, which is the main source of cytokines and represents the main defense system of the central nervous system. Nerve cells undergo neurotransmission or gliotransmission, which regulates neuronal activity via the ion channels, receptors, or transporters expressed on nerve cell membranes. Ion channels, composed of large transmembrane proteins, play crucial roles in maintaining nerve cell homeostasis. These channels are also important for control of the membrane potential and in the secretion of neurotransmitters. A variety of cellular functions and life activities, including functional regulation of the central nervous system, the generation and conduction of nerve excitation, the occurrence of receptor potential, heart pulsation, smooth muscle peristalsis, skeletal muscle contraction, and hormone secretion, are closely related to ion channels associated with passive transmembrane transport. Two types of ion channels in the central nervous system, potassium channels and calcium channels, are closely related to various neurological disorders, including Alzheimer's disease, Parkinson's disease, and epilepsy. Accordingly, various drugs that can affect these ion channels have been explored deeply to provide new directions for the treatment of these neurological disorders. In this review, we focus on the functions of potassium and calcium ion channels in different nerve cells and their involvement in neurological disorders such as Parkinson's disease, Alzheimer's disease, depression, epilepsy, autism, and rare disorders. We also describe several clinical drugs that target potassium or calcium channels in nerve cells and could be used to treat these disorders. We concluded that there are few clinical drugs that can improve the pathology these diseases by acting on potassium or calcium ions. Although a few novel ion-channel-specific modulators have been discovered, meaningful therapies have largely not yet been realized. The lack of target-specific drugs, their requirement to cross the blood-brain barrier, and their exact underlying mechanisms all need further attention. This review aims to explain the urgent problems that need research progress and provide comprehensive information aiming to arouse the research community's interest in the development of ion channel-targeting drugs and the identification of new therapeutic targets for that can increase the cure rate of nervous system diseases and reduce the occurrence of adverse reactions in other systems.
Collapse
Affiliation(s)
- Qing Qiu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
- Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu Province, China
| | - Mengting Yang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
- Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu Province, China
| | - Danfeng Gong
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
- Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu Province, China
| | - Haiying Liang
- Department of Pharmacy, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian Province, China
| | - Tingting Chen
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
- Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu Province, China
| |
Collapse
|
2
|
Zhang D, Xiang W, Liu J, Li W, Qiao Z, Wang K, Shao L. Design, synthesis, and structure-activity relationship of 5,7- dimethylbenzo[d]thiazoles as novel Kv7.2/7.3 activators with antiepileptic effects. Eur J Med Chem 2025; 292:117660. [PMID: 40315730 DOI: 10.1016/j.ejmech.2025.117660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 04/03/2025] [Accepted: 04/18/2025] [Indexed: 05/04/2025]
Abstract
The activation of neuronal Kv7 channels has emerged as an important therapeutic strategy for epilepsy due to their role in regulating neuronal excitability. Retigabine (RTG), a Kv7.2/7.3 channel activator, was previously approved for epilepsy treatment but was withdrawn in 2017 because of its side effects of ophthalmological and dermatological pigmentation. Despite this setback, Kv7.2/7.3 channel remains a promising target for the development of antiepileptic drugs (AEDs). Previous studies have attributed the toxic metabolic quinone/azaquinone diimines and associated blue discoloration of RTG to its electron-rich tri-amine aromatic scaffold. A common strategy to mitigate this toxicity involves removing the ortho-aniline moiety of RTG. In this study, we designed and synthesized a series of compounds based on dimethylbenzene heterocyclic scaffolds as Kv7.2/7.3 activators. Among them, compound 2c demonstrated improved efficacy in Rb+ efflux assays and exhibited comparable activity in whole-cell patch clamp recordings on Kv7.2/7.3 channels. Moreover, compound 2c was effective in both maximal electroshock seizure (MES) and subcutaneous pentylenetetrazol (sc-PTZ) mouse models, with ED50 values of 4.02 mg/kg and 43.17 mg/kg, respectively. The LD50 value of 2c in acute toxicity experiments was 340.35 mg/kg (95 % CI: 293.68-394.45) in mice. Additionally, 2c exhibited locomotor impairment with at TD50 of 48.93 mg/kg in an open field test and 49.25 mg/kg in a rotarod test. Compound 2c also demonstrated reasonable pharmacokinetic (PK) properties and blood-brain barrier (BBB) penetration, along with good photostability. Site-directed mutagenesis, combined with molecular docking, confirmed that 2c interacted with key residues (W236, F305, and L299) in the Kv7.2 channel. Our findings suggest that compound 2c is a promising lead compound with a novel scaffold as a Kv7.2/7.3 activator for the management of epilepsy.
Collapse
Affiliation(s)
- Denggao Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai, 201203, China
| | - Wei Xiang
- Departments of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao, 266073, China
| | - Jie Liu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai, 201203, China
| | - Wei Li
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai, 201203, China.
| | - Zhen Qiao
- Shandong Key Laboratory of Neurorehabilitation, School of Life Sciences and Health, University of Health and Rehabilitation Sciences, Qingdao, 266113, China; Qingdao Key Laboratory of Neurorehabilitation, University of Health and Rehabilitation Sciences, Qingdao, 266113, China.
| | - KeWei Wang
- Departments of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao, 266073, China; Institute of Innovative Drug Discovery, Qingdao University Medical College, 38 Dengzhou Road, Qingdao, 266021, China.
| | - Liming Shao
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai, 201203, China.
| |
Collapse
|
3
|
Viteri JA, Kerr NR, Brennan CD, Kick GR, Wang M, Ketabforoush A, Snyder HK, Moore PJ, Darvishi FB, Dashtmian AR, Ayyagari SN, Rich K, Zhu Y, Arnold WD. Targeting senescence in Amyotrophic Lateral Sclerosis: senolytic treatment improves neuromuscular function and preserves cortical excitability in a TDP-43 Q331K mouse model. RESEARCH SQUARE 2025:rs.3.rs-6081213. [PMID: 40196013 PMCID: PMC11975006 DOI: 10.21203/rs.3.rs-6081213/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder marked by progressive motor neuron degeneration in the primary motor cortex (PMC) and spinal cord. Aging is a key factor in ALS onset and progression, with evidence suggesting that biological aging-a process involving cellular decline- far outpaces chronological aging in ALS. This promotes senescent cell accumulation-marked by irreversible cell-cycle arrest, impaired apoptosis, and chronic inflammation-disrupting tissue homeostasis and impairing neuronal support functions. Thus, targeting senescence presents a novel therapeutic strategy for ALS. Here, we investigated the senolytic combination Dasatinib and Quercetin (D&Q) in TDP-43Q331K ALS mice. D&Q improved neuromuscular function and reduced plasma neurofilament light chain, a biomarker of axonal damage. The most pronounced improvement was the improved cortical excitability, accompanied by reductions in senescence and TDP-43 in the PMC. These findings highlight the potential of senolytics to mitigate ALS-related dysfunction, supporting their viability as a therapeutic strategy.
Collapse
Affiliation(s)
- Jose A Viteri
- Department of Physical Medicine and Rehabilitation, University of Missouri-Columbia, Columbia, MO USA; NextGen Precision Health, University of Missouri-Columbia, Columbia, MO USA
| | - Nathan R Kerr
- Department of Physical Medicine and Rehabilitation, University of Missouri-Columbia, Columbia, MO USA; NextGen Precision Health, University of Missouri-Columbia, Columbia, MO USA
| | - Charles D Brennan
- Department of Physical Medicine and Rehabilitation, University of Missouri-Columbia, Columbia, MO USA; NextGen Precision Health, University of Missouri-Columbia, Columbia, MO USA
| | - Grace R Kick
- Department of Ophthalmology, University of Missouri-Columbia, Columbia, MO USA
| | - Meifang Wang
- Department of Physical Medicine and Rehabilitation, University of Missouri-Columbia, Columbia, MO USA; NextGen Precision Health, University of Missouri-Columbia, Columbia, MO USA
| | - Arsh Ketabforoush
- Department of Physical Medicine and Rehabilitation, University of Missouri-Columbia, Columbia, MO USA; NextGen Precision Health, University of Missouri-Columbia, Columbia, MO USA
| | - Harper K Snyder
- Department of Physical Medicine and Rehabilitation, University of Missouri-Columbia, Columbia, MO USA; NextGen Precision Health, University of Missouri-Columbia, Columbia, MO USA
| | - Peter J Moore
- Department of Physical Medicine and Rehabilitation, University of Missouri-Columbia, Columbia, MO USA; NextGen Precision Health, University of Missouri-Columbia, Columbia, MO USA
| | - Fereshteh B Darvishi
- Department of Physical Medicine and Rehabilitation, University of Missouri-Columbia, Columbia, MO USA; NextGen Precision Health, University of Missouri-Columbia, Columbia, MO USA
| | - Anna Roshani Dashtmian
- Department of Physical Medicine and Rehabilitation, University of Missouri-Columbia, Columbia, MO USA; NextGen Precision Health, University of Missouri-Columbia, Columbia, MO USA
| | - Sindhuja N Ayyagari
- Department of Physical Medicine and Rehabilitation, University of Missouri-Columbia, Columbia, MO USA; NextGen Precision Health, University of Missouri-Columbia, Columbia, MO USA
| | - Kelly Rich
- Department of Genetics, Harvard Medical School, Boston, MA USA
| | - Yi Zhu
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA; Robert & Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - W David Arnold
- Department of Physical Medicine and Rehabilitation, University of Missouri-Columbia, Columbia, MO USA; NextGen Precision Health, University of Missouri-Columbia, Columbia, MO USA
| |
Collapse
|
4
|
Dierksen F, Geibel JS, Albrecht J, Hofer S, Dechent P, Hesse AC, Frahm J, Bähr M, Koch JC, Liman J, Maier IL. T1-relaxation times along the corticospinal tract as a diagnostic marker in patients with amyotrophic lateral sclerosis. FRONTIERS IN NEUROIMAGING 2025; 4:1549727. [PMID: 40017821 PMCID: PMC11865248 DOI: 10.3389/fnimg.2025.1549727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 01/28/2025] [Indexed: 03/01/2025]
Abstract
Background and purpose In the differential diagnostic workup of amyotrophic lateral sclerosis (ALS), magnetic resonance imaging (MRI) is primarily used to rule out significant differential diagnoses. So far, whole-brain T1-mapping has not been assessed as a diagnostic tool in this patient population. Methods We investigated the diagnostic potential of a novel T1-mapping method based on real-time MRI with 0.5 mm in-plane resolution and 4s acquisition time per slice. The study included patients aged 18 to 90 years who met the revised El Escorial criteria for at least possible ALS. T1-relaxation times were measured along the corticospinal tract in predefined regions of interest. Results Twenty-nine ALS-patients and 43 control group patients (CG) were included in the study. Median ALS Functional Rating Scale revised (ALSFRS-R) was 37 (IQR, 35-44) points and the mean duration from symptom onset to MRI was 21 ± 17 (SD) months. ALS patients showed significantly higher T1-relaxation times in all ROIs compared to CG with mean differences in the hand knob of 50 ms (p < 0.001), corona radiata 24 ms (p = 0.034), internal capsule 27 ms (p = 0.002) and midbrain peduncles 48 ms (p < 0.001). There was a consistent negative correlation between the ALSFRS-R and T1-relaxation times in all ROIs. Conclusions T1-relaxation times along the corticospinal tract are significantly elevated in ALS patients compared to CG and associated with lower ALSFRS-R. These results imply the analysis of T1-relaxation times as a promising diagnostic tool that can distinguish ALS patients from the control group. Ongoing longitudinal studies may provide deeper insights into disease progression and the effects of therapeutic interventions.
Collapse
Affiliation(s)
- Fiona Dierksen
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Johanna S. Geibel
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Janika Albrecht
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Sabine Hofer
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Peter Dechent
- Department of Cognitive Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Amelie C. Hesse
- Institute for Diagnostic and Interventional Radiology, University Medical Center Göttingen, Göttingen, Germany
| | - Jens Frahm
- Biomedical NMR, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Mathias Bähr
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Jan C. Koch
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Jan Liman
- Department of Neurology, Klinikum Nürnberg, Nürnberg, Germany
| | - Ilko L. Maier
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
5
|
Calancie B, Alexeeva N. Revisiting motor unit recruitment to TMS in amyotrophic lateral sclerosis: cortical inhibition is retained during voluntary contractions. Exp Brain Res 2025; 243:51. [PMID: 39843865 DOI: 10.1007/s00221-024-06961-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 10/10/2024] [Indexed: 01/24/2025]
Abstract
Transcranial magnetic stimulation (TMS) has been used for many years to study the pathophysiology of amyotrophic lateral sclerosis (ALS). Based on single- or dual-pulse TMS and EMG and/or single motor unit (MU) recordings, many groups have described a loss of central inhibition as an early marker of ALS dysfunction, reflecting a state of cortical 'hyperexcitability'. This conclusion is not without its detractors, however, leading us to reexamine this issue using 4-pulse TMS, shown previously to be more effective for testing central motor pathway functional integrity. A total of 221 motor units were tested in 13 subjects (6 controls; 7 with ALS) across a total of 798 unique TMS conditions. MUs were studied from hand muscles (usually first dorsal interosseus) and from tibialis anterior (TA). Subjects were required to recruit a MU to fire rhythmically, during which time 4-pulse trains of TMS were delivered. A given motor unit's recruitment was examined for different stimulus intensities and interpulse intervals (IPI). All motor units from control subjects showed short latency excitation to TMS, and short latency inhibition for TMS pulses of slightly weaker intensity (i.e. the threshold for inhibition was lower than that for excitation). The same was largely true for MUs studied in subjects with ALS, with the primary difference between control and ALS subjects being the need for stronger stimulus intensities to effect recruitment in subjects with ALS. We saw no evidence for a loss or reduction of inhibition of central motor output in persons with ALS, at least when tested during voluntary contractions.
Collapse
Affiliation(s)
- Blair Calancie
- Dept. of Neurosurgery, Upstate Medical University, 750 E. Adams St, Syracuse, NY, 13210, USA.
| | - Natalia Alexeeva
- Dept. of Neurosurgery, Upstate Medical University, 750 E. Adams St, Syracuse, NY, 13210, USA
| |
Collapse
|
6
|
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
|
7
|
Hui KK, Yamanaka S. iPS cell therapy 2.0: Preparing for next-generation regenerative medicine. Bioessays 2024; 46:e2400072. [PMID: 38922935 DOI: 10.1002/bies.202400072] [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: 03/27/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024]
Abstract
This year marks the tenth anniversary of the world's first transplantation of tissue generated from induced pluripotent stem cells (iPSCs). There is now a growing number of clinical trials worldwide examining the efficacy and safety of autologous and allogeneic iPSC-derived products for treating various pathologic conditions. As we patiently wait for the results from these and future clinical trials, it is imperative to strategize for the next generation of iPSC-based therapies. This review examines the lessons learned from the development of another advanced cell therapy, chimeric antigen receptor (CAR) T cells, and the possibility of incorporating various new bioengineering technologies in development, from RNA engineering to tissue fabrication, to apply iPSCs not only as a means to achieve personalized medicine but also as designer medical applications.
Collapse
Affiliation(s)
- Kelvin K Hui
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Shinya Yamanaka
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
- CiRA Foundation, Kyoto, Japan
- Gladstone Institute of Cardiovascular Disease, San Francisco, California, USA
| |
Collapse
|
8
|
Xie C, Kessi M, Yin F, Peng J. Roles of KCNA2 in Neurological Diseases: from Physiology to Pathology. Mol Neurobiol 2024; 61:8491-8517. [PMID: 38517617 DOI: 10.1007/s12035-024-04120-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 03/10/2024] [Indexed: 03/24/2024]
Abstract
Potassium voltage-gated channel subfamily a member 2 (Kv1.2, encoded by KCNA2) is highly expressed in the central and peripheral nervous systems. Based on the patch clamp studies, gain-of function (GOF), loss-of-function (LOF), and a mixed type (GOF/LOF) variants can cause different conditions/disorders. KCNA2-related neurological diseases include epilepsy, intellectual disability (ID), attention deficit/hyperactive disorder (ADHD), autism spectrum disorder (ASD), pain as well as autoimmune and movement disorders. Currently, the molecular mechanisms for the reported variants in causing diverse disorders are unknown. Consequently, this review brings up to date the related information regarding the structure and function of Kv1.2 channel, expression patterns, neuronal localizations, and tetramerization as well as important cell and animal models. In addition, it provides updates on human genetic variants, genotype-phenotype correlations especially highlighting the deep insight into clinical prognosis of KCNA2-related developmental and epileptic encephalopathy, mechanisms, and the potential treatment targets for all KCNA2-related neurological disorders.
Collapse
Affiliation(s)
- Changning Xie
- Department of Pediatrics, Xiangya Hospital, Central South University, Xiangya Road 87, Hunan, Changsha, 410008, China
| | - Miriam Kessi
- Department of Pediatrics, Xiangya Hospital, Central South University, Xiangya Road 87, Hunan, Changsha, 410008, China
| | - Fei Yin
- Department of Pediatrics, Xiangya Hospital, Central South University, Xiangya Road 87, Hunan, Changsha, 410008, China
| | - Jing Peng
- Department of Pediatrics, Xiangya Hospital, Central South University, Xiangya Road 87, Hunan, Changsha, 410008, China.
- Hunan Intellectual and Development Disabilities Research Center, Hunan, Changsha, 410008, China.
| |
Collapse
|
9
|
Rizea RE, Corlatescu AD, Costin HP, Dumitru A, Ciurea AV. Understanding Amyotrophic Lateral Sclerosis: Pathophysiology, Diagnosis, and Therapeutic Advances. Int J Mol Sci 2024; 25:9966. [PMID: 39337454 PMCID: PMC11432652 DOI: 10.3390/ijms25189966] [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: 08/01/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
This review offers an in-depth examination of amyotrophic lateral sclerosis (ALS), addressing its epidemiology, pathophysiology, clinical presentation, diagnostic techniques, and current as well as emerging treatments. The purpose is to condense key findings and illustrate the complexity of ALS, which is shaped by both genetic and environmental influences. We reviewed the literature to discuss recent advancements in understanding molecular mechanisms such as protein misfolding, mitochondrial dysfunction, oxidative stress, and axonal transport defects, which are critical for identifying potential therapeutic targets. Significant progress has been made in refining diagnostic criteria and identifying biomarkers, leading to earlier and more precise diagnoses. Although current drug treatments provide some benefits, there is a clear need for more effective therapies. Emerging treatments, such as gene therapy and stem cell therapy, show potential in modifying disease progression and improving the quality of life for ALS patients. The review emphasizes the importance of continued research to address challenges such as disease variability and the limited effectiveness of existing treatments. Future research should concentrate on further exploring the molecular foundations of ALS and developing new therapeutic approaches. The implications for clinical practice include ensuring the accessibility of new treatments and that healthcare systems are equipped to support ongoing research and patient care.
Collapse
Affiliation(s)
- Radu Eugen Rizea
- Department of Neurosurgery, University of Medicine and Pharmacy, "Carol Davila", 020021 Bucharest, Romania
- Department of Neurosurgery, "Bagdasar-Arseni" Clinical Emergency Hospital, 041915 Bucharest, Romania
| | - Antonio-Daniel Corlatescu
- Department of Neurosurgery, University of Medicine and Pharmacy, "Carol Davila", 020021 Bucharest, Romania
| | - Horia Petre Costin
- Department of Neurosurgery, University of Medicine and Pharmacy, "Carol Davila", 020021 Bucharest, Romania
| | - Adrian Dumitru
- Department of Neurosurgery, University of Medicine and Pharmacy, "Carol Davila", 020021 Bucharest, Romania
- Department of Morphopathology, University of Medicine and Pharmacy, "Carol Davila", 020021 Bucharest, Romania
- Emergency University Hospital Bucharest, 050098 Bucharest, Romania
| | - Alexandru Vlad Ciurea
- Department of Neurosurgery, University of Medicine and Pharmacy, "Carol Davila", 020021 Bucharest, Romania
- Sanador Clinical Hospital, 010991 Bucharest, Romania
| |
Collapse
|
10
|
Ren K, Wang Q, Jiang D, Liu E, Alsmaan J, Jiang R, Rutkove SB, Tian F. A comprehensive review of electrophysiological techniques in amyotrophic lateral sclerosis research. Front Cell Neurosci 2024; 18:1435619. [PMID: 39280794 PMCID: PMC11393746 DOI: 10.3389/fncel.2024.1435619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 08/08/2024] [Indexed: 09/18/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS), a devastating neurodegenerative disease, is characterized by progressive motor neuron degeneration, leading to widespread weakness and respiratory failure. While a variety of mechanisms have been proposed as causes of this disease, a full understanding remains elusive. Electrophysiological alterations, including increased motor axon excitability, likely play an important role in disease progression. There remains a critical need for non-animal disease models that can integrate electrophysiological tools to better understand underlying mechanisms, track disease progression, and evaluate potential therapeutic interventions. This review explores the integration of electrophysiological technologies with ALS disease models. It covers cellular and clinical electrophysiological tools and their applications in ALS research. Additionally, we examine conventional animal models and highlight advancements in humanized models and 3D organoid technologies. By bridging the gap between these models, we aim to enhance our understanding of ALS pathogenesis and facilitate the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Keyuan Ren
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Qinglong Wang
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Douglas Jiang
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Scripps Institution of Oceanography, San Diego, CA, United States
| | - Ethan Liu
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Julie Alsmaan
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- School of Arts and Science, Harvard College, Cambridge, MA, United States
| | - Rui Jiang
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- School of Arts and Science, Harvard College, Cambridge, MA, United States
| | - Seward B. Rutkove
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Feng Tian
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
11
|
Pain O, Jones A, Al Khleifat A, Agarwal D, Hramyka D, Karoui H, Kubica J, Llewellyn DJ, Ranson JM, Yao Z, Iacoangeli A, Al-Chalabi A. Harnessing transcriptomic signals for amyotrophic lateral sclerosis to identify novel drugs and enhance risk prediction. Heliyon 2024; 10:e35342. [PMID: 39170265 PMCID: PMC11336650 DOI: 10.1016/j.heliyon.2024.e35342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 07/19/2024] [Accepted: 07/26/2024] [Indexed: 08/23/2024] Open
Abstract
Introduction Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease. This study integrates common genetic association results from the latest ALS genome-wide association study (GWAS) summary statistics with functional genomic annotations with the aim of providing mechanistic insights into ALS risk loci, inferring drug repurposing opportunities, and enhancing prediction of ALS risk and clinical characteristics. Methods Genes associated with ALS were identified using GWAS summary statistic methodology including SuSiE SNP-based fine-mapping, and transcriptome- and proteome-wide association study (TWAS/PWAS) analyses. Using several approaches, gene associations were integrated with the DrugTargetor drug-gene interaction database to identify drugs that could be repurposed for the treatment of ALS. Furthermore, ALS gene associations from TWAS were combined with observed blood expression in two external ALS case-control datasets to calculate polytranscriptomic scores and evaluate their utility for prediction of ALS risk and clinical characteristics, including site of onset, age at onset, and survival. Results SNP-based fine-mapping, TWAS and PWAS identified 118 genes associated with ALS, with TWAS and PWAS providing novel mechanistic insights. Drug repurposing analyses identified six drugs significantly enriched for interactions with ALS associated genes, though directionality could not be determined. Additionally, drug class enrichment analysis showed gene signatures linked to calcium channel blockers may reduce ALS risk, whereas antiepileptic drugs may increase ALS risk. Across the two observed expression target samples, ALS polytranscriptomic scores significantly predicted ALS risk (R 2 = 5.1 %; p-value = 3.2 × 10-27) and clinical characteristics. Conclusions Functionally-informed analyses of ALS GWAS summary statistics identified novel mechanistic insights into ALS aetiology, highlighted several therapeutic research avenues, and enabled statistically significant prediction of ALS risk.
Collapse
Affiliation(s)
- Oliver Pain
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Ashley Jones
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Ahmad Al Khleifat
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Devika Agarwal
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, Old Road Campus, University of Oxford, Oxford, United Kingdom
| | - Dzmitry Hramyka
- Core Unit Bioinformatics (CUBI), Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Hajer Karoui
- Multiple Sclerosis and Parkinson's Tissue Bank, Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Jędrzej Kubica
- Laboratory of Structural Bioinformatics, Institute of Evolutionary Biology, University of Warsaw, Poland
- Laboratory of Theory of Biopolimers, Faculty of Chemistry, University of Warsaw, Poland
| | - David J. Llewellyn
- University of Exeter Medical School, Exeter, United Kingdom
- Alan Turing Institute, London, United Kingdom
| | | | - Zhi Yao
- LifeArc, Stevenage, United Kingdom
| | - Alfredo Iacoangeli
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
- National Institute for Health Research Biomedical Research Centre and Dementia Unit at South London and Maudsley NHS Foundation Trust and King's College London, London, United Kingdom
| | - Ammar Al-Chalabi
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| |
Collapse
|
12
|
Pavey N, Hannaford A, Higashihara M, van den Bos M, Geevasinga N, Vucic S, Menon P. Cortical inexcitability in ALS: correlating a clinical phenotype. J Neurol Neurosurg Psychiatry 2024; 96:jnnp-2024-333928. [PMID: 39137976 PMCID: PMC12015075 DOI: 10.1136/jnnp-2024-333928] [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: 03/26/2024] [Accepted: 07/23/2024] [Indexed: 08/15/2024]
Abstract
BACKGROUND Cortical inexcitability, a less studied feature of upper motor neuron (UMN) dysfunction in amyotrophic lateral sclerosis (ALS), was identified in a large cross-sectional cohort of ALS patients and their demographic and clinical characteristics were contrasted with normal or hyperexcitable ALS cohorts to assess the impact of cortical inexcitability on ALS phenotype and survival. METHODS Threshold-tracking transcranial magnetic stimulation (TMS) technique with measurement of mean short interval intracortical inhibition (SICI) differentiated ALS patients into three groups (1) inexcitable (no TMS response at maximal stimulator output in the setting of preserved lower motor neuron (LMN) function), (2) hyperexcitable (SICI≤5.5%) and (3) normal cortical excitability (SICI>5.5%). Clinical phenotyping and neurophysiological assessment of LMN function were undertaken, and survival was recorded in the entire cohort. RESULTS 417 ALS patients were recruited, of whom 26.4% exhibited cortical inexcitability. Cortical inexcitability was associated with a younger age of disease onset (p<0.05), advanced Awaji criteria (p<0.01) and Kings stage (p<0.01) scores. Additionally, patients with cortical inexcitability had higher UMN score (p<0.01), lower revised ALS Functional Rating Scale score (p<0.01) and reduced upper limb strength score (MRC UL, p<0.01). Patient survival (p=0.398) was comparable across the groups, despite lower riluzole use in the cortical inexcitability patient group (p<0.05). CONCLUSION The present study established that cortical inexcitability was associated with a phenotype characterised by prominent UMN signs, greater motor and functional decline, and a younger age of onset. The present findings inform patient management and could improve patient stratification in clinical trials.
Collapse
Affiliation(s)
- Nathan Pavey
- The University of Sydney, Sydney, New South Wales, Australia
- Department of Neurology, Concord Repatriation General Hospital, Sydney, New South Wales, Australia
| | - Andrew Hannaford
- The University of Sydney, Sydney, New South Wales, Australia
- Department of Neurology, Concord Repatriation General Hospital, Sydney, New South Wales, Australia
| | - Mana Higashihara
- Department of Neurology, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi-ku, Tokyo, Japan
| | - Mehdi van den Bos
- The University of Sydney, Sydney, New South Wales, Australia
- Department of Neurology, Concord Repatriation General Hospital, Sydney, New South Wales, Australia
| | - Nimeshan Geevasinga
- The University of Sydney, Sydney, New South Wales, Australia
- Westmead Hospital, Westmead, New South Wales, Australia
| | - Steve Vucic
- The University of Sydney, Sydney, New South Wales, Australia
- Department of Neurology, Concord Repatriation General Hospital, Sydney, New South Wales, Australia
| | - Parvathi Menon
- The University of Sydney, Sydney, New South Wales, Australia
- Department of Neurology, Concord Repatriation General Hospital, Sydney, New South Wales, Australia
| |
Collapse
|
13
|
Stikvoort García DJL, Goedee HS, van Eijk RPA, van Schelven LJ, van den Berg LH, Sleutjes BTHM. Revisiting distinct nerve excitability patterns in patients with amyotrophic lateral sclerosis. Brain 2024; 147:2842-2853. [PMID: 38662766 PMCID: PMC11491535 DOI: 10.1093/brain/awae131] [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: 11/17/2023] [Revised: 03/29/2024] [Accepted: 04/08/2024] [Indexed: 08/02/2024] Open
Abstract
Amyotrophic lateral sclerosis is a devastating neurodegenerative disease, characterized by loss of central and peripheral motor neurons. Although the disease is clinically and genetically heterogeneous, axonal hyperexcitability is a commonly observed feature that has been suggested to reflect an early pathophysiological step linked to the neurodegenerative cascade. Therefore, it is important to clarify the mechanisms causing axonal hyperexcitability and how these relate to the clinical characteristics of patients. Measures derived directly from a nerve excitability recording are frequently used as study end points, although their biophysical basis is difficult to deduce. Mathematical models can aid in the interpretation but are reliable only when applied to group-averaged recordings. Consequently, model estimates of membrane properties cannot be compared with clinical characteristics or treatment effects in individual patients, posing a considerable limitation in heterogeneous diseases, such as amyotrophic lateral sclerosis. To address these challenges, we revisited nerve excitability using a new pattern analysis-based approach (principal component analysis). We evaluated disease-specific patterns of excitability changes and established their biophysical origins. Based on the observed patterns, we developed new compound measures of excitability that facilitate the implementation of this approach in clinical settings. We found that excitability changes in amyotrophic lateral sclerosis patients (n = 161, median disease duration = 11 months) were characterized by four unique patterns compared with controls (n = 50, age and sex matched). These four patterns were best explained by changes in resting membrane potential (modulated by Na+/K+ currents), slow potassium and sodium currents (modulated by their gating kinetics) and refractory properties of the nerve. Consequently, we were able to show that altered gating of slow potassium channels was associated with, and predictive of, the rate of progression of the disease on the amyotrophic lateral sclerosis functional rating scale. Based on these findings, we designed four composite measures that capture these properties to facilitate implementation outside this study. Our findings demonstrate that changes in nerve excitability in patients with amyotrophic lateral sclerosis are dominated by four distinct patterns, each with a distinct biophysical origin. Based on this new approach, we provide evidence that altered slow potassium-channel function might play a role in the rate of disease progression. The magnitudes of these patterns, quantified using a similar approach or our new composite measures, have potential as efficient measures to study membrane properties directly in amyotrophic lateral sclerosis patients, and thus aid prognostic stratification and trial design.
Collapse
Affiliation(s)
| | - H Stephan Goedee
- Department of Neurology, Brain Centre Utrecht, University Medical Centre Utrecht, Utrecht, 3584CX, The Netherlands
| | - Ruben P A van Eijk
- Department of Neurology, Brain Centre Utrecht, University Medical Centre Utrecht, Utrecht, 3584CX, The Netherlands
- Biostatistics and Research Support, Julius Centre for Health Sciences and Primary Care, University Medical Centre Utrecht, Universiteitsweg 100, 3584CX, Utrecht, The Netherlands
| | - Leonard J van Schelven
- Department of Medical Technology and Clinical Physics, University Medical Centre Utrecht, 3584CX, Utrecht, The Netherlands
| | - Leonard H van den Berg
- Department of Neurology, Brain Centre Utrecht, University Medical Centre Utrecht, Utrecht, 3584CX, The Netherlands
| | - Boudewijn T H M Sleutjes
- Department of Neurology, Brain Centre Utrecht, University Medical Centre Utrecht, Utrecht, 3584CX, The Netherlands
| |
Collapse
|
14
|
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
|
15
|
Urena ES, Diezel CC, Serna M, Hala'ufia G, Majuta L, Barber KR, Vanderah TW, Riegel AC. K v7 channel opener retigabine reduces self-administration of cocaine but not sucrose in rats. Addict Biol 2024; 29:e13428. [PMID: 39087789 PMCID: PMC11292668 DOI: 10.1111/adb.13428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/17/2024] [Accepted: 07/03/2024] [Indexed: 08/02/2024]
Abstract
The increasing rates of drug misuse highlight the urgency of identifying improved therapeutics for treatment. Most drug-seeking behaviours that can be modelled in rodents utilize the repeated intravenous self-administration (SA) of drugs. Recent studies examining the mesolimbic pathway suggest that Kv7/KCNQ channels may contribute to the transition from recreational to chronic drug use. However, to date, all such studies used noncontingent, experimenter-delivered drug model systems, and the extent to which this effect generalizes to rats trained to self-administer drugs is not known. Here, we tested the ability of retigabine (ezogabine), a Kv7 channel opener, to regulate instrumental behaviour in male Sprague Dawley rats. We first validated the ability of retigabine to target experimenter-delivered cocaine in a conditioned place preference (CPP) assay and found that retigabine reduced the acquisition of place preference. Next, we trained rats for cocaine-SA under a fixed-ratio or progressive-ratio reinforcement schedule and found that retigabine pretreatment attenuated the SA of low to moderate doses of cocaine. This was not observed in parallel experiments, with rats self-administering sucrose, a natural reward. Compared with sucrose-SA, cocaine-SA was associated with reductions in the expression of the Kv7.5 subunit in the nucleus accumbens, without alterations in Kv7.2 and Kv7.3. Therefore, these studies reveal a reward-specific reduction in SA behaviour and support the notion that Kv7 is a potential therapeutic target for human psychiatric diseases with dysfunctional reward circuitry.
Collapse
Affiliation(s)
- Esteban S. Urena
- Department of Pharmacology, College of MedicineUniversity of ArizonaTucsonArizonaUSA
| | - Cody C. Diezel
- Department of Pharmacology, College of MedicineUniversity of ArizonaTucsonArizonaUSA
| | - Mauricio Serna
- Department of Pharmacology, College of MedicineUniversity of ArizonaTucsonArizonaUSA
| | - Grace Hala'ufia
- Department of Pharmacology, College of MedicineUniversity of ArizonaTucsonArizonaUSA
| | - Lisa Majuta
- Department of Pharmacology, College of MedicineUniversity of ArizonaTucsonArizonaUSA
| | - Kara R. Barber
- Department of Pharmacology, College of MedicineUniversity of ArizonaTucsonArizonaUSA
| | - Todd W. Vanderah
- Department of Pharmacology, College of MedicineUniversity of ArizonaTucsonArizonaUSA
- Neuroscience Graduate Interdisciplinary ProgramUniversity of ArizonaTucsonArizonaUSA
- Comprehensive Pain and Addiction‐Center (CPA‐C)University of Arizona Health SciencesTucsonArizonaUSA
- The Center of Excellence in Addiction Studies (CEAS)University of ArizonaTucsonArizonaUSA
| | - Arthur C. Riegel
- Department of Pharmacology, College of MedicineUniversity of ArizonaTucsonArizonaUSA
- Neuroscience Graduate Interdisciplinary ProgramUniversity of ArizonaTucsonArizonaUSA
- Comprehensive Pain and Addiction‐Center (CPA‐C)University of Arizona Health SciencesTucsonArizonaUSA
- The Center of Excellence in Addiction Studies (CEAS)University of ArizonaTucsonArizonaUSA
- Department of Neuroscience, College of ScienceUniversity of ArizonaTucsonArizonaUSA
- James C. Wyant College of Optical SciencesUniversity of ArizonaTucsonArizonaUSA
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
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
|
19
|
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
|
20
|
Limone F, Mordes DA, Couto A, Joseph BJ, Mitchell JM, Therrien M, Ghosh SD, Meyer D, Zhang Y, Goldman M, Bortolin L, Cobos I, Stevens B, McCarroll SA, Kadiu I, Burberry A, Pietiläinen O, Eggan K. Single-nucleus sequencing reveals enriched expression of genetic risk factors in extratelencephalic neurons sensitive to degeneration in ALS. NATURE AGING 2024; 4:984-997. [PMID: 38907103 PMCID: PMC11257952 DOI: 10.1038/s43587-024-00640-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 05/01/2024] [Indexed: 06/23/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder characterized by a progressive loss of motor function linked to degenerating extratelencephalic neurons/Betz cells (ETNs). The reasons why these neurons are selectively affected remain unclear. Here, to understand the unique molecular properties that may sensitize ETNs to ALS, we performed RNA sequencing of 79,169 single nuclei from cortices of patients and controls. In both patients and unaffected individuals, we found significantly higher expression of ALS risk genes in THY1+ ETNs, regardless of diagnosis. In patients, this was accompanied by the induction of genes involved in protein homeostasis and stress responses that were significantly induced in a wide collection of ETNs. Examination of oligodendroglial and microglial nuclei revealed patient-specific downregulation of myelinating genes in oligodendrocytes and upregulation of an endolysosomal reactive state in microglia. Our findings suggest that selective vulnerability of extratelencephalic neurons is partly connected to their intrinsic molecular properties sensitizing them to genetics and mechanisms of degeneration.
Collapse
Affiliation(s)
- Francesco Limone
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA.
| | - Daniel A Mordes
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Alexander Couto
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Brian J Joseph
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Jana M Mitchell
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Martine Therrien
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- FM Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - Sulagna Dia Ghosh
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Daniel Meyer
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Yingying Zhang
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Melissa Goldman
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Laura Bortolin
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Inma Cobos
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Beth Stevens
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- FM Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Steven A McCarroll
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Irena Kadiu
- Neuroinflammation Focus Area, UCB Pharma, Braine-l'Alleud, Belgium
| | - Aaron Burberry
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Olli Pietiläinen
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Kevin Eggan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
21
|
Sharma R, Khan Z, Mehan S, Das Gupta G, Narula AS. Unraveling the multifaceted insights into amyotrophic lateral sclerosis: Genetic underpinnings, pathogenesis, and therapeutic horizons. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2024; 794:108518. [PMID: 39491718 DOI: 10.1016/j.mrrev.2024.108518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 09/19/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
Amyotrophic Lateral Sclerosis (ALS), a progressive neurodegenerative disease, primarily impairs upper and lower motor neurons, leading to debilitating motor dysfunction and eventually respiratory failure, widely known as Lou Gehrig's disease. ALS presents with diverse symptomatology, including dysarthria, dysphagia, muscle atrophy, and hyperreflexia. The prevalence of ALS varies globally, with incidence rates ranging from 1.5 to 3.8 per 100,000 individuals, significantly affecting populations aged 45-80. A complex interplay of genetic and environmental factors underpins ALS pathogenesis. Key genetic contributors include mutations in chromosome 9 open reading frame 72 (C9ORF72), superoxide dismutase type 1 (SOD1), Fusedin sarcoma (FUS), and TAR DNA-binding protein (TARDBP) genes, accounting for a considerable fraction of both familial (fALS) and sporadic (sALS) cases. The disease mechanism encompasses aberrant protein folding, mitochondrial dysfunction, oxidative stress, excitotoxicity, and neuroinflammation, contributing to neuronal death. This review consolidates current insights into ALS's multifaceted etiology, highlighting the roles of environmental exposures (e.g., toxins, heavy metals) and their interaction with genetic predispositions. We emphasize the polygenic nature of ALS, where multiple genetic variations cumulatively influence disease susceptibility and progression. This aspect underscores the challenges in ALS diagnosis, which currently lacks specific biomarkers and relies on symptomatology and familial history. Therapeutic strategies for ALS, still in nascent stages, involve symptomatic management and experimental approaches targeting molecular pathways implicated in ALS pathology. Gene therapy, focusing on specific ALS mutations, and stem cell therapy emerge as promising avenues. However, effective treatments remain elusive, necessitating a deeper understanding of ALS's genetic architecture and the development of targeted therapies based on personalized medicine principles. This review aims to provide a comprehensive understanding of ALS, encouraging further research into its complex genetic underpinnings and the development of innovative, effective treatment modalities.
Collapse
Affiliation(s)
- Ramaish Sharma
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India (Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Zuber Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India (Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India (Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India.
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India (Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC 27516, USA
| |
Collapse
|
22
|
Shin-Yi Lin C, Howells J, Rutkove S, Nandedkar S, Neuwirth C, Noto YI, Shahrizaila N, Whittaker RG, Bostock H, Burke D, Tankisi H. Neurophysiological and imaging biomarkers of lower motor neuron dysfunction in motor neuron diseases/amyotrophic lateral sclerosis: IFCN handbook chapter. Clin Neurophysiol 2024; 162:91-120. [PMID: 38603949 DOI: 10.1016/j.clinph.2024.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 02/07/2024] [Accepted: 03/12/2024] [Indexed: 04/13/2024]
Abstract
This chapter discusses comprehensive neurophysiological biomarkers utilised in motor neuron disease (MND) and, in particular, its commonest form, amyotrophic lateral sclerosis (ALS). These encompass the conventional techniques including nerve conduction studies (NCS), needle and high-density surface electromyography (EMG) and H-reflex studies as well as novel techniques. In the last two decades, new methods of assessing the loss of motor units in a muscle have been developed, that are more convenient than earlier methods of motor unit number estimation (MUNE),and may use either electrical stimulation (e.g. MScanFit MUNE) or voluntary activation (MUNIX). Electrical impedance myography (EIM) is another novel approach for the evaluation that relies upon the application and measurement of high-frequency, low-intensity electrical current. Nerve excitability techniques (NET) also provide insights into the function of an axon and reflect the changes in resting membrane potential, ion channel dysfunction and the structural integrity of the axon and myelin sheath. Furthermore, imaging ultrasound techniques as well as magnetic resonance imaging are capable of detecting the constituents of morphological changes in the nerve and muscle. The chapter provides a critical description of the ability of each technique to provide neurophysiological insight into the complex pathophysiology of MND/ALS. However, it is important to recognise the strengths and limitations of each approach in order to clarify utility. These neurophysiological biomarkers have demonstrated reliability, specificity and provide additional information to validate and assess lower motor neuron dysfunction. Their use has expanded the knowledge about MND/ALS and enhanced our understanding of the relationship between motor units, axons, reflexes and other neural circuits in relation to clinical features of patients with MND/ALS at different stages of the disease. Taken together, the ultimate goal is to aid early diagnosis, distinguish potential disease mimics, monitor and stage disease progression, quantify response to treatment and develop potential therapeutic interventions.
Collapse
Affiliation(s)
- Cindy Shin-Yi Lin
- Faculty of Medicine and Health, Central Clinical School, Brain and Mind Centre, University of Sydney, Sydney 2006, Australia.
| | - James Howells
- Central Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Seward Rutkove
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sanjeev Nandedkar
- Natus Medical Inc, Middleton, Wisconsin, USA and Medical College of Wisconsin, Milwaukee, WI, USA
| | - Christoph Neuwirth
- Neuromuscular Diseases Unit/ALS Clinic, Kantonsspital, St. Gallen, Switzerland
| | - Yu-Ichi Noto
- Department of Neurology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Nortina Shahrizaila
- Division of Neurology, Department of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Roger G Whittaker
- Newcastle University Translational and Clinical Research Institute (NUTCRI), Newcastle University., Newcastle Upon Tyne, United Kingdom
| | - Hugh Bostock
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, Queen Square, WC1N 3BG, London, United Kingdom
| | - David Burke
- Central Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Hatice Tankisi
- Department of Clinical Neurophysiology, Aarhus University Hospital and Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
23
|
Evans LJ, O'Brien D, Shaw PJ. Current neuroprotective therapies and future prospects for motor neuron disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 176:327-384. [PMID: 38802178 DOI: 10.1016/bs.irn.2024.04.003] [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
Four medications with neuroprotective disease-modifying effects are now in use for motor neuron disease (MND). With FDA approvals for tofersen, relyvrio and edaravone in just the past year, 2022 ended a quarter of a century when riluzole was the sole such drug to offer to patients. The acceleration of approvals may mean we are witnessing the beginning of a step-change in how MND can be treated. Improvements in understanding underlying disease biology has led to more therapies being developed to target specific and multiple disease mechanisms. Consideration for how the pipeline of new therapeutic agents coming through in clinical and preclinical development can be more effectively evaluated with biomarkers, advances in patient stratification and clinical trial design pave the way for more successful translation for this archetypal complex neurodegenerative disease. While it must be cautioned that only slowed rates of progression have so far been demonstrated, pre-empting rapid neurodegeneration by using neurofilament biomarkers to signal when to treat, as is currently being trialled with tofersen, may be more effective for patients with known genetic predisposition to MND. Early intervention with personalized medicines could mean that for some patients at least, in future we may be able to substantially treat what is considered by many to be one of the most distressing diseases in medicine.
Collapse
Affiliation(s)
- Laura J Evans
- The Sheffield Institute for Translational Neuroscience, and the NIHR Sheffield Biomedical Research Centre, University of Sheffield, Sheffield, United Kingdom
| | - David O'Brien
- The Sheffield Institute for Translational Neuroscience, and the NIHR Sheffield Biomedical Research Centre, University of Sheffield, Sheffield, United Kingdom
| | - Pamela J Shaw
- The Sheffield Institute for Translational Neuroscience, and the NIHR Sheffield Biomedical Research Centre, University of Sheffield, Sheffield, United Kingdom.
| |
Collapse
|
24
|
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
|
25
|
Rogers M, Obergrussberger A, Kondratskyi A, Fertig N. Using automated patch clamp electrophysiology platforms in ion channel drug discovery: an industry perspective. Expert Opin Drug Discov 2024; 19:523-535. [PMID: 38481119 DOI: 10.1080/17460441.2024.2329104] [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: 11/30/2023] [Accepted: 03/06/2024] [Indexed: 04/25/2024]
Abstract
INTRODUCTION Automated patch clamp (APC) is now well established as a mature technology for ion channel drug discovery in academia, biotech and pharma companies, and in contract research organizations (CRO), for a variety of applications including channelopathy research, compound screening, target validation and cardiac safety testing. AREAS COVERED Ion channels are an important class of drugged and approved drug targets. The authors present a review of the current state of ion channel drug discovery along with new and exciting developments in ion channel research involving APC. This includes topics such as native and iPSC-derived cells in ion channel drug discovery, channelopathy research, organellar and biologics in ion channel drug discovery. EXPERT OPINION It is our belief that APC will continue to play a critical role in ion channel drug discovery, not only in 'classical' hit screening, target validation and cardiac safety testing, but extending these applications to include high throughput organellar recordings and optogenetics. In this way, with advancements in APC capabilities and applications, together with high resolution cryo-EM structures, ion channel drug discovery will be re-invigorated, leading to a growing list of ion channel ligands in clinical development.
Collapse
Affiliation(s)
- Marc Rogers
- Albion Drug Discovery Services Ltd, Cambridge, UK
| | | | | | | |
Collapse
|
26
|
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
|
27
|
De Cock L, Bercier V, Van Den Bosch L. New developments in pre-clinical models of ALS to guide translation. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 176:477-524. [PMID: 38802181 DOI: 10.1016/bs.irn.2024.04.008] [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
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder in which selective death of motor neurons leads to muscle weakness and paralysis. Most research has focused on understanding and treating monogenic familial forms, most frequently caused by mutations in SOD1, FUS, TARDBP and C9orf72, although ALS is mostly sporadic and without a clear genetic cause. Rodent models have been developed to study monogenic ALS, but despite numerous pre-clinical studies and clinical trials, few disease-modifying therapies are available. ALS is a heterogeneous disease with complex underlying mechanisms where several genes and molecular pathways appear to play a role. One reason for the high failure rate of clinical translation from the current models could be oversimplification in pre-clinical studies. Here, we review advances in pre-clinical models to better capture the heterogeneous nature of ALS and discuss the value of novel model systems to guide translation and aid in the development of precision medicine.
Collapse
Affiliation(s)
- Lenja De Cock
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Louvain-University of Leuven, Leuven, Belgium; Center for Brain and Disease Research, Laboratory of Neurobiology, VIB, Leuven, Belgium
| | - Valérie Bercier
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Louvain-University of Leuven, Leuven, Belgium; Center for Brain and Disease Research, Laboratory of Neurobiology, VIB, Leuven, Belgium
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Louvain-University of Leuven, Leuven, Belgium; Center for Brain and Disease Research, Laboratory of Neurobiology, VIB, Leuven, Belgium.
| |
Collapse
|
28
|
Ceccarelli L, Verriello L, Pauletto G, Valente M, Spadea L, Salati C, Zeppieri M, Ius T. The Role of Human Pluripotent Stem Cells in Amyotrophic Lateral Sclerosis: From Biological Mechanism to Practical Implications. FRONT BIOSCI-LANDMRK 2024; 29:114. [PMID: 38538275 DOI: 10.31083/j.fbl2903114] [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: 11/27/2023] [Revised: 01/02/2024] [Accepted: 01/24/2024] [Indexed: 03/28/2024]
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a neurodegenerative disorder, characterized by progressive loss of both upper and lower motor neurons, resulting in clinical features such as muscle weakness, paralysis, and ultimately, respiratory failure. Nowadays, there is not effective treatment to reverse the progression of the disease, that leads to death within 3-5 years after the onset. Nevertheless, the induced pluripotent stem cells (iPS) technology could be the answer, providing disease modelling, drug testing, and cell-based therapies for this pathology. The aim of this work was to conduct a literature review of the past 5 years about the role of iPS in ALS, to better define the neurobiological mechanisms involved in the pathogenesis and the potential future therapies. The review also deals with advanced and currently available technologies used to reprogram cell lines and generate human motor neurons in vitro, which represent the source to study the pathological processes, the relationship between phenotype and genotype, the disease progression and the potential therapeutic targets of these group of disorders. Specific treatment options with stem cells involve Advance Gene Editing Technology, neuroprotective agents, and cells or exosomes transplantation, aimed to replace dead or damaged nerve cells. In summary, this review comprehensively addresses the role of human pluripotent stem cells (hPSCs) in motor neuron diseases (MND), with a focus on physiopathology, diagnostic and prognostic implications, specific and potential future treatment options. Understanding the biological mechanisms and practical implications of hPSCs in MND is crucial for advancing therapeutic strategies and improving outcomes for patients affected by these devastating diseases.
Collapse
Affiliation(s)
- Laura Ceccarelli
- Clinical Neurology Unit, Head-Neck and Neurosciences Department, Santa Maria della Misericordia University Hospital, ASUFC, 33100 Udine, Italy
- Department of Medicine (DMED), Santa Maria della Misericordia University Hospital, ASUFC, 33100 Udine, Italy
| | - Lorenzo Verriello
- Neurology Unit, Head-Neck and Neurosciences Department, Santa Maria della Misericordia University Hospital, ASUFC, 33100 Udine, Italy
| | - Giada Pauletto
- Neurology Unit, Head-Neck and Neurosciences Department, Santa Maria della Misericordia University Hospital, ASUFC, 33100 Udine, Italy
| | - Mariarosaria Valente
- Clinical Neurology Unit, Head-Neck and Neurosciences Department, Santa Maria della Misericordia University Hospital, ASUFC, 33100 Udine, Italy
- Department of Medicine (DMED), Santa Maria della Misericordia University Hospital, ASUFC, 33100 Udine, Italy
| | - Leopoldo Spadea
- Eye Clinic, Policlinico Umberto I, "Sapienza" University of Rome, 00185 Rome, Italy
| | - Carlo Salati
- Department of Ophthalmology, University Hospital of Udine, 33100 Udine, Italy
| | - Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, 33100 Udine, Italy
| | - Tamara Ius
- Neurosurgery Unit, Head-Neck and Neurosciences Department, Santa Maria della Misericordia University Hospital, ASUFC, 33100 Udine, Italy
| |
Collapse
|
29
|
Marques C, Held A, Dorfman K, Sung J, Song C, Kavuturu AS, Aguilar C, Russo T, Oakley DH, Albers MW, Hyman BT, Petrucelli L, Lagier-Tourenne C, Wainger BJ. Neuronal STING activation in amyotrophic lateral sclerosis and frontotemporal dementia. Acta Neuropathol 2024; 147:56. [PMID: 38478117 PMCID: PMC10937762 DOI: 10.1007/s00401-024-02688-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 12/15/2023] [Accepted: 01/09/2024] [Indexed: 03/17/2024]
Abstract
The stimulator of interferon genes (STING) pathway has been implicated in neurodegenerative diseases, including Parkinson's disease and amyotrophic lateral sclerosis (ALS). While prior studies have focused on STING within immune cells, little is known about STING within neurons. Here, we document neuronal activation of the STING pathway in human postmortem cortical and spinal motor neurons from individuals affected by familial or sporadic ALS. This process takes place selectively in the most vulnerable cortical and spinal motor neurons but not in neurons that are less affected by the disease. Concordant STING activation in layer V cortical motor neurons occurs in a mouse model of C9orf72 repeat-associated ALS and frontotemporal dementia (FTD). To establish that STING activation occurs in a neuron-autonomous manner, we demonstrate the integrity of the STING signaling pathway, including both upstream activators and downstream innate immune response effectors, in dissociated mouse cortical neurons and neurons derived from control human induced pluripotent stem cells (iPSCs). Human iPSC-derived neurons harboring different familial ALS-causing mutations exhibit increased STING signaling with DNA damage as a main driver. The elevated downstream inflammatory markers present in ALS iPSC-derived neurons can be suppressed with a STING inhibitor. Our results reveal an immunophenotype that consists of innate immune signaling driven by the STING pathway and occurs specifically within vulnerable neurons in ALS/FTD.
Collapse
Affiliation(s)
- Christine Marques
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Aaron Held
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Katherine Dorfman
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, MA, USA
| | - Joon Sung
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, MA, USA
| | - Catherine Song
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, MA, USA
| | - Amey S Kavuturu
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, MA, USA
| | - Corey Aguilar
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, MA, USA
| | - Tommaso Russo
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, MA, USA
| | - Derek H Oakley
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Mark W Albers
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Alzheimer Disease Research Unit, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Bradley T Hyman
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Alzheimer Disease Research Unit, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | | | - Clotilde Lagier-Tourenne
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard University and MIT, Cambridge, MA, USA
| | - Brian J Wainger
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of Harvard University and MIT, Cambridge, MA, USA.
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
30
|
Masegosa VM, Navarro X, Herrando-Grabulosa M. ICA-27243 improves neuromuscular function and preserves motoneurons in the transgenic SOD1 G93A mice. Neurotherapeutics 2024; 21:e00319. [PMID: 38262101 DOI: 10.1016/j.neurot.2024.e00319] [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: 12/07/2023] [Accepted: 01/04/2024] [Indexed: 01/25/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease characterized by the death of upper and lower motor neurons (MNs). Excessive neuronal excitability has been implicated in MN degeneration; thus, modulation of hyperexcitability appears as a promising therapeutic strategy. Potassium channels are attractive targets since they can be activated at subthreshold voltages and can regulate neuronal excitability. In this study, we assayed the effects of N-(6-Chloro-pyridin-3-yl)-3,4-difluorobenzamide compound, known as ICA-27243, as a potential treatment for ALS. ICA-27243 is a highly selective Kv7.2/7.3 opener used mainly in epilepsy models. In the in vitro model of spinal cord organotypic cultures (SCOCs) exposed to acute excitotoxicity, ICA-27243 prevented MN degeneration at a dose-of 10 μM. Administration of ICA-27243 to transgenic SOD1G93A ALS mice improved the decline of neuromuscular function, maintained locomotion and coordination in the rotarod, decreased spinal MN death and attenuated glial reactivity. In conclusion, we report here for the first time that ICA-27243 is an effective treatment for ALS, emphasizing the potential of targeting Kv channels to reduce neuronal hyperexcitability.
Collapse
Affiliation(s)
- Vera M Masegosa
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience, Universitat Autònoma de Barcelona, Bellaterra, Spain; Centro de Investigación Biomédica en Red de Enfermedades Degenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Xavier Navarro
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience, Universitat Autònoma de Barcelona, Bellaterra, Spain; Centro de Investigación Biomédica en Red de Enfermedades Degenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Mireia Herrando-Grabulosa
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience, Universitat Autònoma de Barcelona, Bellaterra, Spain; Centro de Investigación Biomédica en Red de Enfermedades Degenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
31
|
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
|
32
|
Sharples SA, Broadhead MJ, Gray JA, Miles GB. M-type potassium currents differentially affect activation of motoneuron subtypes and tune recruitment gain. J Physiol 2023; 601:5751-5775. [PMID: 37988235 DOI: 10.1113/jp285348] [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: 07/27/2023] [Accepted: 10/25/2023] [Indexed: 11/23/2023] Open
Abstract
The size principle is a key mechanism governing the orderly recruitment of motor units and is believed to be dependent on passive properties of the constituent motoneurons. However, motoneurons are endowed with voltage-sensitive ion channels that create non-linearities in their input-output functions. Here we describe a role for the M-type potassium current, conducted by KCNQ channels, in the control of motoneuron recruitment in mice. Motoneurons were studied with whole-cell patch clamp electrophysiology in transverse spinal slices and identified based on delayed (fast) and immediate (slow) onsets of repetitive firing. M-currents were larger in delayed compared to immediate firing motoneurons, which was not reflected by variations in the presence of Kv7.2 or Kv7.3 subunits. Instead, a more depolarized spike threshold in delayed-firing motoneurons afforded a greater proportion of the total M-current to become activated within the subthreshold voltage range, which translated to a greater influence on their recruitment with little influence on their firing rates. Pharmacological activation of M-currents also influenced motoneuron recruitment at the population level, producing a rightward shift in the recruitment curve of monosynaptic reflexes within isolated mouse spinal cords. These results demonstrate a prominent role for M-type potassium currents in regulating the function of motor units, which occurs primarily through the differential control of motoneuron subtype recruitment. More generally, these findings highlight the importance of active properties mediated by voltage-sensitive ion channels in the differential control of motoneuron recruitment, which is a key mechanism for the gradation of muscle force. KEY POINTS: M-currents exert an inhibitory influence on spinal motor output. This inhibitory influence is exerted by controlling the recruitment, but not the firing rate, of high-threshold fast-like motoneurons, with limited influence on low-threshold slow-like motoneurons. Preferential control of fast motoneurons may be linked to a larger M-current that is activated within the subthreshold voltage range compared to slow motoneurons. Larger M-currents in fast compared to slow motoneurons are not accounted for by differences in Kv7.2 or Kv7.3 channel composition. The orderly recruitment of motoneuron subtypes is shaped by differences in the contribution of voltage-gated ion channels, including KCNQ channels. KCNQ channels may provide a target to dynamically modulate the recruitment gain across the motor pool and readily adjust movement vigour.
Collapse
Affiliation(s)
- Simon A Sharples
- School of Psychology and Neuroscience, University of St Andrews, Fife, UK
| | | | - James A Gray
- School of Psychology and Neuroscience, University of St Andrews, Fife, UK
| | - Gareth B Miles
- School of Psychology and Neuroscience, University of St Andrews, Fife, UK
| |
Collapse
|
33
|
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
|
34
|
Okano H, Morimoto S, Kato C, Nakahara J, Takahashi S. Induced pluripotent stem cells-based disease modeling, drug screening, clinical trials, and reverse translational research for amyotrophic lateral sclerosis. J Neurochem 2023; 167:603-614. [PMID: 37952981 DOI: 10.1111/jnc.16005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/09/2023] [Accepted: 10/19/2023] [Indexed: 11/14/2023]
Abstract
It has been more than 10 years since the hopes for disease modeling and drug discovery using induced pluripotent stem cell (iPSC) technology boomed. Recently, clinical trials have been conducted with drugs identified using this technology, and some promising results have been reported. For amyotrophic lateral sclerosis (ALS), a devastating neurodegenerative disease, several groups have identified candidate drugs, ezogabine (retigabine), bosutinib, and ropinirole, using iPSCs-based drug discovery, and clinical trials using these drugs have been conducted, yielding interesting results. In our previous study, an iPSCs-based drug repurposing approach was utilized to show the potential of ropinirole hydrochloride (ROPI) in reducing ALS-specific pathological phenotypes. Recently, a phase 1/2a trial was conducted to investigate the effects of ropinirole on ALS further. This double-blind, randomized, placebo-controlled study confirmed the safety and tolerability of and provided evidence of its ability to delay disease progression and prolong the time to respiratory failure in ALS patients. Furthermore, in the reverse translational research, in vitro characterization of patient-derived iPSCs-motor neurons (MNs) mimicked the therapeutic effects of ROPI in vivo, suggesting the potential application of this technology to the precision medicine of ALS. Interestingly, RNA-seq data showed that ROPI treatment suppressed the sterol regulatory element-binding protein 2-dependent cholesterol biosynthesis pathway. Therefore, this pathway may be involved in the therapeutic effect of ROPI on ALS. The possibility that this pathway may be involved in the therapeutic effect of ALS was demonstrated. Finally, new future strategies for ALS using iPSCs technology will be discussed in this paper.
Collapse
Affiliation(s)
- Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Satoru Morimoto
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Chris Kato
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Jin Nakahara
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Shinichi Takahashi
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
- Department of Neurology and Stroke, Saitama Medical University International Medical Center, Saitama, Japan
| |
Collapse
|
35
|
Maragakis NJ, de Carvalho M, Weiss MD. Therapeutic targeting of ALS pathways: Refocusing an incomplete picture. Ann Clin Transl Neurol 2023; 10:1948-1971. [PMID: 37641443 PMCID: PMC10647018 DOI: 10.1002/acn3.51887] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/04/2023] [Accepted: 08/14/2023] [Indexed: 08/31/2023] Open
Abstract
Numerous potential amyotrophic lateral sclerosis (ALS)-relevant pathways have been hypothesized and studied preclinically, with subsequent translation to clinical trial. However, few successes have been observed with only modest effects. Along with an improved but incomplete understanding of ALS as a neurodegenerative disease is the evolution of more sophisticated and diverse in vitro and in vivo preclinical modeling platforms, as well as clinical trial designs. We highlight proposed pathological pathways that have been major therapeutic targets for investigational compounds. It is likely that the failures of so many of these therapeutic compounds may not have occurred because of lack of efficacy but rather because of a lack of preclinical modeling that would help define an appropriate disease pathway, as well as a failure to establish target engagement. These challenges are compounded by shortcomings in clinical trial design, including lack of biomarkers that could predict clinical success and studies that are underpowered. Although research investments have provided abundant insights into new ALS-relevant pathways, most have not yet been developed more fully to result in clinical study. In this review, we detail some of the important, well-established pathways, the therapeutics targeting them, and the subsequent clinical design. With an understanding of some of the shortcomings in translational efforts over the last three decades of ALS investigation, we propose that scientists and clinicians may choose to revisit some of these therapeutic pathways reviewed here with an eye toward improving preclinical modeling, biomarker development, and the investment in more sophisticated clinical trial designs.
Collapse
Affiliation(s)
| | - Mamede de Carvalho
- Faculdade de MedicinaInsqatituto de Medicina Molecular João Lobo Antunes, Centro Académico de Medicina de Lisboa, Universidade de LisboaLisbonPortugal
| | - Michael D. Weiss
- Department of NeurologyUniversity of WashingtonSeattleWashingtonUSA
| |
Collapse
|
36
|
Tyberghein M, Janssen A, Wang FC. Strength-duration time constant and rheobase measurements: Comparison of the threshold tracking method and a manual procedure. Clin Neurophysiol 2023; 154:27-33. [PMID: 37541074 DOI: 10.1016/j.clinph.2023.06.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/22/2023] [Accepted: 06/29/2023] [Indexed: 08/06/2023]
Abstract
OBJECTIVE To compare the strength-duration time constant (SDTC) and rheobase measurements obtained by the threshold tracking method (TT) and by a non-automated method (MM). METHODS The MM procedure involved measuring, using a routine electrodiagnostic device, the intensity required to evoke a motor response whose amplitude corresponds to 40% of the maximum amplitude for four stimulus duration (1.0, 0.7, 0.5, 0.2 ms), and studying the linear relationship between stimulus charge and stimulus duration (slope = rheobase, intercept on the x-axis = SDTC). Using TT and MM, 30 successive healthy subjects (mean age = 38 years old) underwent a prospective evaluation of SDTC and rheobase of the median nerve motor axons at the wrist. Nerve stimulation and bipolar recording of evoked motor responses were performed with disposable self-adhesive surface electrodes. RESULTS The Spearman correlations between the two methods were 0.78 (p < 0.0001) for SDTC and 0.96 (p < 0.0001) for the rheobase. The Bland-Altman analysis did not reveal any systematic bias of MM compared to TT. CONCLUSIONS The MM procedure was reliable for strength-duration relationship analysis. SIGNIFICANCE We encourage neurophysiologists, who do not have dedicated threshold tracking equipment, not to hesitate to use these simple tools to assess peripheral nerve excitability.
Collapse
Affiliation(s)
- Maelle Tyberghein
- CHU Liège, Department of Neurophysiology, Sart Tilman B35, 4000 Liège, Belgium
| | - Arnaud Janssen
- CHU Liège, Department of Neurophysiology, Sart Tilman B35, 4000 Liège, Belgium
| | | |
Collapse
|
37
|
Stikvoort García DJL, Sleutjes BTHM, van Schelven LJ, Goedee HS, van den Berg LH. Diagnostic accuracy of nerve excitability and compound muscle action potential scan derived biomarkers in amyotrophic lateral sclerosis. Eur J Neurol 2023; 30:3068-3078. [PMID: 37354059 DOI: 10.1111/ene.15954] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 06/26/2023]
Abstract
BACKGROUND AND PURPOSE The lack of reliable early biomarkers still causes substantial diagnostic delays in amyotrophic lateral sclerosis (ALS). The aim was to assess the diagnostic accuracy of a novel electrophysiological protocol in patients with suspected motor neuron disease (MND). METHODS Consecutive patients with suspected MND were prospectively recruited at our tertiary referral centre for MND in Utrecht, The Netherlands. Procedures were performed in accordance with the Standards for Reporting of Diagnostic Accuracy. In addition to the standard diagnostic workup, an electrophysiological protocol of compound muscle action potential (CMAP) scans and nerve excitability tests was performed on patients' thenar muscles. The combined diagnostic yield of nerve excitability and CMAP scan based motor unit number estimation was compared to the Awaji and Gold Coast criteria and their added value was determined. RESULTS In all, 153 ALS or progressive muscular atrophy patients, 63 disease controls and 43 healthy controls were included. Our electrophysiological protocol had high diagnostic accuracy (area under the curve [AUC] 0.85, 95% confidence interval [95% CI] 0.80-0.90), even in muscles with undetectable axon loss (AUC 0.78, 95% CI 0.70-0.85) and in bulbar-onset patients (AUC 0.85, 95% CI 0.73-0.95). Twenty-four of 33 (73%) ALS patients who could not be diagnosed during the same visit were correctly identified, as well as 8/13 (62%) ALS patients not meeting the Gold Coast criteria and 49/59 (83%) ALS patients not meeting the Awaji criteria during this first visit. CONCLUSIONS Our practical and non-invasive electrophysiological protocol may improve early diagnosis in clinically challenging patients with suspected ALS. Routine incorporation may boost early diagnosis, enhance patient selection and generate baseline measures for clinical trials.
Collapse
Affiliation(s)
- D J L Stikvoort García
- Department of Neurology, Brain Center Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands
| | - B T H M Sleutjes
- Department of Neurology, Brain Center Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands
| | - L J van Schelven
- Department of Medical Technology and Clinical Physics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - H S Goedee
- Department of Neurology, Brain Center Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands
| | - L H van den Berg
- Department of Neurology, Brain Center Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
38
|
Held A, Adler M, Marques C, Reyes CJ, Kavuturu AS, Quadros ARAA, Ndayambaje IS, Lara E, Ward M, Lagier-Tourenne C, Wainger BJ. iPSC motor neurons, but not other derived cell types, capture gene expression changes in postmortem sporadic ALS motor neurons. Cell Rep 2023; 42:113046. [PMID: 37651231 PMCID: PMC10622181 DOI: 10.1016/j.celrep.2023.113046] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 05/10/2023] [Accepted: 08/11/2023] [Indexed: 09/02/2023] Open
Abstract
Motor neuron degeneration, the defining feature of amyotrophic lateral sclerosis (ALS), is a primary example of cell-type specificity in neurodegenerative diseases. Using isogenic pairs of induced pluripotent stem cells (iPSCs) harboring different familial ALS mutations, we assess the capacity of iPSC-derived lower motor neurons, sensory neurons, astrocytes, and superficial cortical neurons to capture disease features including transcriptional and splicing dysregulation observed in human postmortem neurons. At early time points, differentially regulated genes in iPSC-derived lower motor neurons, but not other cell types, overlap with one-third of the differentially regulated genes in laser-dissected motor neurons from ALS compared with control postmortem spinal cords. For genes altered in both the iPSC model and bona fide human lower motor neurons, expression changes correlate between the two populations. In iPSC-derived lower motor neurons, but not other derived cell types, we detect the downregulation of genes affected by TDP-43-dependent splicing. This reduction takes place exclusively within genotypes known to involve TDP-43 pathology.
Collapse
Affiliation(s)
- Aaron Held
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Michelle Adler
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Christine Marques
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Charles Jourdan Reyes
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; The Collaborative Center for X-Linked Dystonia-Parkinsonism, Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Amey S Kavuturu
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Ana R A A Quadros
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - I Sandra Ndayambaje
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Erika Lara
- iPSC Neurodegenerative Research Initiative, Center for Alzheimer's and Related Dementias, National Institute on Aging, NIH, Bethesda, MD 20892, USA
| | - Michael Ward
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Clotilde Lagier-Tourenne
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Broad Institute of Harvard University and MIT, Cambridge MA 02142, USA
| | - Brian J Wainger
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Broad Institute of Harvard University and MIT, Cambridge MA 02142, USA; Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston MA 02114, USA; Harvard Stem Cell Institute, Cambridge MA 02138, USA.
| |
Collapse
|
39
|
Tsehay Y, Zeng Y, Weber-Levine C, Awosika T, Kerensky M, Hersh AM, Ou Z, Jiang K, Bhimreddy M, Bauer SJ, Theodore JN, Quiroz VM, Suk I, Alomari S, Sun J, Tong S, Thakor N, Doloff JC, Theodore N, Manbachi A. Low-Intensity Pulsed Ultrasound Neuromodulation of a Rodent's Spinal Cord Suppresses Motor Evoked Potentials. IEEE Trans Biomed Eng 2023; 70:1992-2001. [PMID: 37018313 PMCID: PMC10510849 DOI: 10.1109/tbme.2022.3233345] [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] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Here we investigate the ability of low-intensity ultrasound (LIUS) applied to the spinal cord to modulate the transmission of motor signals. METHODS Male adult Sprague-Dawley rats (n = 10, 250-300 g, 15 weeks old) were used in this study. Anesthesia was initially induced with 2% isoflurane carried by oxygen at 4 L/min via a nose cone. Cranial, upper extremity, and lower extremity electrodes were placed. A thoracic laminectomy was performed to expose the spinal cord at the T11 and T12 vertebral levels. A LIUS transducer was coupled to the exposed spinal cord, and motor evoked potentials (MEPs) were acquired each minute for either 5- or 10-minutes of sonication. Following the sonication period, the ultrasound was turned off and post-sonication MEPs were acquired for an additional 5 minutes. RESULTS Hindlimb MEP amplitude significantly decreased during sonication in both the 5- (p < 0.001) and 10-min (p = 0.004) cohorts with a corresponding gradual recovery to baseline. Forelimb MEP amplitude did not demonstrate any statistically significant changes during sonication in either the 5- (p = 0.46) or 10-min (p = 0.80) trials. CONCLUSION LIUS applied to the spinal cord suppresses MEP signals caudal to the site of sonication, with recovery of MEPs to baseline after sonication. SIGNIFICANCE LIUS can suppress motor signals in the spinal cord and may be useful in treating movement disorders driven by excessive excitation of spinal neurons.
Collapse
|
40
|
Hosaka T, Tsuji H, Kwak S. Roles of Aging, Circular RNAs, and RNA Editing in the Pathogenesis of Amyotrophic Lateral Sclerosis: Potential Biomarkers and Therapeutic Targets. Cells 2023; 12:1443. [PMID: 37408276 DOI: 10.3390/cells12101443] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/18/2023] [Accepted: 05/19/2023] [Indexed: 07/07/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an incurable motor neuron disease caused by upper and lower motor neuron death. Despite advances in our understanding of ALS pathogenesis, effective treatment for this fatal disease remains elusive. As aging is a major risk factor for ALS, age-related molecular changes may provide clues for the development of new therapeutic strategies. Dysregulation of age-dependent RNA metabolism plays a pivotal role in the pathogenesis of ALS. In addition, failure of RNA editing at the glutamine/arginine (Q/R) site of GluA2 mRNA causes excitotoxicity due to excessive Ca2+ influx through Ca2+-permeable α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors, which is recognized as an underlying mechanism of motor neuron death in ALS. Circular RNAs (circRNAs), a circular form of cognate RNA generated by back-splicing, are abundant in the brain and accumulate with age. Hence, they are assumed to play a role in neurodegeneration. Emerging evidence has demonstrated that age-related dysregulation of RNA editing and changes in circRNA expression are involved in ALS pathogenesis. Herein, we review the potential associations between age-dependent changes in circRNAs and RNA editing, and discuss the possibility of developing new therapies and biomarkers for ALS based on age-related changes in circRNAs and dysregulation of RNA editing.
Collapse
Affiliation(s)
- Takashi Hosaka
- Department of Neurology, Division of Clinical Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan
- University of Tsukuba Hospital/Jichi Medical University Joint Ibaraki Western Regional Clinical Education Center, Chikusei 308-0813, Japan
- Department of Internal Medicine, Ibaraki Western Medical Center, Chikusei 308-0813, Japan
| | - Hiroshi Tsuji
- Department of Neurology, Division of Clinical Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan
| | - Shin Kwak
- Department of Neurology, Tokyo Medical University, Tokyo 160-0023, Japan
| |
Collapse
|
41
|
Kondo T, Ebinuma I, Tanaka H, Nishikawa Y, Komiya T, Ishikawa M, Okano H. Rapid and Robust Multi-Phenotypic Assay System for ALS Using Human iPS Cells with Mutations in Causative Genes. Int J Mol Sci 2023; 24:ijms24086987. [PMID: 37108151 PMCID: PMC10138792 DOI: 10.3390/ijms24086987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/06/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a major life-threatening disease caused by motor neuron degeneration. More effective treatments through drug discovery are urgently needed. Here, we established an effective high-throughput screening system using induced pluripotent stem cells (iPSCs). Using a Tet-On-dependent transcription factor expression system carried on the PiggyBac vector, motor neurons were efficiently and rapidly generated from iPSCs by a single-step induction method. Induced iPSC transcripts displayed characteristics similar to those of spinal cord neurons. iPSC-generated motor neurons carried a mutation in fused in sarcoma (FUS) and superoxide dismutase 1 (SOD1) genes and had abnormal protein accumulation corresponding to each mutation. Calcium imaging and multiple electrode array (MEA) recordings demonstrated that ALS neurons were abnormally hyperexcitable. Noticeably, protein accumulation and hyperexcitability were ameliorated by treatment with rapamycin (mTOR inhibitor) and retigabine (Kv7 channel activator), respectively. Furthermore, rapamycin suppressed ALS neuronal death and hyperexcitability, suggesting that protein aggregate clearance through the activation of autophagy effectively normalized activity and improved neuronal survival. Our culture system reproduced several ALS phenotypes, including protein accumulation, hyperexcitability, and neuronal death. This rapid and robust phenotypic screening system will likely facilitate the discovery of novel ALS therapeutics and stratified and personalized medicine for sporadic motor neuron diseases.
Collapse
Affiliation(s)
- Tosho Kondo
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
- Research Center of Neurology, Ono Pharmaceutical Co., Ltd., Osaka 618-8585, Japan
| | - Ihori Ebinuma
- Research Center of Neurology, Ono Pharmaceutical Co., Ltd., Osaka 618-8585, Japan
| | - Hirotaka Tanaka
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
- Research Center of Neurology, Ono Pharmaceutical Co., Ltd., Osaka 618-8585, Japan
| | - Yukitoshi Nishikawa
- Research Center of Neurology, Ono Pharmaceutical Co., Ltd., Osaka 618-8585, Japan
| | - Takaki Komiya
- Research Center of Neurology, Ono Pharmaceutical Co., Ltd., Osaka 618-8585, Japan
| | - Mitsuru Ishikawa
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
- Department of Clinical Regenerative Medicine, Fujita Health University School of Medicine, 1-98, Dengakugakubo, Kutsukake-cho, Toyoake 470-1192, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
- International Center for Brain Science, Fujita Health University, 1-98, Dengakugakubo, Kutsukake-cho, Toyoake 470-1192, Japan
| |
Collapse
|
42
|
Marshall KL, Rajbhandari L, Venkatesan A, Maragakis NJ, Farah MH. Enhanced axonal regeneration of ALS patient iPSC-derived motor neurons harboring SOD1 A4V mutation. Sci Rep 2023; 13:5597. [PMID: 37020097 PMCID: PMC10076424 DOI: 10.1038/s41598-023-31720-7] [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: 12/19/2022] [Accepted: 03/16/2023] [Indexed: 04/07/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease, characterized by degeneration of upper and lower motor neurons that leads to muscle weakness, paralysis, and death, but the effects of disease-causing mutations on axonal outgrowth of neurons derived from human induced pluripotent stem cells (iPSC)-derived motor neurons (hiPSC-MN) are poorly understood. The use of hiPSC-MN is a promising tool to develop more relevant models for target identification and drug development in ALS research, but questions remain concerning the effects of distinct disease-causing mutations on axon regeneration. Mutations in superoxide dismutase 1 (SOD1) were the first to be discovered in ALS patients. Here, we investigated the effect of the SOD1A4V mutation on axonal regeneration of hiPSC-MNs, utilizing compartmentalized microfluidic devices, which are powerful tools for studying hiPSC-MN distal axons. Surprisingly, SOD1+/A4V hiPSC-MNs regenerated axons more quickly following axotomy than those expressing the native form of SOD1. Though initial axon regrowth was not significantly different following axotomy, enhanced regeneration was apparent at later time points, indicating an increased rate of outgrowth. This regeneration model could be used to identify factors that enhance the rate of human axon regeneration.
Collapse
Affiliation(s)
- Katherine L Marshall
- Neuromuscular Division, Department of Neurology, Johns Hopkins University School of Medicine, The John G. Rangos Sr. Building, Room 239, 855 N. Wolfe Street, Baltimore, MD, 21205, USA
| | - Labchan Rajbhandari
- Neuromuscular Division, Department of Neurology, Johns Hopkins University School of Medicine, The John G. Rangos Sr. Building, Room 239, 855 N. Wolfe Street, Baltimore, MD, 21205, USA
| | - Arun Venkatesan
- Neuromuscular Division, Department of Neurology, Johns Hopkins University School of Medicine, The John G. Rangos Sr. Building, Room 239, 855 N. Wolfe Street, Baltimore, MD, 21205, USA
| | - Nicholas J Maragakis
- Neuromuscular Division, Department of Neurology, Johns Hopkins University School of Medicine, The John G. Rangos Sr. Building, Room 239, 855 N. Wolfe Street, Baltimore, MD, 21205, USA
| | - Mohamed H Farah
- Neuromuscular Division, Department of Neurology, Johns Hopkins University School of Medicine, The John G. Rangos Sr. Building, Room 239, 855 N. Wolfe Street, Baltimore, MD, 21205, USA.
| |
Collapse
|
43
|
Kapell H, Fazio L, Dyckow J, Schwarz S, Cruz-Herranz A, Mayer C, Campos J, D’Este E, Möbius W, Cordano C, Pröbstel AK, Gharagozloo M, Zulji A, Narayanan Naik V, Delank A, Cerina M, Müntefering T, Lerma-Martin C, Sonner JK, Sin JH, Disse P, Rychlik N, Sabeur K, Chavali M, Srivastava R, Heidenreich M, Fitzgerald KC, Seebohm G, Stadelmann C, Hemmer B, Platten M, Jentsch TJ, Engelhardt M, Budde T, Nave KA, Calabresi PA, Friese MA, Green AJ, Acuna C, Rowitch DH, Meuth SG, Schirmer L. Neuron-oligodendrocyte potassium shuttling at nodes of Ranvier protects against inflammatory demyelination. J Clin Invest 2023; 133:e164223. [PMID: 36719741 PMCID: PMC10065072 DOI: 10.1172/jci164223] [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: 08/09/2022] [Accepted: 01/27/2023] [Indexed: 02/01/2023] Open
Abstract
Multiple sclerosis (MS) is a progressive inflammatory demyelinating disease of the CNS. Increasing evidence suggests that vulnerable neurons in MS exhibit fatal metabolic exhaustion over time, a phenomenon hypothesized to be caused by chronic hyperexcitability. Axonal Kv7 (outward-rectifying) and oligodendroglial Kir4.1 (inward-rectifying) potassium channels have important roles in regulating neuronal excitability at and around the nodes of Ranvier. Here, we studied the spatial and functional relationship between neuronal Kv7 and oligodendroglial Kir4.1 channels and assessed the transcriptional and functional signatures of cortical and retinal projection neurons under physiological and inflammatory demyelinating conditions. We found that both channels became dysregulated in MS and experimental autoimmune encephalomyelitis (EAE), with Kir4.1 channels being chronically downregulated and Kv7 channel subunits being transiently upregulated during inflammatory demyelination. Further, we observed that pharmacological Kv7 channel opening with retigabine reduced neuronal hyperexcitability in human and EAE neurons, improved clinical EAE signs, and rescued neuronal pathology in oligodendrocyte-Kir4.1-deficient (OL-Kir4.1-deficient) mice. In summary, our findings indicate that neuron-OL compensatory interactions promoted resilience through Kv7 and Kir4.1 channels and identify pharmacological activation of nodal Kv7 channels as a neuroprotective strategy against inflammatory demyelination.
Collapse
Affiliation(s)
- Hannah Kapell
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Luca Fazio
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster (UKM), Münster, Germany
- Department of Neurology, University of Düsseldorf, Dusseldorf, Germany
| | - Julia Dyckow
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Sophia Schwarz
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Andrés Cruz-Herranz
- Weill Institute for Neurosciences, Department of Neurology, UCSF, San Francisco, California, USA
| | - Christina Mayer
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Joaquin Campos
- Chica and Heinz Schaller Research Group, Institute of Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Elisa D’Este
- Optical Microscopy Facility, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Wiebke Möbius
- Electron Microscopy Core Unit, Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
- Cluster of Excellence, “Multiscale Bioimaging: from Molecular Machines to Network of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
| | - Christian Cordano
- Weill Institute for Neurosciences, Department of Neurology, UCSF, San Francisco, California, USA
| | - Anne-Katrin Pröbstel
- Weill Institute for Neurosciences, Department of Neurology, UCSF, San Francisco, California, USA
- Neurologic Clinic and Policlinic and Research Center for Clinical Neuroimmunology and Neuroscience Basel, Departments of Medicine, Biomedicine, and Clinical Research, University Hospital of Basel, University of Basel, Basel, Switzerland
| | - Marjan Gharagozloo
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Amel Zulji
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Venu Narayanan Naik
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster (UKM), Münster, Germany
| | - Anna Delank
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster (UKM), Münster, Germany
| | - Manuela Cerina
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster (UKM), Münster, Germany
| | | | - Celia Lerma-Martin
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Jana K. Sonner
- Chica and Heinz Schaller Research Group, Institute of Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Jung Hyung Sin
- Weill Institute for Neurosciences, Department of Neurology, UCSF, San Francisco, California, USA
| | - Paul Disse
- Institute for Genetics of Heart Diseases (IfGH), Cellular Electrophysiology and Molecular Biology, UKM, Münster, Germany
- University of Münster, Chembion, Münster, Germany
| | - Nicole Rychlik
- University of Münster, Chembion, Münster, Germany
- Institute of Physiology I, University of Münster, Münster, Germany
| | - Khalida Sabeur
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research and
- Department of Pediatrics, UCSF, San Francisco, California, USA
| | - Manideep Chavali
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research and
- Department of Pediatrics, UCSF, San Francisco, California, USA
| | - Rajneesh Srivastava
- Department of Neurology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Matthias Heidenreich
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) and Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | - Kathryn C. Fitzgerald
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Guiscard Seebohm
- Institute for Genetics of Heart Diseases (IfGH), Cellular Electrophysiology and Molecular Biology, UKM, Münster, Germany
| | - Christine Stadelmann
- Electron Microscopy Core Unit, Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
- Institute of Neuropathology, University Medical Center, Göttingen, Germany
| | - Bernhard Hemmer
- Department of Neurology, School of Medicine, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology, Munich, Germany
| | - Michael Platten
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), INF 280, Heidelberg, Germany
- Interdisciplinary Center for Neurosciences (IZN) and
- Mannheim Center for Translational Neuroscience and Institute for Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Thomas J. Jentsch
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) and Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
- Neurocure Cluster of Excellence, Charité University Medicine Berlin, Berlin, Germany
| | - Maren Engelhardt
- Mannheim Center for Translational Neuroscience and Institute for Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Institute of Neuroanatomy, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Institute of Anatomy and Cell Biology, Johannes Kepler University Linz, Linz, Austria
| | - Thomas Budde
- Institute of Physiology I, University of Münster, Münster, Germany
| | - Klaus-Armin Nave
- Electron Microscopy Core Unit, Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Peter A. Calabresi
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Manuel A. Friese
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Ari J. Green
- Weill Institute for Neurosciences, Department of Neurology, UCSF, San Francisco, California, USA
- Department of Ophthalmology, UCSF, San Francisco, California, USA
| | - Claudio Acuna
- Chica and Heinz Schaller Research Group, Institute of Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - David H. Rowitch
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research and
- Department of Pediatrics, UCSF, San Francisco, California, USA
- Wellcome Trust–Medical Research Council Stem Cell Institute and
- Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom
- Department of Neurosurgery, UCSF, San Francisco, California, USA
| | - Sven G. Meuth
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster (UKM), Münster, Germany
- Department of Neurology, University of Düsseldorf, Dusseldorf, Germany
| | - Lucas Schirmer
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Interdisciplinary Center for Neurosciences (IZN) and
- Mannheim Center for Translational Neuroscience and Institute for Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
44
|
Antel JP, Kennedy TE, Kuhlmann T. Seeking neuroprotection in multiple sclerosis: an ongoing challenge. J Clin Invest 2023; 133:168595. [PMID: 37009896 PMCID: PMC10065069 DOI: 10.1172/jci168595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease of the CNS, featuring inflammation and demyelination with variable recovery. In this issue of the JCI, Kapell, Fazio, and authors address the potential for targeting neuron-oligodendrocyte potassium shuttling at the nodes of Ranvier as a neuroprotective strategy during inflammatory demyelination of the CNS in experimental MS. Their extensive and impressive study could serve as a template for defining the physiologic properties of a putative protective pathway. The authors examined MS features in existent disease models, investigated the impact of pharmacologic intervention, and evaluated its status in tissues from patients with MS. We await future studies that will tackle the challenge of translating these findings into a clinical therapy.
Collapse
Affiliation(s)
- Jack P Antel
- Montreal Neurological Institute, McGill University, Québec, Canada
| | | | - Tanja Kuhlmann
- Institute of Neuropathology, University Hospital Münster, Münster, Germany
| |
Collapse
|
45
|
Palasantzas VEJM, Tamargo-Rubio I, Le K, Slager J, Wijmenga C, Jonkers IH, Kumar V, Fu J, Withoff S. iPSC-derived organ-on-a-chip models for personalized human genetics and pharmacogenomics studies. Trends Genet 2023; 39:268-284. [PMID: 36746737 DOI: 10.1016/j.tig.2023.01.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 11/24/2022] [Accepted: 01/12/2023] [Indexed: 02/07/2023]
Abstract
Genome-wide association studies (GWAS) have now correlated hundreds of genetic variants with complex genetic diseases and drug efficacy. Functional characterization of these factors remains challenging, particularly because of the lack of human model systems. Molecular and nanotechnological advances, in particular the ability to generate patient-specific PSC lines, differentiate them into diverse cell types, and seed and combine them on microfluidic chips, have led to the establishment of organ-on-a-chip (OoC) platforms that recapitulate organ biology. OoC technology thus provides unique personalized platforms for studying the effects of host genetics and environmental factors on organ physiology. In this review we describe the technology and provide examples of how OoCs may be used for disease modeling and pharmacogenetic research.
Collapse
Affiliation(s)
- Victoria E J M Palasantzas
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Isabel Tamargo-Rubio
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Kieu Le
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jelle Slager
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Cisca Wijmenga
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Iris H Jonkers
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Vinod Kumar
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Internal Medicine and Radboud Centre for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jingyuan Fu
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Sebo Withoff
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
46
|
Du H, Huo Z, Chen Y, Zhao Z, Meng F, Wang X, Liu S, Zhang H, Zhou F, Liu J, Zhang L, Zhou S, Guan Y, Wang X. Induced Pluripotent Stem Cells and Their Applications in Amyotrophic Lateral Sclerosis. Cells 2023; 12:cells12060971. [PMID: 36980310 PMCID: PMC10047679 DOI: 10.3390/cells12060971] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/20/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease that results in the loss of motor function in the central nervous system (CNS) and ultimately death. The mechanisms underlying ALS pathogenesis have not yet been fully elucidated, and ALS cannot be treated effectively. Most studies have applied animal or single-gene intervention cell lines as ALS disease models, but they cannot accurately reflect the pathological characteristics of ALS. Induced pluripotent stem cells (iPSCs) can be reprogrammed from somatic cells, possessing the ability to self-renew and differentiate into a variety of cells. iPSCs can be obtained from ALS patients with different genotypes and phenotypes, and the genetic background of the donor cells remains unchanged during reprogramming. iPSCs can differentiate into neurons and glial cells related to ALS. Therefore, iPSCs provide an excellent method to evaluate the impact of diseases on ALS patients. Moreover, patient-derived iPSCs are obtained from their own somatic cells, avoiding ethical concerns and posing only a low risk of immune rejection. The iPSC technology creates new hope for ALS treatment. Here, we review recent studies on iPSCs and their applications in disease modeling, drug screening and cell therapy in ALS, with a particular focus on the potential for ALS treatment.
Collapse
Affiliation(s)
- Hongmei Du
- Department of Histology and Embryology, School of Basic Medical Sciences, Weifang Medical University, Weifang 261053, China
- Neurologic Disorders and Regenerative Repair Laboratory, Weifang Medical University, Weifang 261053, China
| | - Zijun Huo
- Department of Histology and Embryology, School of Basic Medical Sciences, Weifang Medical University, Weifang 261053, China
| | - Yanchun Chen
- Department of Histology and Embryology, School of Basic Medical Sciences, Weifang Medical University, Weifang 261053, China
- Neurologic Disorders and Regenerative Repair Laboratory, Weifang Medical University, Weifang 261053, China
| | - Zhenhan Zhao
- Department of Histology and Embryology, School of Basic Medical Sciences, Weifang Medical University, Weifang 261053, China
| | - Fandi Meng
- Department of Histology and Embryology, School of Basic Medical Sciences, Weifang Medical University, Weifang 261053, China
| | - Xuemei Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Weifang Medical University, Weifang 261053, China
| | - Shiyue Liu
- Neurologic Disorders and Regenerative Repair Laboratory, Weifang Medical University, Weifang 261053, China
| | - Haoyun Zhang
- Neurologic Disorders and Regenerative Repair Laboratory, Weifang Medical University, Weifang 261053, China
| | - Fenghua Zhou
- Neurologic Disorders and Regenerative Repair Laboratory, Weifang Medical University, Weifang 261053, China
- Department of Pathology, School of Basic Medical Sciences, Weifang Medical University, Weifang 261053, China
| | - Jinmeng Liu
- Neurologic Disorders and Regenerative Repair Laboratory, Weifang Medical University, Weifang 261053, China
| | - Lingyun Zhang
- Neurologic Disorders and Regenerative Repair Laboratory, Weifang Medical University, Weifang 261053, China
| | - Shuanhu Zhou
- Harvard Medical School and Harvard Stem Cell Institute, Harvard University, Boston, MA 02115, USA
| | - Yingjun Guan
- Department of Histology and Embryology, School of Basic Medical Sciences, Weifang Medical University, Weifang 261053, China
- Neurologic Disorders and Regenerative Repair Laboratory, Weifang Medical University, Weifang 261053, China
| | - Xin Wang
- Harvard Medical School and Harvard Stem Cell Institute, Harvard University, Boston, MA 02115, USA
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
47
|
Mead RJ, Shan N, Reiser HJ, Marshall F, Shaw PJ. Amyotrophic lateral sclerosis: a neurodegenerative disorder poised for successful therapeutic translation. Nat Rev Drug Discov 2023; 22:185-212. [PMID: 36543887 PMCID: PMC9768794 DOI: 10.1038/s41573-022-00612-2] [Citation(s) in RCA: 183] [Impact Index Per Article: 91.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2022] [Indexed: 12/24/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating disease caused by degeneration of motor neurons. As with all major neurodegenerative disorders, development of disease-modifying therapies has proven challenging for multiple reasons. Nevertheless, ALS is one of the few neurodegenerative diseases for which disease-modifying therapies are approved. Significant discoveries and advances have been made in ALS preclinical models, genetics, pathology, biomarkers, imaging and clinical readouts over the last 10-15 years. At the same time, novel therapeutic paradigms are being applied in areas of high unmet medical need, including neurodegenerative disorders. These developments have evolved our knowledge base, allowing identification of targeted candidate therapies for ALS with diverse mechanisms of action. In this Review, we discuss how this advanced knowledge, aligned with new approaches, can enable effective translation of therapeutic agents from preclinical studies through to clinical benefit for patients with ALS. We anticipate that this approach in ALS will also positively impact the field of drug discovery for neurodegenerative disorders more broadly.
Collapse
Affiliation(s)
- Richard J Mead
- Sheffield Institute for Translational Neuroscience, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
- Keapstone Therapeutics, The Innovation Centre, Broomhall, Sheffield, UK
| | - Ning Shan
- Aclipse Therapeutics, Radnor, PA, US
| | | | - Fiona Marshall
- MSD UK Discovery Centre, Merck, Sharp and Dohme (UK) Limited, London, UK
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, UK.
- Neuroscience Institute, University of Sheffield, Sheffield, UK.
- Keapstone Therapeutics, The Innovation Centre, Broomhall, Sheffield, UK.
| |
Collapse
|
48
|
Suzuki N, Nishiyama A, Warita H, Aoki M. Genetics of amyotrophic lateral sclerosis: seeking therapeutic targets in the era of gene therapy. J Hum Genet 2023; 68:131-152. [PMID: 35691950 PMCID: PMC9968660 DOI: 10.1038/s10038-022-01055-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/17/2022] [Accepted: 05/29/2022] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is an intractable disease that causes respiratory failure leading to mortality. The main locus of ALS is motor neurons. The success of antisense oligonucleotide (ASO) therapy in spinal muscular atrophy (SMA), a motor neuron disease, has triggered a paradigm shift in developing ALS therapies. The causative genes of ALS and disease-modifying genes, including those of sporadic ALS, have been identified one after another. Thus, the freedom of target choice for gene therapy has expanded by ASO strategy, leading to new avenues for therapeutic development. Tofersen for superoxide dismutase 1 (SOD1) was a pioneer in developing ASO for ALS. Improving protocols and devising early interventions for the disease are vital. In this review, we updated the knowledge of causative genes in ALS. We summarized the genetic mutations identified in familial ALS and their clinical features, focusing on SOD1, fused in sarcoma (FUS), and transacting response DNA-binding protein. The frequency of the C9ORF72 mutation is low in Japan, unlike in Europe and the United States, while SOD1 and FUS are more common, indicating that the target mutations for gene therapy vary by ethnicity. A genome-wide association study has revealed disease-modifying genes, which could be the novel target of gene therapy. The current status and prospects of gene therapy development were discussed, including ethical issues. Furthermore, we discussed the potential of axonal pathology as new therapeutic targets of ALS from the perspective of early intervention, including intra-axonal transcription factors, neuromuscular junction disconnection, dysregulated local translation, abnormal protein degradation, mitochondrial pathology, impaired axonal transport, aberrant cytoskeleton, and axon branching. We simultaneously discuss important pathological states of cell bodies: persistent stress granules, disrupted nucleocytoplasmic transport, and cryptic splicing. The development of gene therapy based on the elucidation of disease-modifying genes and early intervention in molecular pathology is expected to become an important therapeutic strategy in ALS.
Collapse
Affiliation(s)
- Naoki Suzuki
- Department of Neurology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Japan.
| | - Ayumi Nishiyama
- Department of Neurology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Japan
| | - Hitoshi Warita
- Department of Neurology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Japan
| | - Masashi Aoki
- Department of Neurology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Japan.
| |
Collapse
|
49
|
Limone F, Guerra San Juan I, Mitchell JM, Smith JLM, Raghunathan K, Meyer D, Ghosh SD, Couto A, Klim JR, Joseph BJ, Gold J, Mello CJ, Nemesh J, Smith BM, Verhage M, McCarroll SA, Pietiläinen O, Nehme R, Eggan K. Efficient generation of lower induced motor neurons by coupling Ngn2 expression with developmental cues. Cell Rep 2023; 42:111896. [PMID: 36596304 PMCID: PMC10117176 DOI: 10.1016/j.celrep.2022.111896] [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: 12/17/2021] [Revised: 06/01/2022] [Accepted: 12/08/2022] [Indexed: 01/03/2023] Open
Abstract
Human pluripotent stem cells (hPSCs) are a powerful tool for disease modeling of hard-to-access tissues (such as the brain). Current protocols either direct neuronal differentiation with small molecules or use transcription-factor-mediated programming. In this study, we couple overexpression of transcription factor Neurogenin2 (Ngn2) with small molecule patterning to differentiate hPSCs into lower induced motor neurons (liMoNes/liMNs). This approach induces canonical MN markers including MN-specific Hb9/MNX1 in more than 95% of cells. liMNs resemble bona fide hPSC-derived MN, exhibit spontaneous electrical activity, express synaptic markers, and can contact muscle cells in vitro. Pooled, multiplexed single-cell RNA sequencing on 50 hPSC lines reveals reproducible populations of distinct subtypes of cervical and brachial MNs that resemble their in vivo, embryonic counterparts. Combining small molecule patterning with Ngn2 overexpression facilitates high-yield, reproducible production of disease-relevant MN subtypes, which is fundamental in propelling our knowledge of MN biology and its disruption in disease.
Collapse
Affiliation(s)
- Francesco Limone
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Stanley Centre for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Leiden University Medical Center, LUMC, 2333 ZA Leiden, the Netherlands.
| | - Irune Guerra San Juan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Stanley Centre for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, the Netherlands; Human Genetics, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Jana M Mitchell
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Stanley Centre for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Janell L M Smith
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Kavya Raghunathan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Stanley Centre for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Daniel Meyer
- Stanley Centre for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Sulagna Dia Ghosh
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Stanley Centre for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Alexander Couto
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Joseph R Klim
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Stanley Centre for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Brian J Joseph
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Departments of Pathology and Cell Biology, Columbia University Irving Medical Centre, New York, NY 10032, USA
| | - John Gold
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Curtis J Mello
- Stanley Centre for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - James Nemesh
- Stanley Centre for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Brittany M Smith
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Matthijs Verhage
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, the Netherlands; Human Genetics, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Steven A McCarroll
- Stanley Centre for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Olli Pietiläinen
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Stanley Centre for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Ralda Nehme
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Stanley Centre for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kevin Eggan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
50
|
Pain O, Jones A, Al Khleifat A, Agarwal D, Hramyka D, Karoui H, Kubica J, Llewellyn DJ, Ranson JM, Yao Z, Iacoangeli A, Al-Chalabi A. Harnessing Transcriptomic Signals for Amyotrophic Lateral Sclerosis to Identify Novel Drugs and Enhance Risk Prediction. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.01.18.23284589. [PMID: 36747854 PMCID: PMC9901068 DOI: 10.1101/2023.01.18.23284589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Introduction Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease. This study integrates the latest ALS genome-wide association study (GWAS) summary statistics with functional genomic annotations with the aim of providing mechanistic insights into ALS risk loci, inferring drug repurposing opportunities, and enhancing prediction of ALS risk and clinical characteristics. Methods Genes associated with ALS were identified using GWAS summary statistic methodology including SuSiE SNP-based fine-mapping, and transcriptome- and proteome-wide association study (TWAS/PWAS) analyses. Using several approaches, gene associations were integrated with the DrugTargetor drug-gene interaction database to identify drugs that could be repurposed for the treatment of ALS. Furthermore, ALS gene associations from TWAS were combined with observed blood expression in two external ALS case-control datasets to calculate polytranscriptomic scores and evaluate their utility for prediction of ALS risk and clinical characteristics, including site of onset, age at onset, and survival. Results SNP-based fine-mapping, TWAS and PWAS identified 117 genes associated with ALS, with TWAS and PWAS providing novel mechanistic insights. Drug repurposing analyses identified five drugs significantly enriched for interactions with ALS associated genes, with directional analyses highlighting α-glucosidase inhibitors may exacerbate ALS pathology. Additionally, drug class enrichment analysis showed calcium channel blockers may reduce ALS risk. Across the two observed expression target samples, ALS polytranscriptomic scores significantly predicted ALS risk (R2 = 4%; p-value = 2.1×10-21). Conclusions Functionally-informed analyses of ALS GWAS summary statistics identified novel mechanistic insights into ALS aetiology, highlighted several therapeutic research avenues, and enabled statistically significant prediction of ALS risk.
Collapse
Affiliation(s)
- Oliver Pain
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Ashley Jones
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Ahmad Al Khleifat
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Devika Agarwal
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, Old Road Campus, University of Oxford, Oxford, United Kingdom
| | - Dzmitry Hramyka
- Core Unit Bioinformatics (CUBI), Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Hajer Karoui
- Multiple Sclerosis and Parkinson’s Tissue Bank, Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Jędrzej Kubica
- Laboratory of Structural Bioinformatics, Institute of Evolutionary Biology, University of Warsaw, Poland
- Laboratory of Theory of Biopolimers, Faculty of Chemistry, University of Warsaw, Poland
| | - David J. Llewellyn
- University of Exeter Medical School, Exeter, United Kingdom
- Alan Turing Institute, London, United Kingdom
| | | | - Zhi Yao
- LifeArc, Stevenage, United Kingdom
| | - Alfredo Iacoangeli
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
- National Institute for Health Research Biomedical Research Centre and Dementia Unit at South London and Maudsley NHS Foundation Trust and King’s College London, London, United Kingdom
| | - Ammar Al-Chalabi
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| |
Collapse
|