1
|
Russo A, Putaggio S, Tellone E, Calderaro A, Cirmi S, Laganà G, Ficarra S, Barreca D, Patanè GT. Emerging Ferroptosis Involvement in Amyotrophic Lateral Sclerosis Pathogenesis: Neuroprotective Activity of Polyphenols. Molecules 2025; 30:1211. [PMID: 40141987 PMCID: PMC11944684 DOI: 10.3390/molecules30061211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
Neurodegenerative diseases are a group of diseases that share common features, such as the generation of misfolded protein deposits and increased oxidative stress. Among them, amyotrophic lateral sclerosis (ALS), whose pathogenesis is still not entirely clear, is a complex neurodegenerative disease linked both to gene mutations affecting different proteins, such as superoxide dismutase 1, Tar DNA binding protein 43, Chromosome 9 open frame 72, and Fused in Sarcoma, and to altered iron homeostasis, mitochondrial dysfunction, oxidative stress, and impaired glutamate metabolism. The purpose of this review is to highlight the molecular targets common to ALS and ferroptosis. Indeed, many pathways implicated in the disease are hallmarks of ferroptosis, a recently discovered type of iron-dependent programmed cell death characterized by increased reactive oxygen species (ROS) and lipid peroxidation. Iron accumulation results in mitochondrial dysfunction and increased levels of ROS, lipid peroxidation, and ferroptosis triggers; in addition, the inhibition of the Xc- system results in reduced cystine levels and glutamate accumulation, leading to excitotoxicity and the inhibition of GPx4 synthesis. These results highlight the potential involvement of ferroptosis in ALS, providing new molecular and biochemical targets that could be exploited in the treatment of the disease using polyphenols.
Collapse
Affiliation(s)
| | - Stefano Putaggio
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (A.R.); (A.C.); (S.C.); (G.L.); (S.F.); (D.B.); (G.T.P.)
| | - Ester Tellone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (A.R.); (A.C.); (S.C.); (G.L.); (S.F.); (D.B.); (G.T.P.)
| | | | | | | | | | | | | |
Collapse
|
2
|
Maier A, Kettemann D, Weyen U, Grehl T, Schulte PC, Steinbach R, Rödiger A, Weydt P, Petri S, Wolf J, Grosskreutz J, Koch JC, Weishaupt JH, Rosseau S, Norden J, Körtvélyessy P, Koch B, Holm T, Hildebrandt B, Schumann P, Walter B, Riitano A, Münch C, Meyer T, Spittel S. Provision, cough efficacy and treatment satisfaction of mechanical insufflation-exsufflation in a large multicenter cohort of patients with amyotrophic lateral sclerosis. Sci Rep 2025; 15:7360. [PMID: 40025240 PMCID: PMC11873142 DOI: 10.1038/s41598-025-91692-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 02/21/2025] [Indexed: 03/04/2025] Open
Abstract
In patients with amyotrophic lateral sclerosis (ALS), mechanical insufflation-exsufflation (MI-E) addresses cough deficiency to achieve major therapeutic goals: improving costal muscle and joint function, reducing atelectasis through insufflation, and clearing bronchial secretions via exsufflation. Despite its perceived benefits, there is limited systematic research on MI-E provision, symptom alleviation, or patient satisfaction. The research platform Ambulanzpartner coordinated this longitudinal observational study conducted in 12 German ALS centers from July 2018 to September 2023. Patients were enrolled based on ALS-related cough deficiency requiring MI-E therapy. The study recorded provision, reasons for withholding MI-E, clinical parameters, therapy frequency, subjective cough deficiency, and symptomatic relief. Satisfaction with MI-E therapy was determined by the likelihood of recommendation. Out of 694 ALS patients indicated for MI-E, 527 (75.9%) received the therapy. The primary reason for non-provision was that the patient had died before provision (n = 66 of 167; 39.5%). These patients were significantly more affected as represented by higher progression rates and lower cough peak flows (CPF) at the time of MI-E indication (p < 0.05). Most patients who received MI-E used it daily (n = 290 of 370; 78.4%). Self-assessed cough deficiency correlated with clinical measurements, especially for patients with higher deficits. At follow-up visits, patients reported reduced cough deficiency (p < 0.001). Frequent MI-E use was linked to greater symptom relief and higher likelihood of recommending the therapy. This study highlights the symptomatic and palliative potential of MI-E therapy for ALS patients.
Collapse
Affiliation(s)
- André Maier
- Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Outpatient Center for ALS and Other Motor Neuron Diseases, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
| | - Dagmar Kettemann
- Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Outpatient Center for ALS and Other Motor Neuron Diseases, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Ute Weyen
- Center for ALS and Other Motor Neuron Disorders, Berufsgenossenschaftliches Universitätsklinikum Bergmannsheil, Bochum, Germany
| | - Torsten Grehl
- Department of Neurology, Center for ALS and Other Motor Neuron Disorders, Alfried Krupp Krankenhaus, Essen, Germany
| | - Peter Caspar Schulte
- Clinic for Pneumology, Gastroenterology and Internal Medicine, Alfried Krupp Krankenhaus, Essen, Germany
| | - Robert Steinbach
- Department of Neurology, Jena University Hospital, Jena, Germany
| | | | - Patrick Weydt
- Department for Neurodegenerative Disorders and Gerontopsychiatry, Bonn University, Bonn, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen, Research Site Bonn, Bonn, Germany
| | - Susanne Petri
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Joachim Wolf
- Department of Neurology, Diako Mannheim, Mannheim, Germany
| | | | - Jan Christoph Koch
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Jochen H Weishaupt
- Division for Neurodegenerative Diseases, Neurology Department, Mannheim Center for Translational Medicine, University Medicine Mannheim, Heidelberg University, Mannheim, Germany
| | - Simone Rosseau
- Pneumological ventilation center, Ernst von Bergmann Klinik Bad Belzig, Bad Belzig, Germany
| | - Jenny Norden
- Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Outpatient Center for ALS and Other Motor Neuron Diseases, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Peter Körtvélyessy
- Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Outpatient Center for ALS and Other Motor Neuron Diseases, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Birgit Koch
- Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Outpatient Center for ALS and Other Motor Neuron Diseases, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Teresa Holm
- Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Outpatient Center for ALS and Other Motor Neuron Diseases, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | | | - Peggy Schumann
- Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Outpatient Center for ALS and Other Motor Neuron Diseases, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Bertram Walter
- Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Outpatient Center for ALS and Other Motor Neuron Diseases, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | | | - Christoph Münch
- Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Outpatient Center for ALS and Other Motor Neuron Diseases, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- Ambulanzpartner Soziotechnologie APST GmbH, Berlin, Germany
| | - Thomas Meyer
- Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Outpatient Center for ALS and Other Motor Neuron Diseases, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- Ambulanzpartner Soziotechnologie APST GmbH, Berlin, Germany
| | - Susanne Spittel
- Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Outpatient Center for ALS and Other Motor Neuron Diseases, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- Ambulanzpartner Soziotechnologie APST GmbH, Berlin, Germany
| |
Collapse
|
3
|
Jin Y, Conneely KN, Ma W, Naviaux RK, Siddique T, Allen EG, Guingrich S, Pascuzzi RM, Jin P. Whole-genome bisulfite sequencing of cell-free DNA unveils age-dependent and ALS-associated methylation alterations. Cell Biosci 2025; 15:26. [PMID: 39980027 PMCID: PMC11843967 DOI: 10.1186/s13578-025-01366-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 02/11/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Cell-free DNA (cfDNA) in plasma carries epigenetic signatures specific to tissue or cell of origin. Aberrant methylation patterns in circulating cfDNA have emerged as valuable tools for noninvasive cancer detection, prenatal diagnostics, and organ transplant assessment. Such epigenetic changes also hold significant promise for the diagnosis of neurodegenerative diseases, which often progresses slowly and has a lengthy asymptomatic period. However, genome-wide cfDNA methylation changes in neurodegenerative diseases remain poorly understood. RESULTS We used whole-genome bisulfite sequencing (WGBS) to profile age-dependent and ALS-associated methylation signatures in cfDNA from 30 individuals, including young and middle-aged controls, as well as ALS patients with matched controls. We identified 5,223 age-related differentially methylated loci (DMLs) (FDR < 0.05), with 51.6% showing hypomethylation in older individuals. Our results significantly overlapped with age-associated CpGs identified in a large blood-based epigenome-wide association study (EWAS). Comparing ALS patients to controls, we detected 1,045 differentially methylated regions (DMRs) in gene bodies, promoters, and intergenic regions. Notably, these DMRs were linked to key ALS-associated pathways, including endocytosis and cell adhesion. Integration with spinal cord transcriptomics revealed that 31% of DMR-associated genes exhibited differential expression in ALS patients compared to controls, with over 20 genes significantly correlating with disease duration. Furthermore, comparison with published single-nucleus RNA sequencing (snRNA-Seq) data of ALS demonstrated that cfDNA methylation changes reflects cell-type-specific gene dysregulation in the brain of ALS patients, particularly in excitatory neurons and astrocytes. Deconvolution of cfDNA methylation profiles suggested altered proportions of immune and liver-derived cfDNA in ALS patients. CONCLUSIONS cfDNA methylation is a powerful tool for assessing age-related changes and ALS-specific molecular dysregulation by revealing perturbed locus, genes, and the proportional contributions of different tissues/cells to the plasma. This technique holds promise for clinical application in biomarker discovery across a broad spectrum of neurodegenerative disorders.
Collapse
Affiliation(s)
- Yulin Jin
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Karen N Conneely
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Wenjing Ma
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Robert K Naviaux
- Departments of Medicine, Pediatrics, and Pathology, and the Mitochondrial and Metabolic Disease Center (MMDC), School of Medicine, University of California San Diego, San Diego, CA, 92103, USA
| | - Teepu Siddique
- Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Emily G Allen
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Sandra Guingrich
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Robert M Pascuzzi
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Peng Jin
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
4
|
Cheng J, Wu BT, Liu HP, Lin WY. Machine learning identified novel players in lipid metabolism, endosomal trafficking, and iron metabolism of the ALS spinal cord. Sci Rep 2025; 15:1564. [PMID: 39794401 PMCID: PMC11723943 DOI: 10.1038/s41598-024-81315-z] [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: 02/20/2024] [Accepted: 11/26/2024] [Indexed: 01/13/2025] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease affecting motor neurons. Although genes causing familial cases have been identified, those of sporadic ALS, which occupies the majority of patients, are still elusive. In this study, we adopted machine learning to build binary classifiers based on the New York Genome Center (NYGC) ALS Consortium's RNA-seq data of the postmortem spinal cord of ALS and non-neurological disease control. The accuracy of the classifiers was greater than 83% and 77% for the training set and the unseen test set, respectively. The classifiers contained 114 genes. Among them, 41 genes have been reported in previous ALS studies, and others are novel in this field. These genes are involved in mitochondrial respiration, lipid metabolism, endosomal trafficking, and iron metabolism, which may promote the progression of ALS pathology.
Collapse
Affiliation(s)
- Jack Cheng
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan
- Department of Medical Research, China Medical University Hospital, Taichung, 40447, Taiwan
| | - Bor-Tsang Wu
- Department of Senior Citizen Service Management, National Taichung University of Science and Technology, Taichung, 40343, Taiwan
| | - Hsin-Ping Liu
- Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan.
| | - Wei-Yong Lin
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan.
- Department of Medical Research, China Medical University Hospital, Taichung, 40447, Taiwan.
| |
Collapse
|
5
|
Canosa A, Martino A, Manera U, Giuliani A, Vasta R, Palumbo F, Grassano M, Morbelli SD, Pardini M, Chiaravalloti A, Schillaci O, Leenders KL, Kogan RV, Polverari G, Zocco G, Pede FD, Mattei FD, Cabras S, Matteoni E, Moglia C, Calvo A, Chiò A, Pagani M. Sex-related differences in amyotrophic lateral sclerosis: A 2-[ 18F]FDG-PET study. Eur J Neurol 2025; 32:e16588. [PMID: 39655539 PMCID: PMC11629101 DOI: 10.1111/ene.16588] [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: 07/29/2024] [Accepted: 11/28/2024] [Indexed: 12/12/2024]
Abstract
PURPOSE We investigated sex-related brain metabolic differences in Amyotrophic Lateral Sclerosis (ALS) and healthy controls (HC). METHODS We collected two equal-sized groups of male (m-ALS) and female ALS (f-ALS) patients (n = 130 each), who underwent 2-[18F]FDG-PET at diagnosis, matched for site of onset, cognitive status and King's stage. We included 168 age-matched healthy controls, half female (f-HC) and half male (m-HC). We compared brain metabolism of males and females separately for ALS and HC, including age as covariate. A differential network analysis was performed to evaluate brain connectivity. RESULTS M-ALS showed relative hypometabolism of bilateral medial frontal, parietal and occipital cortices, and left temporal cortex, compared to f-ALS. In node-wise comparison, f-ALS showed significantly higher connectivity in right middle cingulate cortex and left superior and medial frontal gyrus. In HC we did not find any sex-related differences. CONCLUSION Sex resulted a major determinant of brain metabolism and connectivity in ALS patients.
Collapse
Affiliation(s)
- Antonio Canosa
- ALS Centre, ‘Rita Levi Montalcini’ Department of NeuroscienceUniversity of TurinTurinItaly
- Azienda Ospedaliero‐Universitaria Città della Salute e della Scienza di TorinoNeurology Unit 1UTurinItaly
- Institute of Cognitive Sciences and Technologies, C.N.RRomeItaly
| | - Alessio Martino
- Department of Business and ManagementLUISS UniversityRomeItaly
| | - Umberto Manera
- ALS Centre, ‘Rita Levi Montalcini’ Department of NeuroscienceUniversity of TurinTurinItaly
- Azienda Ospedaliero‐Universitaria Città della Salute e della Scienza di TorinoNeurology Unit 1UTurinItaly
| | | | - Rosario Vasta
- ALS Centre, ‘Rita Levi Montalcini’ Department of NeuroscienceUniversity of TurinTurinItaly
| | - Francesca Palumbo
- ALS Centre, ‘Rita Levi Montalcini’ Department of NeuroscienceUniversity of TurinTurinItaly
| | - Maurizio Grassano
- ALS Centre, ‘Rita Levi Montalcini’ Department of NeuroscienceUniversity of TurinTurinItaly
| | - Silvia Daniela Morbelli
- Department of Medical SciencesUniversity of TurinTurinItaly
- Azienda Ospedaliero‐Universitaria Città della Salute e della Scienza di TorinoNuclear Medicine UnitTurinItaly
| | - Matteo Pardini
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI)University of GenoaGenoaItaly
- IRCCS Ospedale Policlinico San MartinoGenoaItaly
| | - Agostino Chiaravalloti
- Department of Biomedicine and PreventionUniversity of Rome ‘Tor Vergata’RomeItaly
- IRCCS NeuromedPozzilliItaly
| | - Orazio Schillaci
- Department of Biomedicine and PreventionUniversity of Rome ‘Tor Vergata’RomeItaly
| | - Klaus Leonard Leenders
- Department of NeurologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
- Department of Nuclear Medicine and Molecular ImagingUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Rosalie Vered Kogan
- Department of Nuclear Medicine and Molecular ImagingUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Giulia Polverari
- Positron Emission Tomography Centre AFFIDEA‐IRMET S.p.ATurinItaly
| | - Grazia Zocco
- ALS Centre, ‘Rita Levi Montalcini’ Department of NeuroscienceUniversity of TurinTurinItaly
| | - Francesca Di Pede
- ALS Centre, ‘Rita Levi Montalcini’ Department of NeuroscienceUniversity of TurinTurinItaly
| | - Filippo De Mattei
- ALS Centre, ‘Rita Levi Montalcini’ Department of NeuroscienceUniversity of TurinTurinItaly
| | - Sara Cabras
- ALS Centre, ‘Rita Levi Montalcini’ Department of NeuroscienceUniversity of TurinTurinItaly
- Center for NeuroscienceUniversity of CamerinoCamerinoItaly
| | - Enrico Matteoni
- ALS Centre, ‘Rita Levi Montalcini’ Department of NeuroscienceUniversity of TurinTurinItaly
| | - Cristina Moglia
- ALS Centre, ‘Rita Levi Montalcini’ Department of NeuroscienceUniversity of TurinTurinItaly
- Azienda Ospedaliero‐Universitaria Città della Salute e della Scienza di TorinoNeurology Unit 1UTurinItaly
| | - Andrea Calvo
- ALS Centre, ‘Rita Levi Montalcini’ Department of NeuroscienceUniversity of TurinTurinItaly
- Azienda Ospedaliero‐Universitaria Città della Salute e della Scienza di TorinoNeurology Unit 1UTurinItaly
- Neuroscience Institute of Turin (NIT)TurinItaly
| | - Adriano Chiò
- ALS Centre, ‘Rita Levi Montalcini’ Department of NeuroscienceUniversity of TurinTurinItaly
- Azienda Ospedaliero‐Universitaria Città della Salute e della Scienza di TorinoNeurology Unit 1UTurinItaly
- Institute of Cognitive Sciences and Technologies, C.N.RRomeItaly
- Neuroscience Institute of Turin (NIT)TurinItaly
| | - Marco Pagani
- Institute of Cognitive Sciences and Technologies, C.N.RRomeItaly
- Department of Medical Radiation Physics and Nuclear MedicineKarolinska University HospitalStockholmSweden
| |
Collapse
|
6
|
Simoes FA, Christoforidou E, Cassel R, Dupuis L, Hafezparast M. Severe dynein dysfunction in cholinergic neurons exacerbates ALS-like phenotypes in a new mouse model. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167540. [PMID: 39428001 DOI: 10.1016/j.bbadis.2024.167540] [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] [Revised: 09/12/2024] [Accepted: 10/06/2024] [Indexed: 10/22/2024]
Abstract
Cytoplasmic dynein 1, a motor protein essential for retrograde axonal transport, is increasingly implicated in the pathogenesis of neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS). In this study, we developed a novel mouse model that combines the Legs at odd angles (Loa, F580Y) point mutation in the dynein heavy chain with a cholinergic neuron-specific knockout of the dynein heavy chain. This model, for the first time, allows us to investigate the impact of Loa allele exclusivity in these neurons into adulthood. Our findings reveal that this selective increase in dynein dysfunction exacerbated the phenotypes observed in heterozygous Loa mice including pre-wean survival, reduced body weight and grip strength. Additionally, it induced ALS-like pathology in neuromuscular junctions (NMJs) not seen in heterozygous Loa mice. Notably, we also found a previously unobserved significant increase in neurons displaying TDP-43 puncta in both Loa mutants, suggesting early TDP-43 mislocalisation - a hallmark of ALS. The novel model also exhibited a concurrent rise in p62 puncta that did not co-localise with TDP-43, indicating broader impairments in autophagic clearance mechanisms. Overall, this new model underscores the fact that dynein impairment alone can induce ALS-like pathology and provides a valuable platform to further explore the role of dynein in ALS.
Collapse
Affiliation(s)
- Fabio A Simoes
- Department of Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom
| | - Eleni Christoforidou
- Department of Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom
| | | | - Luc Dupuis
- University of Strasbourg, INSERM, UMR-S1329, Strasbourg, France
| | - Majid Hafezparast
- Department of Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom.
| |
Collapse
|
7
|
Wu H, Erenay FS, Özaltın OY, Dalgıç ÖO, Sır MY, He QM, Crum BA, Pasupathy KS. Prognostic factors affecting ALS progression through disease tollgates. J Neurol 2024; 272:69. [PMID: 39680215 DOI: 10.1007/s00415-024-12819-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 09/11/2024] [Accepted: 09/28/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND AND OBJECTIVES Understanding factors affecting the timing of critical clinical events in ALS progression. METHODS We captured ALS progression based on the timing of critical events (tollgates), by augmenting 6366 patients' data from the PRO-ACT database with tollgate-passed information using classification. Time trajectories of passing ALS tollgates after the first visit were derived using Kaplan-Meier analyses. The significant prognostic factors were found using log-rank tests. Decision-tree-based classifications identified significant ALS phenotypes characterized by the list of body segments involved at the first visit. RESULTS Standard (e.g., gender and onset type) and tollgate-related (phenotype and initial tollgate level) prognostic factors affect the timing of ALS tollgates. For instance, by the third year after the first visit, 80-100% of bulbar-onset patients vs. 43-48% of limb-onset patients, and 65-73% of females vs. 42-49% of males lost the ability to talk and started using a feeding tube. Compared to the standard factors, tollgate-related factors had a stronger effect on ALS progression. The initial impairment level significantly impacted subsequent ALS progression in a segment while affected segment combinations further characterized progression speed. For instance, patients with normal speech (Tollgate Level 0) at the first visit had less than a 10% likelihood of losing speech within a year, while for patients with Tollgate Level 1 (affected speech), this likelihood varied between 23 and 53% based on additional segment (leg) involvement. CONCLUSIONS Tollgate- and phenotype-related factors have a strong effect on the timing of ALS tollgates. All factors should be jointly considered to better characterize patient groups with different progression aggressiveness.
Collapse
Affiliation(s)
- Haoran Wu
- School of Business, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - F Safa Erenay
- Department of Management Sciences and Engineering, University of Waterloo, Waterloo, ON, Canada
| | - Osman Y Özaltın
- E.P. Fitts Department of Industrial & Systems Engineering, NC State University, Raleigh, NC, USA
| | | | - Mustafa Y Sır
- Mayo Clinic Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Rochester, USA
| | - Qi-Ming He
- Department of Management Sciences and Engineering, University of Waterloo, Waterloo, ON, Canada
| | - Brian A Crum
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Kalyan S Pasupathy
- Mayo Clinic Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Rochester, USA
- Biomedical & Health Information Sciences, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
8
|
Klose V, Jesse S, Lewerenz J, Kassubek J, Dorst J, Rosenbohm A, Nagel G, Wernecke D, Roselli F, Tumani H, Ludolph AC. Blood-CSF barrier integrity in amyotrophic lateral sclerosis. Brain 2024; 147:4254-4264. [PMID: 38743595 DOI: 10.1093/brain/awae144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 05/16/2024] Open
Abstract
The integrity of the blood-CSF barrier plays a major role in inflammation, but also in shielding the CNS from external and systemic-potentially toxic-factors. Here we report results of measurements of the albumin quotient-which is thought to mirror the integrity of the blood-CSF barrier-in 1059 patients with amyotrophic lateral sclerosis. The results were compared with groups of patients suffering from Alzheimer's disease, facial palsy and tension headache. The albumin quotient, an accepted measure of the blood-CSF barrier integrity, was not significantly different from control populations. In addition, we found that the albumin quotient correlated with survival of the patients; this effect was mainly driven by male patients and influenced by age, body mass index and diabetes mellitus. We conclude that the blood-CSF barrier is intact in this large cohort of patients with amyotrophic lateral sclerosis and that the albumin quotient correlates with survival. Whether this is important for the pathogenesis of the disease, requires mechanistic studies.
Collapse
Affiliation(s)
- Veronika Klose
- Department of Neurology, Ulm University, 89081 Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE) Ulm, 89081 Ulm, Germany
| | - Sarah Jesse
- Department of Neurology, Ulm University, 89081 Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE) Ulm, 89081 Ulm, Germany
| | - Jan Lewerenz
- Department of Neurology, Ulm University, 89081 Ulm, Germany
| | - Jan Kassubek
- Department of Neurology, Ulm University, 89081 Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE) Ulm, 89081 Ulm, Germany
| | - Johannes Dorst
- Department of Neurology, Ulm University, 89081 Ulm, Germany
| | | | - Gabriele Nagel
- Institute of Epidemiology and Medical Biometry, Ulm University, 89081 Ulm, Germany
| | - Deborah Wernecke
- Institute of Epidemiology and Medical Biometry, Ulm University, 89081 Ulm, Germany
| | - Francesco Roselli
- Department of Neurology, Ulm University, 89081 Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE) Ulm, 89081 Ulm, Germany
| | | | - Albert C Ludolph
- Department of Neurology, Ulm University, 89081 Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE) Ulm, 89081 Ulm, Germany
| |
Collapse
|
9
|
Cox OH, Seifuddin F, Guo J, Pirooznia M, Boersma GJ, Wang J, Tamashiro KL, Lee RS. Implementation of the Methyl-Seq platform to identify tissue- and sex-specific DNA methylation differences in the rat epigenome. Epigenetics 2024; 19:2393945. [PMID: 39306700 PMCID: PMC11418217 DOI: 10.1080/15592294.2024.2393945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 07/23/2024] [Accepted: 08/13/2024] [Indexed: 09/25/2024] Open
Abstract
Epigenomic annotations for the rat lag far behind those of human and mouse, despite the rat's immense utility in pharmacological and behavioral studies and the need to understand their epigenetic mechanisms. We have designed a targeted-enrichment method followed by next-generation sequencing (Methyl-Seq) to identify DNA methylation (DNAm) signatures across the rat genome. The design reflected an attempt to create a more comprehensive investigation of the rat epigenome, as it included promoters, CpG islands, and island shores of all RefSeq genes. In this study, we implemented the rat Methyl-Seq platform and tested its ability to distinguish differentially methylated regions (DMRs) among three different tissue types, three distinct brain regions, and, in the hippocampus, between males and females. These comparisons yielded DNAm differences of differing magnitudes, many of which were independently validated by bisulfite pyrosequencing, including autosomal regions that were predicted to show the least degree of difference in DNAm between males and females. Quantitative reverse transcription PCR revealed that most genes associated with the DMRs showed tissue-, brain region-, and sex-specific differences in expression. In particular, we found evidence for sex-specific DNAm and expression differences at Tubb6, Lrrn2, Tex26, and Sox5l1, all of which play important roles in neurodevelopment and have been implicated in studies examining sex differences. Our results demonstrate the utility of the rat Methyl-Seq platform and suggest the presence of DNAm differences between the male and female hippocampus. The rat Methyl-Seq has the potential to provide epigenomic insights into pharmacological and behavioral studies performed in the rat.
Collapse
Affiliation(s)
- Olivia H. Cox
- Mood Disorders Center, Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Fayaz Seifuddin
- Mood Disorders Center, Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Jeffrey Guo
- Mood Disorders Center, Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Mehdi Pirooznia
- Mood Disorders Center, Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Gretha J. Boersma
- GGZ Drenthe Mental Health Institute, Department of Forensic Psychiatry, Assen, The Netherlands
| | - Josh Wang
- Agilent Technologies, Inc., Santa Clara, USA
| | - Kellie L.K. Tamashiro
- Mood Disorders Center, Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Richard S. Lee
- Mood Disorders Center, Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, USA
| |
Collapse
|
10
|
Grant OA, Iacoangeli A, Zwamborn RAJ, van Rheenen W, Byrne R, Van Eijk KR, Kenna K, van Vugt JJFA, Cooper-Knock J, Kenna B, Vural A, Topp S, Campos Y, Weber M, Smith B, Dobson R, van Es MA, Vourc'h P, Corcia P, de Carvalho M, Gotkine M, Panades MP, Mora JS, Mill J, Garton F, McRae A, Wray NR, Shaw PJ, Landers JE, Glass JD, Shaw CE, Basak N, Hardiman O, Van Damme P, McLaughlin RL, van den Berg LH, Veldink JH, Al-Chalabi A, Al Khleifat A. Sex-specific DNA methylation differences in Amyotrophic lateral sclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.22.624866. [PMID: 39651197 PMCID: PMC11623544 DOI: 10.1101/2024.11.22.624866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Sex is an important covariate in all genetic and epigenetic research due to its role in the incidence, progression and outcome of many phenotypic characteristics and human diseases. Amyotrophic lateral sclerosis (ALS) is a motor neuron disease with a sex bias towards higher incidence in males. Here, we report for the first time a blood-based epigenome-wide association study meta-analysis in 9274 individuals after stringent quality control (5529 males and 3975 females). We identified a total of 226 ALS saDMPs (sex-associated DMPs) annotated to a total of 159 unique genes. These ALS saDMPs were depleted at transposable elements yet significantly enriched at enhancers and slightly enriched at 3'UTRs. These ALS saDMPs were enriched for transcription factor motifs such as ESR1 and REST. Moreover, we identified an additional 10 genes associated with ALS saDMPs through chromatin loop interactions, suggesting a potential regulatory role for these saDMPs on distant genes. Furthermore, we investigated the relationship between DNA methylation at specific CpG sites and overall survival in ALS using Cox proportional hazards models. We identified two ALS saDMPs, cg14380013 and cg06729676, that showed significant associations with survival. Overall, our study reports a reliable catalogue of sex-associated ALS saDMPs in ALS and elucidates several characteristics of these sites using a large-scale dataset. This resource will benefit future studies aiming to investigate the role of sex in the incidence, progression and risk for ALS.
Collapse
|
11
|
Białobrodzka E, Flis DJ, Akdogan B, Borkowska A, Wieckowski MR, Antosiewicz J, Zischka H, Dzik KP, Kaczor JJ, Ziolkowski W. Amyotrophic Lateral Sclerosis and swim training affect copper metabolism in skeletal muscle in a mouse model of disease. Muscle Nerve 2024; 70:1111-1118. [PMID: 39225106 DOI: 10.1002/mus.28237] [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: 07/28/2023] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION/AIMS Swim training and regulation of copper metabolism result in clinical benefits in amyotrophic lateral sclerosis (ALS) mice. Therefore, the study aimed to determine whether swim training improves copper metabolism by modifying copper metabolism in the skeletal muscles of ALS mice. METHODS SOD1G93A mice (n = 6 per group) were used as the ALS model, and wild-type B6SJL (WT) mice as controls (n = 6). Mice with ALS were analyzed before the onset of ALS (ALS BEFORE), at baseline ALS (first disease symptoms, trained and untrained, ALS ONSET), and at the end of ALS (last stage disease, trained and untrained, ALS TERMINAL). Copper concentrations and the level of copper metabolism proteins in the skeletal muscles of the lower leg were determined. RESULTS ALS disease caused a reduction in the copper concentration in ALS TERMINAL untrained mice compared with the ALS BEFORE (10.43 ± 1.81 and 38.67 ± 11.50 μg/mg, respectively, p = .0213). The copper chaperon for SOD1 protein, which supplies copper to SOD1, and ATPase7a protein (copper exporter), increased at the terminal stage of disease by 57% (p = .0021) and 34% (p = .0372), while the CTR1 protein (copper importer) decreased by 45% (p = .002). Swim training moderately affected the copper concentration and the concentrations of proteins responsible for copper metabolism in skeletal muscles. DISCUSSION The results show disturbances in skeletal muscle copper metabolism associated with ALS progression, which is moderately affected by swim training. From a clinical point of view, exercise in water for ALS patients should be an essential element of rehabilitation for maintaining quality of life.
Collapse
Affiliation(s)
| | - Damian Jozef Flis
- Department of Pharmaceutical Pathophysiology, Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland
| | - Banu Akdogan
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Andzelika Borkowska
- Department of Bioenergetics and Physiology of Exercise, Faculty of Health Sciences, Medical University of Gdansk, Gdansk, Poland
| | - Mariusz Roman Wieckowski
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Jedrzej Antosiewicz
- Department of Bioenergetics and Physiology of Exercise, Faculty of Health Sciences, Medical University of Gdansk, Gdansk, Poland
| | - Hans Zischka
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Toxicology and Environmental Hygiene, Technical University Munich School of Medicine, Munich, Germany
| | | | - Jan Jacek Kaczor
- Department of Animal and Human Physiology, University of Gdansk, Gdansk, Poland
| | - Wieslaw Ziolkowski
- Department of Rehabilitation Medicine, Faculty of Health Sciences, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
12
|
Zhao Y, Li X, Wang K, Iyer G, Sakowski SA, Zhao L, Teener S, Bakulski KM, Dou JF, Traynor BJ, Karnovsky A, Batterman SA, Feldman EL, Sartor MA, Goutman SA. Epigenetic age acceleration is associated with occupational exposures, sex, and survival in amyotrophic lateral sclerosis. EBioMedicine 2024; 109:105383. [PMID: 39369616 PMCID: PMC11491892 DOI: 10.1016/j.ebiom.2024.105383] [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: 02/15/2024] [Revised: 09/16/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is linked to ageing and genetic and environmental risk factors, yet underlying mechanisms are incompletely understood. We aimed to evaluate epigenetic age acceleration (EAA), i.e., DNA methylation (DNAm) age acceleration, and its association with ALS case status and survival. METHODS In this study, we included 428 ALS and 288 control samples collected between 2011 and 2021. We calculated EAA using the GrimAge residual method from ALS and control blood samples and grouped participants with ALS into three ageing groups (fast, normal, slow). We associated EAA with ALS case status and survival, stratified by sex, and correlated it with environmental and biological factors through occupational exposure assessments, immune cell proportions, and transcriptome changes. FINDINGS Participants with ALS had higher average EAA by 1.80 ± 0.30 years (p < 0.0001) versus controls. Participants with ALS in the fast ageing group had a hazard ratio of 1.52 (95% confidence interval 1.16-2.00, p = 0.0028) referenced to the normal ageing group. In males, this hazard ratio was 1.55 (95% confidence interval 1.11-2.17, p = 0.010), and EAA was positively correlated with high-risk occupational exposures including particulate matter (adj.p < 0.0001) and metals (adj.p = 0.0087). Also, in male participants with ALS, EAA was positively correlated with neutrophil proportions and was negatively correlated with CD4+ T cell proportions. Pathways dysregulated in participants with ALS with fast ageing included spliceosome, nucleocytoplasmic transport, axon guidance, and interferons. INTERPRETATION EAA was associated with ALS case status and, at least in males, with shorter survival after diagnosis. The effect of EAA on ALS was partially explained by occupational exposures and immune cell proportions in a sex-dependent manner. These findings highlight the complex interactions of ageing and exposures in ALS. FUNDING NIH, CDC/National ALS Registry, ALS Association, Dr. Randall Whitcomb Fund for ALS Genetics, Peter Clark Fund for ALS Research, Sinai Medical Staff Foundation, Scott L. Pranger ALS Clinic Fund, NeuroNetwork Therapeutic Discovery Fund, NeuroNetwork for Emerging Therapies.
Collapse
Affiliation(s)
- Yue Zhao
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Xiayan Li
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Kai Wang
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Gayatri Iyer
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Stacey A Sakowski
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA; NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, USA
| | - Lili Zhao
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Samuel Teener
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, USA
| | - Kelly M Bakulski
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - John F Dou
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Bryan J Traynor
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Alla Karnovsky
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Stuart A Batterman
- Environmental Health Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA; NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, USA
| | - Maureen A Sartor
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA; Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA.
| | - Stephen A Goutman
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA; NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
13
|
Al-Khayri JM, Ravindran M, Banadka A, Vandana CD, Priya K, Nagella P, Kukkemane K. Amyotrophic Lateral Sclerosis: Insights and New Prospects in Disease Pathophysiology, Biomarkers and Therapies. Pharmaceuticals (Basel) 2024; 17:1391. [PMID: 39459030 PMCID: PMC11510162 DOI: 10.3390/ph17101391] [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: 09/02/2024] [Revised: 09/30/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a severe neurodegenerative disorder marked by the gradual loss of motor neurons, leading to significant disability and eventual death. Despite ongoing research, there are still limited treatment options, underscoring the need for a deeper understanding of the disease's complex mechanisms and the identification of new therapeutic targets. This review provides a thorough examination of ALS, covering its epidemiology, pathology, and clinical features. It investigates the key molecular mechanisms, such as protein aggregation, neuroinflammation, oxidative stress, and excitotoxicity that contribute to motor neuron degeneration. The role of biomarkers is highlighted for their importance in early diagnosis and disease monitoring. Additionally, the review explores emerging therapeutic approaches, including inhibitors of protein aggregation, neuroinflammation modulators, antioxidant therapies, gene therapy, and stem cell-based treatments. The advantages and challenges of these strategies are discussed, with an emphasis on the potential for precision medicine to tailor treatments to individual patient needs. Overall, this review aims to provide a comprehensive overview of the current state of ALS research and suggest future directions for developing effective therapies.
Collapse
Affiliation(s)
- Jameel M. Al-Khayri
- Department of Agricultural Biotechnology, College of Agriculture and Food Sciences, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Mamtha Ravindran
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed-to-be-University), Bangalore 560027, India; (M.R.); (A.B.); (C.D.V.); (K.P.)
| | - Akshatha Banadka
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed-to-be-University), Bangalore 560027, India; (M.R.); (A.B.); (C.D.V.); (K.P.)
| | - Chendanda Devaiah Vandana
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed-to-be-University), Bangalore 560027, India; (M.R.); (A.B.); (C.D.V.); (K.P.)
| | - Kushalva Priya
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed-to-be-University), Bangalore 560027, India; (M.R.); (A.B.); (C.D.V.); (K.P.)
| | - Praveen Nagella
- Department of Life Sciences, School of Sciences, Christ University, Bengaluru 560029, India;
| | - Kowshik Kukkemane
- Department of Life Sciences, School of Sciences, Christ University, Bengaluru 560029, India;
| |
Collapse
|
14
|
Zhang LY, Zhang SY, Wen R, Zhang TN, Yang N. Role of histone deacetylases and their inhibitors in neurological diseases. Pharmacol Res 2024; 208:107410. [PMID: 39276955 DOI: 10.1016/j.phrs.2024.107410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
Histone deacetylases (HDACs) are zinc-dependent deacetylases that remove acetyl groups from lysine residues of histones or form protein complexes with other proteins for transcriptional repression, changing chromatin structure tightness, and inhibiting gene expression. Recent in vivo and in vitro studies have amply demonstrated the critical role of HDACs in the cell biology of the nervous system during both physiological and pathological processes and have provided new insights into the conduct of research on neurological disease targets. In addition, in vitro and in vivo studies on HDAC inhibitors show promise for the treatment of various diseases. This review summarizes the regulatory mechanisms of HDAC and the important role of its downstream targets in nervous system diseases, and summarizes the therapeutic mechanisms and efficacy of HDAC inhibitors in various nervous system diseases. Additionally, the current pharmacological situation, problems, and developmental prospects of HDAC inhibitors are described. A better understanding of the pathogenic mechanisms of HDACs in the nervous system may reveal new targets for therapeutic interventions in diseases and help to relieve healthcare pressure through preventive measures.
Collapse
Affiliation(s)
- Li-Ying Zhang
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Sen-Yu Zhang
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ri Wen
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Tie-Ning Zhang
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Ni Yang
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
15
|
Jacob SM, Lee S, Kim SH, Sharkey KA, Pfeffer G, Nguyen MD. Brain-body mechanisms contribute to sexual dimorphism in amyotrophic lateral sclerosis. Nat Rev Neurol 2024; 20:475-494. [PMID: 38965379 DOI: 10.1038/s41582-024-00991-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2024] [Indexed: 07/06/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is the most common form of human motor neuron disease. It is characterized by the progressive degeneration of upper and lower motor neurons, leading to generalized motor weakness and, ultimately, respiratory paralysis and death within 3-5 years. The disease is shaped by genetics, age, sex and environmental stressors, but no cure or routine biomarkers exist for the disease. Male individuals have a higher propensity to develop ALS, and a different manifestation of the disease phenotype, than female individuals. However, the mechanisms underlying these sex differences remain a mystery. In this Review, we summarize the epidemiology of ALS, examine the sexually dimorphic presentation of the disease and highlight the genetic variants and molecular pathways that might contribute to sex differences in humans and animal models of ALS. We advance the idea that sexual dimorphism in ALS arises from the interactions between the CNS and peripheral organs, involving vascular, metabolic, endocrine, musculoskeletal and immune systems, which are strikingly different between male and female individuals. Finally, we review the response to treatments in ALS and discuss the potential to implement future personalized therapeutic strategies for the disease.
Collapse
Affiliation(s)
- Sarah M Jacob
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Sukyoung Lee
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Seung Hyun Kim
- Department of Neurology, Hanyang University Hospital, Seoul, South Korea
| | - Keith A Sharkey
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Gerald Pfeffer
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| | - Minh Dang Nguyen
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
16
|
Weinreich M, McDonough H, Yacovzada N, Magen I, Cohen Y, Harvey C, Gornall S, Boddy S, Alix J, Mohseni N, Kurz JM, Kenna KP, Zhang S, Iacoangeli A, Al-Khleifat A, Snyder MP, Hobson E, Al-Chalabi A, Hornstein E, Elhaik E, Shaw PJ, McDermott C, Cooper-Knock J. predicTTE: An accessible and optimal tool for time-to-event prediction in neurological diseases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.20.604416. [PMID: 39091819 PMCID: PMC11291041 DOI: 10.1101/2024.07.20.604416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Time-to-event prediction is a key task for biological discovery, experimental medicine, and clinical care. This is particularly true for neurological diseases where development of reliable biomarkers is often limited by difficulty visualising and sampling relevant cell and molecular pathobiology. To date, much work has relied on Cox regression because of ease-of-use, despite evidence that this model includes incorrect assumptions. We have implemented a set of deep learning and spline models for time-to-event modelling within a fully customizable 'app' and accompanying online portal, both of which can be used for any time-to-event analysis in any disease by a non-expert user. Our online portal includes capacity for end-users including patients, Neurology clinicians, and researchers, to access and perform predictions using a trained model, and to contribute new data for model improvement, all within a data-secure environment. We demonstrate a pipeline for use of our app with three use-cases including imputation of missing data, hyperparameter tuning, model training and independent validation. We show that predictions are optimal for use in downstream applications such as genetic discovery, biomarker interpretation, and personalised choice of medication. We demonstrate the efficiency of an ensemble configuration, including focused training of a deep learning model. We have optimised a pipeline for imputation of missing data in combination with time-to-event prediction models. Overall, we provide a powerful and accessible tool to develop, access and share time-to-event prediction models; all software and tutorials are available at www.predictte.org.
Collapse
Affiliation(s)
- Marcel Weinreich
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, UK
- Department of Clinical Neurobiology at the German Cancer Research Center (DKFZ) and the Medical Faculty of the Heidelberg University, Heidelberg, Germany
| | - Harry McDonough
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, UK
| | - Nancy Yacovzada
- Department of Molecular Genetics and Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Iddo Magen
- Department of Molecular Genetics and Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Yahel Cohen
- Department of Molecular Genetics and Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Calum Harvey
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, UK
| | - Sarah Gornall
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, UK
| | - Sarah Boddy
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, UK
| | - James Alix
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, UK
- NIHR Sheffield Biomedical Research Centre, Royal Hallamshire Hospital, Glossop Road, Sheffield S10 2JF, UK
| | | | - Julian M Kurz
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, UK
| | - Kevin P Kenna
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sai Zhang
- Department of Epidemiology, University of Florida, Gainesville, FL, USA
| | - Alfredo Iacoangeli
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, London, UK
| | - Ahmad Al-Khleifat
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, London, UK
| | - Michael P Snyder
- Center for Genomics and Personalized Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Esther Hobson
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, UK
| | - Ammar Al-Chalabi
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, London, UK
| | - Eran Hornstein
- Department of Molecular Genetics and Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Elhaik
- Department of Biology, Lund University, Sweden
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, UK
- NIHR Sheffield Biomedical Research Centre, Royal Hallamshire Hospital, Glossop Road, Sheffield S10 2JF, UK
| | - Christopher McDermott
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, UK
- NIHR Sheffield Biomedical Research Centre, Royal Hallamshire Hospital, Glossop Road, Sheffield S10 2JF, UK
| | - Johnathan Cooper-Knock
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, UK
- NIHR Sheffield Biomedical Research Centre, Royal Hallamshire Hospital, Glossop Road, Sheffield S10 2JF, UK
| |
Collapse
|
17
|
Niccolai E, Pedone M, Martinelli I, Nannini G, Baldi S, Simonini C, Di Gloria L, Zucchi E, Ramazzotti M, Spezia PG, Maggi F, Quaranta G, Masucci L, Bartolucci G, Stingo FC, Mandrioli J, Amedei A. Amyotrophic lateral sclerosis stratification: unveiling patterns with virome, inflammation, and metabolism molecules. J Neurol 2024; 271:4310-4325. [PMID: 38644373 PMCID: PMC11233352 DOI: 10.1007/s00415-024-12348-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/23/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is an untreatable and clinically heterogeneous condition primarily affecting motor neurons. The ongoing quest for reliable biomarkers that mirror the disease status and progression has led to investigations that extend beyond motor neurons' pathology, encompassing broader systemic factors such as metabolism, immunity, and the microbiome. Our study contributes to this effort by examining the potential role of microbiome-related components, including viral elements, such as torque tenovirus (TTV), and various inflammatory factors, in ALS. In our analysis of serum samples from 100 ALS patients and 34 healthy controls (HC), we evaluated 14 cytokines, TTV DNA load, and 18 free fatty acids (FFA). We found that the evaluated variables are effective in differentiating ALS patients from healthy controls. In addition, our research identifies four unique patient clusters, each characterized by distinct biological profiles. Intriguingly, no correlations were found with site of onset, sex, progression rate, phenotype, or C9ORF72 expansion. A remarkable aspect of our findings is the discovery of a gender-specific relationship between levels of 2-ethylhexanoic acid and patient survival. In addition to contributing to the growing body of evidence suggesting altered peripheral immune responses in ALS, our exploratory research underscores metabolic diversity challenging conventional clinical classifications. If our exploratory findings are validated by further research, they could significantly impact disease understanding and patient care customization. Identifying groups based on biological profiles might aid in clustering patients with varying responses to treatments.
Collapse
Affiliation(s)
- Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Matteo Pedone
- Department of Statistics, Computer Science, Applications "G. Parenti", University of Florence, Florence, Italy
| | - Ilaria Martinelli
- Neurology Unit, Department of Neuroscience, Azienda Ospedaliero Universitaria Di Modena, Modena, Italy
| | - Giulia Nannini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Simone Baldi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Cecilia Simonini
- Neurology Unit, Department of Neuroscience, Azienda Ospedaliero Universitaria Di Modena, Modena, Italy
| | - Leandro Di Gloria
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Elisabetta Zucchi
- Neurology Unit, Department of Neuroscience, Azienda Ospedaliero Universitaria Di Modena, Modena, Italy
| | - Matteo Ramazzotti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Pietro Giorgio Spezia
- Department of Translational Research, Retrovirus Center - University of Pisa, Pisa, Italy
| | - Fabrizio Maggi
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani - IRCCS, Rome, Italy
| | - Gianluca Quaranta
- Department of Laboratory and Infectious Sciences, A. Gemelli University Hospital IRCCS, Rome, Italy
| | - Luca Masucci
- Department of Laboratory and Infectious Sciences, A. Gemelli University Hospital IRCCS, Rome, Italy
| | - Gianluca Bartolucci
- Department of Neurosciences, Psychology, Drug Research and Child Health Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Florence, Italy
| | - Francesco Claudio Stingo
- Department of Statistics, Computer Science, Applications "G. Parenti", University of Florence, Florence, Italy
| | - Jessica Mandrioli
- Neurology Unit, Department of Neuroscience, Azienda Ospedaliero Universitaria Di Modena, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.
| |
Collapse
|
18
|
Parvanovova P, Hnilicova P, Kolisek M, Tatarkova Z, Halasova E, Kurca E, Holubcikova S, Koprusakova MT, Baranovicova E. Disturbances in Muscle Energy Metabolism in Patients with Amyotrophic Lateral Sclerosis. Metabolites 2024; 14:356. [PMID: 39057679 PMCID: PMC11278632 DOI: 10.3390/metabo14070356] [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: 05/22/2024] [Revised: 06/17/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neuromuscular disease type of motor neuron disorder characterized by degeneration of the upper and lower motor neurons resulting in dysfunction of the somatic muscles of the body. The ALS condition is manifested in progressive skeletal muscle atrophy and spasticity. It leads to death, mostly due to respiratory failure. Within the pathophysiology of the disease, muscle energy metabolism seems to be an important part. In our study, we used blood plasma from 25 patients with ALS diagnosed by definitive El Escorial criteria according to ALSFR-R (Revised Amyotrophic Lateral Sclerosis Functional Rating Scale) criteria and 25 age and sex-matched subjects. Aside from standard clinical biochemical parameters, we used the NMR (nuclear magnetic resonance) metabolomics approach to determine relative plasma levels of metabolites. We observed a decrease in total protein level in blood; however, despite accelerated skeletal muscle catabolism characteristic for ALS patients, we did not detect changes in plasma levels of essential amino acids. When focused on alterations in energy metabolism within muscle, compromised creatine uptake was accompanied by decreased plasma creatinine. We did not observe changes in plasma levels of BCAAs (branched chain amino acids; leucine, isoleucine, valine); however, the observed decrease in plasma levels of all three BCKAs (branched chain alpha-keto acids derived from BCAAs) suggests enhanced utilization of BCKAs as energy substrate. Glutamine, found to be increased in blood plasma in ALS patients, besides serving for ammonia detoxification, could also be considered a potential TCA (tricarboxylic acid) cycle contributor in times of decreased pyruvate utilization. When analyzing the data by using a cross-validated Random Forest algorithm, it finished with an AUC of 0.92, oob error of 8%, and an MCC (Matthew's correlation coefficient) of 0.84 when relative plasma levels of metabolites were used as input variables. Although the discriminatory power of the system used was promising, additional features are needed to create a robust discriminatory model.
Collapse
Affiliation(s)
- Petra Parvanovova
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia; (P.P.); (Z.T.); (S.H.)
| | - Petra Hnilicova
- Biomedical Centre Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia; (P.H.); (M.K.); (E.H.)
| | - Martin Kolisek
- Biomedical Centre Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia; (P.H.); (M.K.); (E.H.)
| | - Zuzana Tatarkova
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia; (P.P.); (Z.T.); (S.H.)
| | - Erika Halasova
- Biomedical Centre Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia; (P.H.); (M.K.); (E.H.)
| | - Egon Kurca
- Department of Neurology, University Hospital Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Kollarova 2, 036 01 Martin, Slovakia;
| | - Simona Holubcikova
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia; (P.P.); (Z.T.); (S.H.)
| | - Monika Turcanova Koprusakova
- Department of Neurology, University Hospital Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Kollarova 2, 036 01 Martin, Slovakia;
| | - Eva Baranovicova
- Biomedical Centre Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia; (P.H.); (M.K.); (E.H.)
| |
Collapse
|
19
|
Murage B, Tan H, Mashimo T, Jackson M, Skehel PA. Spinal cord neurone loss and foot placement changes in a rat knock-in model of amyotrophic lateral sclerosis Type 8. Brain Commun 2024; 6:fcae184. [PMID: 38846532 PMCID: PMC11154649 DOI: 10.1093/braincomms/fcae184] [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: 08/08/2023] [Revised: 04/10/2024] [Accepted: 05/23/2024] [Indexed: 06/09/2024] Open
Abstract
Amyotrophic lateral sclerosis is an age-dependent cell type-selective degenerative disease. Genetic studies indicate that amyotrophic lateral sclerosis is part of a spectrum of disorders, ranging from spinal muscular atrophy to frontotemporal dementia that share common pathological mechanisms. Amyotrophic lateral sclerosis Type 8 is a familial disease caused by mis-sense mutations in VAPB. VAPB is localized to the cytoplasmic surface of the endoplasmic reticulum, where it serves as a docking point for cytoplasmic proteins and mediates inter-organelle interactions with the endoplasmic reticulum membrane. A gene knock-in model of amyotrophic lateral sclerosis Type 8 based on the VapBP56S mutation and VapB gene deletion has been generated in rats. These animals display a range of age-dependent phenotypes distinct from those previously reported in mouse models of amyotrophic lateral sclerosis Type 8. A loss of motor neurones in VapBP56S/+ and VapBP56S/P56S animals is indicated by a reduction in the number of large choline acetyl transferase-staining cells in the spinal cord. VapB-/- animals exhibit a relative increase in cytoplasmic TDP-43 levels compared with the nucleus, but no large protein aggregates. Concomitant with these spinal cord pathologies VapBP56S/+ , VapBP56S/P56S and VapB-/- animals exhibit age-dependent changes in paw placement and exerted pressures when traversing a CatWalk apparatus, consistent with a somatosensory dysfunction. Extramotor dysfunction is reported in half the cases of motor neurone disease, and this is the first indication of an associated sensory dysfunction in a rodent model of amyotrophic lateral sclerosis. Different rodent models may offer complementary experimental platforms with which to understand the human disease.
Collapse
Affiliation(s)
- Brenda Murage
- Centre for Discovery Brain Sciences, Edinburgh University, Edinburgh EH8 9XD, UK
- Euan MacDonald Centre for MND Research, Edinburgh University, Edinburgh EH16 4SB, UK
| | - Han Tan
- Centre for Discovery Brain Sciences, Edinburgh University, Edinburgh EH8 9XD, UK
| | - Tomoji Mashimo
- Division of Animal Genetics, Laboratory Animal Research Center, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Mandy Jackson
- Centre for Discovery Brain Sciences, Edinburgh University, Edinburgh EH8 9XD, UK
- Euan MacDonald Centre for MND Research, Edinburgh University, Edinburgh EH16 4SB, UK
| | - Paul A Skehel
- Centre for Discovery Brain Sciences, Edinburgh University, Edinburgh EH8 9XD, UK
- Euan MacDonald Centre for MND Research, Edinburgh University, Edinburgh EH16 4SB, UK
| |
Collapse
|
20
|
Nguyen L. Updates on Disease Mechanisms and Therapeutics for Amyotrophic Lateral Sclerosis. Cells 2024; 13:888. [PMID: 38891021 PMCID: PMC11172142 DOI: 10.3390/cells13110888] [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: 03/29/2024] [Revised: 05/08/2024] [Accepted: 05/15/2024] [Indexed: 06/20/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS), or Lou Gehrig's disease, is a motor neuron disease. In ALS, upper and lower motor neurons in the brain and spinal cord progressively degenerate during the course of the disease, leading to the loss of the voluntary movement of the arms and legs. Since its first description in 1869 by a French neurologist Jean-Martin Charcot, the scientific discoveries on ALS have increased our understanding of ALS genetics, pathology and mechanisms and provided novel therapeutic strategies. The goal of this review article is to provide a comprehensive summary of the recent findings on ALS mechanisms and related therapeutic strategies to the scientific audience. Several highlighted ALS research topics discussed in this article include the 2023 FDA approved drug for SOD1 ALS, the updated C9orf72 GGGGCC repeat-expansion-related mechanisms and therapeutic targets, TDP-43-mediated cryptic splicing and disease markers and diagnostic and therapeutic options offered by these recent discoveries.
Collapse
Affiliation(s)
- Lien Nguyen
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL 32610, USA;
- Center for NeuroGenetics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Genetics Institute, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
21
|
Christoforidou E, Moody L, Joilin G, Simoes FA, Gordon D, Talbot K, Hafezparast M. An ALS-associated mutation dysregulates microglia-derived extracellular microRNAs in a sex-specific manner. Dis Model Mech 2024; 17:dmm050638. [PMID: 38721655 PMCID: PMC11152562 DOI: 10.1242/dmm.050638] [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: 11/30/2023] [Accepted: 04/29/2024] [Indexed: 05/30/2024] Open
Abstract
Evidence suggests the presence of microglial activation and microRNA (miRNA) dysregulation in amyotrophic lateral sclerosis (ALS), the most common form of adult motor neuron disease. However, few studies have investigated whether the miRNA dysregulation originates from microglia. Furthermore, TDP-43 (encoded by TARDBP), involved in miRNA biogenesis, aggregates in tissues of ∼98% of ALS cases. Thus, this study aimed to determine whether expression of the ALS-linked TDP-43M337V mutation in a transgenic mouse model dysregulates microglia-derived miRNAs. RNA sequencing identified several dysregulated miRNAs released by transgenic microglia and a differential miRNA release by lipopolysaccharide-stimulated microglia, which was more pronounced in cells from female mice. We validated the downregulation of three candidate miRNAs, namely, miR-16-5p, miR-99a-5p and miR-191-5p, by reverse transcription quantitative polymerase chain reaction (RT-qPCR) and identified their predicted targets, which primarily include genes involved in neuronal development and function. These results suggest that altered TDP-43 function leads to changes in the miRNA population released by microglia, which may in turn be a source of the miRNA dysregulation observed in the disease. This has important implications for the role of neuroinflammation in ALS pathology and could provide potential therapeutic targets.
Collapse
Affiliation(s)
- Eleni Christoforidou
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| | - Libby Moody
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| | - Greig Joilin
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| | - Fabio A. Simoes
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| | - David Gordon
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 9DU, UK
| | - Kevin Talbot
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 9DU, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, OX1 3QU, UK
| | - Majid Hafezparast
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| |
Collapse
|
22
|
Murdock BJ, Zhao B, Pawlowski KD, Famie JP, Piecuch CE, Webber-Davis IF, Teener SJ, Feldman EL, Zhao L, Goutman SA. Peripheral Immune Profiles Predict ALS Progression in an Age- and Sex-Dependent Manner. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200241. [PMID: 38626361 PMCID: PMC11087030 DOI: 10.1212/nxi.0000000000200241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 02/12/2024] [Indexed: 04/18/2024]
Abstract
BACKGROUND AND OBJECTIVES Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease whose pathobiology associates with peripheral blood immune cell levels and activation patterns in an age and sex-dependent manner. This study's objective was to identify immune profile associations with ALS progression, whether the associations are age and sex-specific, and whether immune profiles can predict a future disease course. METHODS Flow cytometry immune profiles (a combination of 22 peripheral blood immune markers) were generated for 241 participants with ALS and linked to ALS progression, using progression-free survival, which is a composite combining the revised ALS Functional Rating Scale and survival. Participants were first grouped by immune profiles using unsupervised hierarchical clustering, and clusters were associated with subsequent progression-free survival. Next, individual immune markers were associated with progression-free survival using least absolute shrinkage and selection operator-Cox regression. Analyses were stratified by age and sex to identify demographic-specific immune mechanisms. Finally, random forest determined the predictive power of immune profiles on ALS progression in the whole population and again stratified by age and sex. RESULTS Progression-free survival differed between clusters of participants with similar immune profiles, particularly reduced natural killer (NK)-cell activation associated with slower progression. Individual markers such as neutrophil levels and NK-cell NKp46 expression associated with faster ALS progression while overall NK-cell levels and NK-cell subpopulations associated with slower progression; the strength of these associations varied by age and sex. Adding these immune markers to prediction models dramatically increased short-term prediction compared with routine clinical prognostic variables alone, and the addition of NK-cell markers further improved the prediction accuracy in female participants. DISCUSSION Specific immune profiles likely contribute to ALS progression in an age and sex-dependent manner, and peripheral immune markers enhance the prediction of short-term clinical outcomes. These findings suggest a complex milieu of immune profiles associated with ALS progression, and more detailed immunophenotyping in ALS will facilitate personalized immunotherapeutics in ALS.
Collapse
Affiliation(s)
- Benjamin J Murdock
- From the Department of Neurology (B.J.M., K.D.P., J.P.F., C.E.P., I.F.W.-D., S.J.T., E.L.F., S.A.G.); and School of Public Health (B.Z., L.Z.), Biostatistics Department, University of Michigan, Ann Arbor
| | - Bangyao Zhao
- From the Department of Neurology (B.J.M., K.D.P., J.P.F., C.E.P., I.F.W.-D., S.J.T., E.L.F., S.A.G.); and School of Public Health (B.Z., L.Z.), Biostatistics Department, University of Michigan, Ann Arbor
| | - Kristen D Pawlowski
- From the Department of Neurology (B.J.M., K.D.P., J.P.F., C.E.P., I.F.W.-D., S.J.T., E.L.F., S.A.G.); and School of Public Health (B.Z., L.Z.), Biostatistics Department, University of Michigan, Ann Arbor
| | - Joshua P Famie
- From the Department of Neurology (B.J.M., K.D.P., J.P.F., C.E.P., I.F.W.-D., S.J.T., E.L.F., S.A.G.); and School of Public Health (B.Z., L.Z.), Biostatistics Department, University of Michigan, Ann Arbor
| | - Caroline E Piecuch
- From the Department of Neurology (B.J.M., K.D.P., J.P.F., C.E.P., I.F.W.-D., S.J.T., E.L.F., S.A.G.); and School of Public Health (B.Z., L.Z.), Biostatistics Department, University of Michigan, Ann Arbor
| | - Ian F Webber-Davis
- From the Department of Neurology (B.J.M., K.D.P., J.P.F., C.E.P., I.F.W.-D., S.J.T., E.L.F., S.A.G.); and School of Public Health (B.Z., L.Z.), Biostatistics Department, University of Michigan, Ann Arbor
| | - Samuel J Teener
- From the Department of Neurology (B.J.M., K.D.P., J.P.F., C.E.P., I.F.W.-D., S.J.T., E.L.F., S.A.G.); and School of Public Health (B.Z., L.Z.), Biostatistics Department, University of Michigan, Ann Arbor
| | - Eva L Feldman
- From the Department of Neurology (B.J.M., K.D.P., J.P.F., C.E.P., I.F.W.-D., S.J.T., E.L.F., S.A.G.); and School of Public Health (B.Z., L.Z.), Biostatistics Department, University of Michigan, Ann Arbor
| | - Lili Zhao
- From the Department of Neurology (B.J.M., K.D.P., J.P.F., C.E.P., I.F.W.-D., S.J.T., E.L.F., S.A.G.); and School of Public Health (B.Z., L.Z.), Biostatistics Department, University of Michigan, Ann Arbor
| | - Stephen A Goutman
- From the Department of Neurology (B.J.M., K.D.P., J.P.F., C.E.P., I.F.W.-D., S.J.T., E.L.F., S.A.G.); and School of Public Health (B.Z., L.Z.), Biostatistics Department, University of Michigan, Ann Arbor
| |
Collapse
|
23
|
Pinilla-González V, Montecinos-Barrientos B, Martin-Kommer C, Chichiarelli S, Saso L, Rodrigo R. Exploring antioxidant strategies in the pathogenesis of ALS. Open Life Sci 2024; 19:20220842. [PMID: 38585631 PMCID: PMC10997151 DOI: 10.1515/biol-2022-0842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/05/2024] [Accepted: 02/20/2024] [Indexed: 04/09/2024] Open
Abstract
The central nervous system is essential for maintaining homeostasis and controlling the body's physiological functions. However, its biochemical characteristics make it highly vulnerable to oxidative damage, which is a common factor in neurodegenerative diseases like amyotrophic lateral sclerosis (ALS). ALS is a leading cause of motor neuron disease, characterized by a rapidly progressing and incurable condition. ALS often results in death from respiratory failure within 3-5 years from the onset of the first symptoms, underscoring the urgent need to address this medical challenge. The aim of this study is to present available data supporting the role of oxidative stress in the mechanisms underlying ALS and to discuss potential antioxidant therapies currently in development. These therapies aim to improve the quality of life and life expectancy for patients affected by this devastating disease.
Collapse
Affiliation(s)
- Víctor Pinilla-González
- Faculty of Medicine, Institute of Biomedical Sciences, University of Chile, Santiago8380000, Chile
| | | | - Clemente Martin-Kommer
- Faculty of Medicine, Institute of Biomedical Sciences, University of Chile, Santiago8380000, Chile
| | - Silvia Chichiarelli
- Department of Biochemical Sciences “A. Rossi-Fanelli”, Sapienza University of Rome, 00185Rome, Italy
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Faculty of Pharmacy and Medicine, Sapienza University, P.le Aldo Moro 5, 00185Rome, Italy
| | - Ramón Rodrigo
- Faculty of Medicine, Institute of Biomedical Sciences, University of Chile, Santiago8380000, Chile
| |
Collapse
|
24
|
Bjørklund G, Đorđević AB, Hamdan H, Wallace DR, Peana M. Metal-induced autoimmunity in neurological disorders: A review of current understanding and future directions. Autoimmun Rev 2024; 23:103509. [PMID: 38159894 DOI: 10.1016/j.autrev.2023.103509] [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: 12/08/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
Autoimmunity is a multifaceted disorder influenced by both genetic and environmental factors, and metal exposure has been implicated as a potential catalyst, especially in autoimmune diseases affecting the central nervous system. Notably, metals like mercury, lead, and aluminum exhibit well-established neurotoxic effects, yet the precise mechanisms by which they elicit autoimmune responses in susceptible individuals remain unclear. Recent studies propose that metal-induced autoimmunity may arise from direct toxic effects on immune cells and tissues, coupled with indirect impacts on the gut microbiome and the blood-brain barrier. These effects can activate self-reactive T cells, prompting the production of autoantibodies, inflammatory responses, and tissue damage. Diagnosing metal-induced autoimmunity proves challenging due to nonspecific symptoms and a lack of reliable biomarkers. Treatment typically involves chelation therapy to eliminate excess metals and immunomodulatory agents to suppress autoimmune responses. Prevention strategies include lifestyle adjustments to reduce metal exposure and avoiding occupational and environmental risks. Prognosis is generally favorable with proper treatment; however, untreated cases may lead to autoimmune disorder progression and irreversible organ damage, particularly in the brain. Future research aims to identify genetic and environmental risk factors, enhance diagnostic precision, and explore novel treatment approaches for improved prevention and management of this intricate and debilitating disease.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Mo i Rana, Norway.
| | | | - Halla Hamdan
- Department of Pharmacology, Oklahoma State University Center for Health Sciences, Tulsa, OK, United States
| | - David R Wallace
- Department of Pharmacology, Oklahoma State University Center for Health Sciences, Tulsa, OK, United States
| | - Massimiliano Peana
- Department of Chemical, Physical, Mathematical and Natural Sciences, University of Sassari, Italy.
| |
Collapse
|
25
|
Zamani A, Thomas E, Wright DK. Sex biology in amyotrophic lateral sclerosis. Ageing Res Rev 2024; 95:102228. [PMID: 38354985 DOI: 10.1016/j.arr.2024.102228] [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: 08/31/2023] [Revised: 02/09/2024] [Accepted: 02/09/2024] [Indexed: 02/16/2024]
Abstract
Although sex differences in amyotrophic lateral sclerosis (ALS) have not been studied systematically, numerous clinical and preclinical studies have shown sex to be influential in disease prognosis. Moreover, with the development of advanced imaging tools, the difference between male and female brain in structure and function and their response to neurodegeneration are more definitive. As discussed in this review, ALS patients exhibit a sex bias pertaining to the features of the disease, and their clinical, pathological, (and pathophysiological) phenotypes. Several epidemiological studies have indicated that this sex disparity stems from various aetiologies, including sex-specific brain structure and neural functioning, genetic predisposition, age, gonadal hormones, susceptibility to traumatic brain injury (TBI)/head trauma and lifestyle factors.
Collapse
Affiliation(s)
- Akram Zamani
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia.
| | - Emma Thomas
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - David K Wright
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| |
Collapse
|
26
|
Limone F, Couto A, Wang JY, Zhang Y, McCourt B, Huang C, Minkin A, Jani M, McNeer S, Keaney J, Gillet G, Gonzalez RL, Goodman WA, Kadiu I, Eggan K, Burberry A. Myeloid and lymphoid expression of C9orf72 regulates IL-17A signaling in mice. Sci Transl Med 2024; 16:eadg7895. [PMID: 38295187 PMCID: PMC11247723 DOI: 10.1126/scitranslmed.adg7895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 01/10/2024] [Indexed: 02/02/2024]
Abstract
A mutation in C9ORF72 is the most common cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Patients with ALS or FTD often develop autoimmunity and inflammation that precedes or coincides with the onset of neurological symptoms, but the underlying mechanisms are poorly understood. Here, we knocked out murine C9orf72 in seven hematopoietic progenitor compartments by conditional mutagenesis and found that myeloid lineage C9orf72 prevents splenomegaly, loss of tolerance, and premature mortality. Furthermore, we demonstrated that C9orf72 plays a role in lymphoid cells to prevent interleukin-17A (IL-17A) production and neutrophilia. Mass cytometry identified early and sustained elevation of the costimulatory molecule CD80 expressed on C9orf72-deficient mouse macrophages, monocytes, and microglia. Enrichment of CD80 was similarly observed in human spinal cord microglia from patients with C9ORF72-mediated ALS compared with non-ALS controls. Single-cell RNA sequencing of murine spinal cord, brain cortex, and spleen demonstrated coordinated induction of gene modules related to antigen processing and presentation and antiviral immunity in C9orf72-deficient endothelial cells, microglia, and macrophages. Mechanistically, C9ORF72 repressed the trafficking of CD80 to the cell surface in response to Toll-like receptor agonists, interferon-γ, and IL-17A. Deletion of Il17a in C9orf72-deficient mice prevented CD80 enrichment in the spinal cord, reduced neutrophilia, and reduced gut T helper type 17 cells. Last, systemic delivery of an IL-17A neutralizing antibody augmented motor performance and suppressed neuroinflammation in C9orf72-deficient mice. Altogether, we show that C9orf72 orchestrates myeloid costimulatory potency and provide support for IL-17A as a therapeutic target for neuroinflammation associated with ALS or FTD.
Collapse
Affiliation(s)
- Francesco Limone
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
- Leiden University Medical Center, LUMC, 2333 ZA Leiden, The Netherlands
| | - Alexander Couto
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
| | - Jin-Yuan Wang
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
| | - Yingying Zhang
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
| | - Blake McCourt
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Cerianne Huang
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Adina Minkin
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Marghi Jani
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Sarah McNeer
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - James Keaney
- Neuroinflammation Focus Area, UCB Biopharma SRL, Braine-l’Alleud, 1420, Belgium
| | - Gaëlle Gillet
- Neuroinflammation Focus Area, UCB Biopharma SRL, Braine-l’Alleud, 1420, Belgium
| | - Rodrigo Lopez Gonzalez
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44196, USA
| | - Wendy A. Goodman
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Irena Kadiu
- Neuroinflammation Focus Area, UCB Biopharma SRL, Braine-l’Alleud, 1420, Belgium
| | - Kevin Eggan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
| | - Aaron Burberry
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| |
Collapse
|
27
|
Dey B, Kumar A, Patel AB. Pathomechanistic Networks of Motor System Injury in Amyotrophic Lateral Sclerosis. Curr Neuropharmacol 2024; 22:1778-1806. [PMID: 37622689 PMCID: PMC11284732 DOI: 10.2174/1570159x21666230824091601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/25/2023] [Accepted: 06/06/2023] [Indexed: 08/26/2023] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is the most common, adult-onset, progressive motor neurodegenerative disorder that results in death within 3 years of the clinical diagnosis. Due to the clinicopathological heterogeneity, any reliable biomarkers for diagnosis or prognosis of ALS have not been identified till date. Moreover, the only three clinically approved treatments are not uniformly effective in slowing the disease progression. Over the last 15 years, there has been a rapid advancement in research on the complex pathomechanistic landscape of ALS that has opened up new avenues for successful clinical translation of targeted therapeutics. Multiple studies suggest that the age-dependent interaction of risk-associated genes with environmental factors and endogenous modifiers is critical to the multi-step process of ALS pathogenesis. In this review, we provide an updated discussion on the dysregulated cross-talk between intracellular homeostasis processes, the unique molecular networks across selectively vulnerable cell types, and the multisystemic nature of ALS pathomechanisms. Importantly, this work highlights the alteration in epigenetic and epitranscriptomic landscape due to gene-environment interactions, which have been largely overlooked in the context of ALS pathology. Finally, we suggest that precision medicine research in ALS will be largely benefitted from the stratification of patient groups based on the clinical phenotype, onset and progression, genome, exposome, and metabolic identities.
Collapse
Affiliation(s)
- Bedaballi Dey
- CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Hyderabad 500007, Telangana, India
- AcSIR-Academy of Scientific and Innovative Research, Ghaziabad 201002, Uttar Pradesh, India
| | - Arvind Kumar
- CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Hyderabad 500007, Telangana, India
- AcSIR-Academy of Scientific and Innovative Research, Ghaziabad 201002, Uttar Pradesh, India
| | - Anant Bahadur Patel
- CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Hyderabad 500007, Telangana, India
- AcSIR-Academy of Scientific and Innovative Research, Ghaziabad 201002, Uttar Pradesh, India
| |
Collapse
|
28
|
Ludolph AC, Grandjean H, Reviers E, De Micheli V, Bianchi C, Cardosi L, Russ H, Silani V. The preferences of people with amyotrophic lateral sclerosis on riluzole treatment in Europe. Sci Rep 2023; 13:22497. [PMID: 38110502 PMCID: PMC10728064 DOI: 10.1038/s41598-023-49424-3] [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: 03/16/2023] [Accepted: 12/07/2023] [Indexed: 12/20/2023] Open
Abstract
The Patient Preference Survey aims to understand unmet needs related to riluzole management in people with Amyotrophic Lateral Sclerosis (ALS) and to identify which characteristics of a new formulation could better match their preferences. The survey involved 117 people with ALS (PALS) treated with riluzole in four European countries. The dysphagic PALS were least satisfied with the riluzole tablet and oral suspension and with ease in self-administration; up to 68% of respondents postponed or missed the treatment due to swallowing difficulties and need of caregiver assistance. Overall, 51% of tablet and 53% of oral suspension users regularly crushed or mixed riluzole with beverages, respectively; PALS who always manipulated riluzole showed low satisfaction with the formulation and considered the risk of choking and pneumonia the most worrisome event. The survey evaluated the driving factors in choosing/switching the therapy: 67% of PALS declared a low risk of choking. The research finally evaluated which attributes of a new formulation would be preferred: the most relevant were ease of use (4.3/5), convenient/portable packaging (4.0/5) and oral-dissolving properties without tongue motility (3.9/5). The Patient Preference Survey suggests that patients have several unmet needs and preferences that could be addressed by a different formulation, e.g. using oral film technologies.
Collapse
Affiliation(s)
- Albert C Ludolph
- Department of Neurology, German Center for Neurodegenerative Diseases (DZNE), University of Ulm, Ulm, Germany
| | | | - Evy Reviers
- European Organization for Professionals and Patients with ALS (EUpALS), Leuven, Belgium
| | | | | | | | - Hermann Russ
- Sirius Scientific Consulting AG, 8852, Altendorf, Switzerland.
| | - Vincenzo Silani
- Department of Neuroscience and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
29
|
Provenzano F, Torazza C, Bonifacino T, Bonanno G, Milanese M. The Key Role of Astrocytes in Amyotrophic Lateral Sclerosis and Their Commitment to Glutamate Excitotoxicity. Int J Mol Sci 2023; 24:15430. [PMID: 37895110 PMCID: PMC10607805 DOI: 10.3390/ijms242015430] [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: 08/25/2023] [Revised: 10/12/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
In the last two decades, there has been increasing evidence supporting non-neuronal cells as active contributors to neurodegenerative disorders. Among glial cells, astrocytes play a pivotal role in driving amyotrophic lateral sclerosis (ALS) progression, leading the scientific community to focus on the "astrocytic signature" in ALS. Here, we summarized the main pathological mechanisms characterizing astrocyte contribution to MN damage and ALS progression, such as neuroinflammation, mitochondrial dysfunction, oxidative stress, energy metabolism impairment, miRNAs and extracellular vesicles contribution, autophagy dysfunction, protein misfolding, and altered neurotrophic factor release. Since glutamate excitotoxicity is one of the most relevant ALS features, we focused on the specific contribution of ALS astrocytes in this aspect, highlighting the known or potential molecular mechanisms by which astrocytes participate in increasing the extracellular glutamate level in ALS and, conversely, undergo the toxic effect of the excessive glutamate. In this scenario, astrocytes can behave as "producers" and "targets" of the high extracellular glutamate levels, going through changes that can affect themselves and, in turn, the neuronal and non-neuronal surrounding cells, thus actively impacting the ALS course. Moreover, this review aims to point out knowledge gaps that deserve further investigation.
Collapse
Affiliation(s)
- Francesca Provenzano
- Department of Pharmacy (DIFAR), University of Genoa, 16148 Genova, Italy; (F.P.); (C.T.); (G.B.); (M.M.)
| | - Carola Torazza
- Department of Pharmacy (DIFAR), University of Genoa, 16148 Genova, Italy; (F.P.); (C.T.); (G.B.); (M.M.)
| | - Tiziana Bonifacino
- Department of Pharmacy (DIFAR), University of Genoa, 16148 Genova, Italy; (F.P.); (C.T.); (G.B.); (M.M.)
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 56122 Pisa, Italy
| | - Giambattista Bonanno
- Department of Pharmacy (DIFAR), University of Genoa, 16148 Genova, Italy; (F.P.); (C.T.); (G.B.); (M.M.)
| | - Marco Milanese
- Department of Pharmacy (DIFAR), University of Genoa, 16148 Genova, Italy; (F.P.); (C.T.); (G.B.); (M.M.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| |
Collapse
|
30
|
Rowe HP, Shellikeri S, Yunusova Y, Chenausky KV, Green JR. Quantifying articulatory impairments in neurodegenerative motor diseases: A scoping review and meta-analysis of interpretable acoustic features. INTERNATIONAL JOURNAL OF SPEECH-LANGUAGE PATHOLOGY 2023; 25:486-499. [PMID: 36001500 PMCID: PMC9950294 DOI: 10.1080/17549507.2022.2089234] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
PURPOSE Neurodegenerative motor diseases (NMDs) have devastating effects on the lives of patients and their loved ones, in part due to the impact of neurologic abnormalities on speech, which significantly limits functional communication. Clinical speech researchers have thus spent decades investigating speech features in populations suffering from NMDs. Features of impaired articulatory function are of particular interest given their detrimental impact on intelligibility, their ability to encode a variety of distinct movement disorders, and their potential as diagnostic indicators of neurodegenerative diseases. The objectives of this scoping review were to identify (1) which components of articulation (i.e. coordination, consistency, speed, precision, and repetition rate) are the most represented in the acoustic literature on NMDs; (2) which acoustic articulatory features demonstrate the most potential for detecting speech motor dysfunction in NMDs; and (3) which articulatory components are the most impaired within each NMD. METHOD This review examined literature published between 1976 and 2020. Studies were identified from six electronic databases using predefined key search terms. The first research objective was addressed using a frequency count of studies investigating each articulatory component, while the second and third objectives were addressed using meta-analyses. RESULT Findings from 126 studies revealed a considerable emphasis on articulatory precision. Of the 24 features included in the meta-analyses, vowel dispersion/distance and stop gap duration exhibited the largest effects when comparing the NMD population to controls. The meta-analyses also revealed divergent patterns of articulatory performance across disease types, providing evidence of unique profiles of articulatory impairment. CONCLUSION This review illustrates the current state of the literature on acoustic articulatory features in NMDs. By highlighting the areas of need within each articulatory component and disease group, this work provides a foundation on which clinical researchers, speech scientists, neurologists, and computer science engineers can develop research questions that will both broaden and deepen the understanding of articulatory impairments in NMDs.
Collapse
Affiliation(s)
- Hannah P Rowe
- MGH Institute of Health Professions, Boston, MA, USA
| | - Sanjana Shellikeri
- Department of Speech-Language Pathology & Rehabilitation Sciences Institute, University of Toronto, Toronto, ON, Canada
- Sunnybrook Research Institute, Toronto, ON, Canada
- Penn Frontotemporal Degeneration Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Yana Yunusova
- Department of Speech-Language Pathology & Rehabilitation Sciences Institute, University of Toronto, Toronto, ON, Canada
- Sunnybrook Research Institute, Toronto, ON, Canada
| | - Karen V Chenausky
- MGH Institute of Health Professions, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA, and
| | - Jordan R Green
- MGH Institute of Health Professions, Boston, MA, USA
- Speech and Hearing Biosciences and Technology Program, Harvard University, Cambridge, MA, USA
| |
Collapse
|
31
|
Pattee GL, Genge A, Couratier P, Lunetta C, Sobue G, Aoki M, Yoshino H, Jackson CE, Wymer J, Salah A, Nelson S. Oral Edaravone - Introducing a Flexible Treatment Option for Amyotrophic Lateral Sclerosis. Expert Rev Neurother 2023; 23:859-866. [PMID: 37646130 DOI: 10.1080/14737175.2023.2251687] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 08/14/2023] [Accepted: 08/21/2023] [Indexed: 09/01/2023]
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) is a progressive and incurable neurodegenerative disease. While pharmacotherapy options remain limited, the Food and Drug Administration (FDA) approved intravenous (IV) and oral edaravone for the treatment of ALS in 2017 and 2022, respectively. With the addition of oral edaravone, patients with ALS may exclusively use oral medications. AREAS COVERED The authors performed a review of the published literature using the United States (US) National Library of Medicine's PubMed.gov resource to describe the pharmacokinetics, pharmacodynamics, safety, and efficacy of oral edaravone, as well as pertinent completed and ongoing clinical trials, including the oral edaravone clinical trial development program. The clinical profile of oral edaravone is also discussed. EXPERT OPINION Edaravone has been shown to slow the rate of motor function deterioration experienced by patients with ALS. As the oral formulation has been approved, patients with ALS may use it alone or in combination with other approved therapeutics. Additional clinical trials and real-world evidence are ongoing to gain further understanding of the clinical profile of oral edaravone.
Collapse
Affiliation(s)
- Gary L Pattee
- Neurological Sciences Department, University of Nebraska Medical Center/Neurology Associates, Lincoln, Nebraska, USA
| | - Angela Genge
- Clinical Research and ALS Clinic, Montreal Neurological Institute and Hospital, Montreal, Canada
| | - Philippe Couratier
- Department of Neurology, University Hospital of Limoges, Limoges, France
| | - Christian Lunetta
- ALS Medical Rehabilitation Department, Istituti Clinici Scientifici Maugeri IRCCS, Neurorehabilitation Unit of Milano, Milan, Italy
| | - Gen Sobue
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Neurology, Aichi Medical University, Nagakute, Aichi, Japan
| | - Masashi Aoki
- Department of Neurology, Tohoku University School of Medicine, Sendai, Miyagi, Japan
| | - Hiide Yoshino
- Department of Neurology, Yoshino Neurology Clinic, Chiba, Japan
| | - Carlayne E Jackson
- Department of Neurology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - James Wymer
- Department of Neuromuscular Neurology, University of Florida, Gainesville, Florida, USA
| | | | - Sally Nelson
- Mitsubishi Tanabe Pharma America, Inc, New Jersey, USA
| |
Collapse
|
32
|
Yamashita S, Tawara N, Hara K, Ueda M. Gender differences in clinical features at the initial examination of late-onset amyotrophic lateral sclerosis. J Neurol Sci 2023; 451:120697. [PMID: 37295193 DOI: 10.1016/j.jns.2023.120697] [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: 04/12/2023] [Revised: 05/26/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease that mainly affects motor neurons in the brain and spinal cord. With the advent of aging societies, the proportion of elderly patients with ALS is expected to increase. METHODS We retrospectively compared the clinical characteristics at the initial examination of patients with onset of ALS at age 74 years or younger (early onset) and those aged 75 years or older at onset (late-onset) at a single regional ALS diagnostic center in Japan. RESULTS The phenotype of late-onset ALS differed between males and females, with late-onset females having more bulbar-onset ALS and significantly lower body mass index, late-onset males having more frequent bulbar and respiratory symptoms at the initial examination, and significantly lower forced vital capacity at the initial examination in both groups compared to early onset patients. CONCLUSION For late-onset patients, maintenance of skeletal muscle mass by early intervention for bulbar and respiratory symptoms may be useful for prolonging survival; however, a prospective analysis is warranted.
Collapse
Affiliation(s)
- Satoshi Yamashita
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan; Department of Neurology, International University of Health and Welfare Narita Hospital, Narita, Japan.
| | - Nozomu Tawara
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kentaro Hara
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Mitsuharu Ueda
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
33
|
Mata S, Bussotti M, Del Mastio M, Barilaro A, Piersanti P, Lombardi M, Cincotta M, Torricelli S, Leccese D, Sperti M, Rodolico GR, Nacmias B, Sorbi S. Epidemiology of amyotrophic lateral sclerosis in the north east Tuscany in the 2018–2021 period. eNeurologicalSci 2023; 31:100457. [PMID: 37008534 PMCID: PMC10063398 DOI: 10.1016/j.ensci.2023.100457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/14/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Background The incidence of Amyotrophic Lateral Sclerosis (ALS) varies among different geographical areas and seems to increase over time. This study aimed to examine the epidemiologic data of ALS in the north-east Tuscany and compare the results with those of similar surveys. Methods Data from ALS cases diagnosed in Florence and Prato Hospitals were prospectively collected from 1st June 2018 to 31st May 2021. Results The age- and sex-adjusted incidence rate of ALS in cases per 100,000 population was 2.71 (M/F ratio: 1.21), significantly higher as compared to that reported in the 1967-1976 decade in the same geographical area (0.714). The age- and sex-adjusted incidence rate among resident strangers was similar to that of the general population (2.69). A slightly higher incidence rate (4.36) was observed in the north-east area of Florence province, which includes the Mugello valley. The mean prevalence was of 7.17/100,00. The mean age at diagnosis was 69.7 years, with a peak between 70 and 79 years among men and a smoother age curve among women. Conclusions ALS epidemiological features in north-east Tuscany are in line with other Italian and European Centers. The dramatic increase of the local disease burden over the last decades probably reflects better ascertainment methods and health system.
Collapse
|
34
|
Workman MJ, Lim RG, Wu J, Frank A, Ornelas L, Panther L, Galvez E, Perez D, Meepe I, Lei S, Valencia V, Gomez E, Liu C, Moran R, Pinedo L, Tsitkov S, Ho R, Kaye JA, Thompson T, Rothstein JD, Finkbeiner S, Fraenkel E, Sareen D, Thompson LM, Svendsen CN. Large-scale differentiation of iPSC-derived motor neurons from ALS and control subjects. Neuron 2023; 111:1191-1204.e5. [PMID: 36764301 PMCID: PMC10557526 DOI: 10.1016/j.neuron.2023.01.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/29/2022] [Accepted: 01/17/2023] [Indexed: 02/11/2023]
Abstract
Using induced pluripotent stem cells (iPSCs) to understand the mechanisms of neurological disease holds great promise; however, there is a lack of well-curated lines from a large array of participants. Answer ALS has generated over 1,000 iPSC lines from control and amyotrophic lateral sclerosis (ALS) patients along with clinical and whole-genome sequencing data. The current report summarizes cell marker and gene expression in motor neuron cultures derived from 92 healthy control and 341 ALS participants using a 32-day differentiation protocol. This is the largest set of iPSCs to be differentiated into motor neurons, and characterization suggests that cell composition and sex are significant sources of variability that need to be carefully controlled for in future studies. These data are reported as a resource for the scientific community that will utilize Answer ALS data for disease modeling using a wider array of omics being made available for these samples.
Collapse
Affiliation(s)
- Michael J Workman
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ryan G Lim
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Jie Wu
- Department of Biological Chemistry, University of California, Irvine, CA, USA
| | - Aaron Frank
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Loren Ornelas
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Lindsay Panther
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Erick Galvez
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Daniel Perez
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Imara Meepe
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Susan Lei
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Viviana Valencia
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Emilda Gomez
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Chunyan Liu
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ruby Moran
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Louis Pinedo
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Stanislav Tsitkov
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ritchie Ho
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Center for Neural Science and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Julia A Kaye
- Center for Systems and Therapeutics, Gladstone Institutes, University of California, San Francisco, San Francisco, CA, USA; Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes, University of California, San Francisco, San Francisco, CA, USA; Departments of Neurology and Physiology, University of California, San Francisco, San Francisco, CA, USA
| | | | - Jeffrey D Rothstein
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Steven Finkbeiner
- Center for Systems and Therapeutics, Gladstone Institutes, University of California, San Francisco, San Francisco, CA, USA; Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes, University of California, San Francisco, San Francisco, CA, USA; Departments of Neurology and Physiology, University of California, San Francisco, San Francisco, CA, USA
| | - Ernest Fraenkel
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Dhruv Sareen
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Leslie M Thompson
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA; Department of Biological Chemistry, University of California, Irvine, CA, USA; Department of Neurobiology and Behavior, University of California, Irvine, CA, USA; Department of Psychiatry and Human Behavior and Sue and Bill Gross Stem Cell Center, University of California, Irvine, CA, USA.
| | - Clive N Svendsen
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
35
|
Abu-Abaa M, Mousa A, Chadalawada S, Abdulsahib A. Bulbar Onset Amyotrophic Lateral Sclerosis in a COVID-19 Patient: A Case Report. Cureus 2023; 15:e37814. [PMID: 37214058 PMCID: PMC10197023 DOI: 10.7759/cureus.37814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2023] [Indexed: 05/23/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disorder with a largely unknown etiology. In this case, we are presenting an 84-year-old male patient who was admitted for acute hypoxemic respiratory failure secondary to coronavirus disease 2019 (COVID-19) infection. He was neurologically intact. His infection improved and oxygen requirement was gradually weaned off allowing for discharge. However, he was admitted again a month later with progressive dysphagia and aspiration that were confirmed on videofluoroscopic study. He was also found to have mild dysarthria, bulbar muscle weakness, bilateral lower motor neuron facial nerve palsy, diffuse hyporeflexia on four extremities with intact sensory function. Diagnosis of ALS was suspected after extensive workup was pursued and ruled out nutritional, structural, autoimmune, infectious and inflammatory disorders. This case is only the third reported case in medical literature to suggest COVID-19 infection as a triggering/accelerating factor of ALS progression.
Collapse
Affiliation(s)
- Mohammad Abu-Abaa
- Internal Medicine, Capital Health Regional Medical Center, Trenton, USA
| | - Aliaa Mousa
- Internal Medicine, Capital Health Regional Medical Center, Trenton, USA
| | | | - Ali Abdulsahib
- Internal Medicine, Capital Health Regional Medical Center, Trenton, USA
| |
Collapse
|
36
|
Bak AN, Djukic S, Kadlecova M, Braunstein TH, Jensen DB, Meehan CF. Cytoplasmic TDP-43 accumulation drives changes in C-bouton number and size in a mouse model of sporadic Amyotrophic Lateral Sclerosis. Mol Cell Neurosci 2023; 125:103840. [PMID: 36921783 DOI: 10.1016/j.mcn.2023.103840] [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: 10/15/2022] [Revised: 02/11/2023] [Accepted: 03/04/2023] [Indexed: 03/14/2023] Open
Abstract
An altered neuronal excitability of spinal motoneurones has consistently been implicated in Amyotrophic Lateral Sclerosis (ALS) leading to several investigations of synaptic input to these motoneurones. One such input that has repeatedly been shown to be affected is a population of large cholinergic synapses terminating mainly on the soma of the motoneurones referred to as C-boutons. Most research on these synapses during disease progression has used transgenic Superoxide Dismutase 1 (SOD1) mouse models of the disease which have not only produced conflicting findings, but also fail to recapitulate the key pathological feature seen in ALS; cytoplasmic accumulations of TAR DNA-binding protein 43 (TDP-43). Additionally, they fail to distinguish between slow and fast motoneurones, the latter of which have more C-boutons, but are lost earlier in the disease. To circumvent these issues, we quantified the frequency and volume of C-boutons on traced soleus and gastrocnemius motoneurones, representing predominantly slow and fast motor pools respectively. Experiments were performed using the TDP-43ΔNLS mouse model that carries a transgenic construct of TDP-43 devoid of its nuclear localization signal, preventing its nuclear import. This results in the emergence of pathological TDP-43 inclusions in the cytoplasm, modelling the main pathology seen in this disorder, accompanied by a severe and lethal ALS phenotype. Our results confirmed changes in both the number and volume of C-boutons with a decrease in number on the more vulnerable, predominantly fast gastrocnemius motoneurones and an increase in number on the less vulnerable, predominantly slow soleus motoneurones. Importantly, these changes were only found in male mice. However, both sexes and motor pools showed a decrease in C-bouton volume. Our experiments confirm that cytoplasmic TDP-43 accumulation is sufficient to drive C-bouton changes.
Collapse
Affiliation(s)
| | - Svetlana Djukic
- Department of Neuroscience, University of Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
37
|
de los Ríos C, Viejo L, Carretero VJ, Juárez NH, Cruz-Martins N, Hernández-Guijo JM. Promising Molecular Targets in Pharmacological Therapy for Neuronal Damage in Brain Injury. Antioxidants (Basel) 2023; 12:118. [PMID: 36670980 PMCID: PMC9854812 DOI: 10.3390/antiox12010118] [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: 11/24/2022] [Revised: 12/19/2022] [Accepted: 12/27/2022] [Indexed: 01/05/2023] Open
Abstract
The complex etiopathogenesis of brain injury associated with neurodegeneration has sparked a lot of studies in the last century. These clinical situations are incurable, and the currently available therapies merely act on symptoms or slow down the course of the diseases. Effective methods are being sought with an intent to modify the disease, directly acting on the properly studied targets, as well as to contribute to the development of effective therapeutic strategies, opening the possibility of refocusing on drug development for disease management. In this sense, this review discusses the available evidence for mitochondrial dysfunction induced by Ca2+ miscommunication in neurons, as well as how targeting phosphorylation events may be used to modulate protein phosphatase 2A (PP2A) activity in the treatment of neuronal damage. Ca2+ tends to be the catalyst for mitochondrial dysfunction, contributing to the synaptic deficiency seen in brain injury. Additionally, emerging data have shown that PP2A-activating drugs (PADs) suppress inflammatory responses by inhibiting different signaling pathways, indicating that PADs may be beneficial for the management of neuronal damage. In addition, a few bioactive compounds have also triggered the activation of PP2A-targeted drugs for this treatment, and clinical studies will help in the authentication of these compounds. If the safety profiles of PADs are proven to be satisfactory, there is a case to be made for starting clinical studies in the setting of neurological diseases as quickly as possible.
Collapse
Affiliation(s)
- Cristóbal de los Ríos
- Department of Pharmacology and Therapeutic and Teófilo Hernando Institute, Faculty of Medicine, University Autónoma de Madrid, C/. Arzobispo Morcillo 4, 28029 Madrid, Spain
- Departamento de Ciencias Básicas de la Salud, University Rey Juan Carlos, Avda. Atenas s/n, 28922 Alcorcón, Spain
| | - Lucía Viejo
- Department of Pharmacology and Therapeutic and Teófilo Hernando Institute, Faculty of Medicine, University Autónoma de Madrid, C/. Arzobispo Morcillo 4, 28029 Madrid, Spain
| | - Victoria Jiménez Carretero
- Department of Pharmacology and Therapeutic and Teófilo Hernando Institute, Faculty of Medicine, University Autónoma de Madrid, C/. Arzobispo Morcillo 4, 28029 Madrid, Spain
| | - Natalia Hernández Juárez
- Department of Pharmacology and Therapeutic and Teófilo Hernando Institute, Faculty of Medicine, University Autónoma de Madrid, C/. Arzobispo Morcillo 4, 28029 Madrid, Spain
| | - Natália Cruz-Martins
- Faculty of Medicine, Institute for Research and Innovation in Health (i3S), University of Porto, 4200-319 Porto, Portugal
- Institute for Research and Advanced Training in Health Sciences and Technologies, Rua Central de Gandra, 1317, 4585-116 Gandra, Portugal
| | - Jesús M. Hernández-Guijo
- Department of Pharmacology and Therapeutic and Teófilo Hernando Institute, Faculty of Medicine, University Autónoma de Madrid, C/. Arzobispo Morcillo 4, 28029 Madrid, Spain
- Ramón y Cajal Institute for Health Research, IRYCIS, Hospital Ramón y Cajal, Ctra. de Colmenar Viejo, Km. 9,100, 28029 Madrid, Spain
| |
Collapse
|
38
|
Nona RJ, Xu Z, Robinson GA, Henderson RD, McCombe PA. Age of Onset and Length of Survival of Queensland Patients with Amyotrophic Lateral Sclerosis: Details of Subjects with Early Onset and Subjects with Long Survival. NEURODEGENER DIS 2022; 22:104-121. [PMID: 36587610 PMCID: PMC10627495 DOI: 10.1159/000528875] [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: 06/16/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022] Open
Abstract
INTRODUCTION The aims of the study were to document the characteristics of amyotrophic lateral sclerosis (ALS) patients in Queensland, to examine factors influencing age of onset, and survival, and to study those with early-onset (<45 years) disease and those with long (>5 years) survival. METHODS We studied subjects seen at the ALS Clinic at the Royal Brisbane and Women's Hospital. We recorded sex, age of onset, region of onset, length of survival, presence of family history, type of disease, and evidence of cognitive involvement. We analysed the influence of these features on age of onset and survival. We analysed the features of patients with early onset of disease and patients with long survival. RESULTS There were 855 ALS patients (505 males) in the cohort. The age of onset was lower in males than females, in patients with a family history of ALS compared to those without, and in patients with spinal onset compared to bulbar onset. Early-onset disease was seen in 10% of patients, and had a greater proportion of males, spinal onset, and classical ALS phenotype compared to late-onset disease. Survival was shorter in females, in patients with bulbar onset, and in patients with classical ALS. Long survival was seen in 18% of patients. Patients with long survival had younger age of onset, greater proportion of males, spinal onset, and fewer patients with classical ALS. CONCLUSION Our study confirms that ALS is more prevalent in males and that spinal onset is more common than bulbar onset. Males have earlier onset but longer survival. We found that overall, patients with classical ALS have worse survival than ALS variants, but some patients who were considered to have classical ALS had long survival. This study confirms the similarity of ALS in our region to ALS in other geographical regions.
Collapse
Affiliation(s)
- Robert J. Nona
- Centre for Clinical Research, University of Queensland, Herston, Queensland, Australia
| | - Zhouwei Xu
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Gail A. Robinson
- Queensland Brain Institute and School of Psychology, University of Queensland, St Lucia, Queensland, Australia
| | - Robert D. Henderson
- Centre for Clinical Research, University of Queensland, Herston, Queensland, Australia
- Department of Neurology, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia
| | - Pamela A. McCombe
- Centre for Clinical Research, University of Queensland, Herston, Queensland, Australia
- Department of Neurology, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia
| |
Collapse
|
39
|
Zalewska T, Pawelec P, Ziabska K, Ziemka-Nalecz M. Sexual Dimorphism in Neurodegenerative Diseases and in Brain Ischemia. Biomolecules 2022; 13:26. [PMID: 36671411 PMCID: PMC9855831 DOI: 10.3390/biom13010026] [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: 11/22/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022] Open
Abstract
Epidemiological studies and clinical observations show evidence of sexual dimorphism in brain responses to several neurological conditions. It is suggested that sex-related differences between men and women may have profound effects on disease susceptibility, pathophysiology, and progression. Sexual differences of the brain are achieved through the complex interplay of several factors contributing to this phenomenon, such as sex hormones, as well as genetic and epigenetic differences. Despite recent advances, the precise link between these factors and brain disorders is incompletely understood. This review aims to briefly outline the most relevant aspects that differ between men and women in ischemia and neurodegenerative disorders (AD, PD, HD, ALS, and SM). Recognition of disparities between both sexes could aid the development of individual approaches to ameliorate or slow the progression of intractable disorders.
Collapse
Affiliation(s)
- Teresa Zalewska
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 A. Pawinskiego Str., 02-106 Warsaw, Poland
| | | | | | | |
Collapse
|
40
|
Metzner K, Darawsha O, Wang M, Gaur N, Cheng Y, Rödiger A, Frahm C, Witte OW, Perocchi F, Axer H, Grosskreutz J, Brill MS. Age-dependent increase of cytoskeletal components in sensory axons in human skin. Front Cell Dev Biol 2022; 10:965382. [PMID: 36393849 PMCID: PMC9664158 DOI: 10.3389/fcell.2022.965382] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 10/12/2022] [Indexed: 01/24/2023] Open
Abstract
Aging is a complex process characterized by several molecular and cellular imbalances. The composition and stability of the neuronal cytoskeleton is essential for the maintenance of homeostasis, especially in long neurites. Using human skin biopsies containing sensory axons from a cohort of healthy individuals, we investigate alterations in cytoskeletal content and sensory axon caliber during aging via quantitative immunostainings. Cytoskeletal components show an increase with aging in both sexes, while elevation in axon diameter is only evident in males. Transcriptomic data from aging males illustrate various patterns in gene expression during aging. Together, the data suggest gender-specific changes during aging in peripheral sensory axons, possibly influencing cytoskeletal functionality and axonal caliber. These changes may cumulatively increase susceptibility of aged individuals to neurodegenerative diseases.
Collapse
Affiliation(s)
- Klara Metzner
- Department of Neurology, Jena University Hospital, Jena, Germany,Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - Omar Darawsha
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - Mengzhe Wang
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - Nayana Gaur
- Department of Neurology, Jena University Hospital, Jena, Germany,Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Yiming Cheng
- Helmholtz Diabetes Center (HDC), Helmholtz Center Munich, Institute for Diabetes and Obesity, Munich, Germany
| | | | - Christiane Frahm
- Department of Neurology, Jena University Hospital, Jena, Germany
| | - Otto W. Witte
- Department of Neurology, Jena University Hospital, Jena, Germany
| | - Fabiana Perocchi
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany,Helmholtz Diabetes Center (HDC), Helmholtz Center Munich, Institute for Diabetes and Obesity, Munich, Germany,Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Hubertus Axer
- Department of Neurology, Jena University Hospital, Jena, Germany
| | - Julian Grosskreutz
- Precision Neurology of the University of Lübeck, Lübeck, Germany,PMI Cluster, University of Lübeck, Lübeck, Germany
| | - Monika S. Brill
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany,Munich Cluster of Systems Neurology (SyNergy), Munich, Germany,*Correspondence: Monika S. Brill,
| |
Collapse
|
41
|
Parrella E, Porrini V, Scambi I, Gennari MM, Gussago C, Bankole O, Benarese M, Mariotti R, Pizzi M. Synergistic association of resveratrol and histone deacetylase inhibitors as treatment in amyotrophic lateral sclerosis. Front Pharmacol 2022; 13:1017364. [PMID: 36339574 PMCID: PMC9633661 DOI: 10.3389/fphar.2022.1017364] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 10/06/2022] [Indexed: 11/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease associated with motor neuron degeneration, progressive paralysis and finally death. Despite the research efforts, currently there is no cure for ALS. In recent years, multiple epigenetic mechanisms have been associated with neurodegenerative diseases. A pathological role for histone hypoacetylation and the abnormal NF-κB/RelA activation involving deacetylation of lysines, with the exclusion of lysine 310, has been established in ALS. Recent findings indicate that the pathological acetylation state of NF-κB/RelA and histone 3 (H3) occurring in the SOD1(G93A) murine model of ALS can be corrected by the synergistic combination of low doses of the AMP-activated kinase (AMPK)-sirtuin 1 pathway activator resveratrol and the histone deacetylase (HDAC) inhibitors MS-275 (entinostat) or valproate. The combination of the epigenetic drugs, by rescuing RelA and the H3 acetylation state, promotes a beneficial and sexually dimorphic effect on disease onset, survival and motor neurons degeneration. In this mini review, we discuss the potential of the epigenetic combination of resveratrol with HDAC inhibitors in the ALS treatment.
Collapse
Affiliation(s)
- Edoardo Parrella
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Vanessa Porrini
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Ilaria Scambi
- Section of Anatomy and Histology, Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Michele M. Gennari
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Cristina Gussago
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Oluwamolakun Bankole
- Section of Anatomy and Histology, Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Marina Benarese
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Raffaella Mariotti
- Section of Anatomy and Histology, Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Marina Pizzi
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
42
|
Poppe C, Verwey M, Wangmo T. "Walking a tightrope": A grounded theory approach to informal caregiving for amyotrophic lateral sclerosis. HEALTH & SOCIAL CARE IN THE COMMUNITY 2022; 30:e1935-e1947. [PMID: 34719073 PMCID: PMC9545073 DOI: 10.1111/hsc.13625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 10/06/2021] [Accepted: 10/18/2021] [Indexed: 06/13/2023]
Abstract
Informal caregivers, mainly family members and friends, provide supportive and palliative care for people with amyotrophic lateral sclerosis (ALS) during their terminal disease course. Informal caregiving for people with ALS continues towards palliative care and end-of-life care with the progression of the disease. In this study, we provide a theoretical understanding of informal caregiving in ALS utilising 23 semi-structured interviews conducted with informal caregivers of people with ALS (pwALS) in Switzerland. Due to the expected death of the care recipient, our grounded theory approach outlines informal caregivers' caregiving work as an effort to secure a balance amongst different caregiving activities, which feed into the final stage of providing palliative care. Overall, our theoretical understanding of ALS informal caregiving work encompasses the core category 'holding the balance' and four secondary categories: 'Organising support', 'being present', 'managing everyday life' and 'keeping up with ALS'. The core category of holding the balance underlines the significance of ensuring care and normalcy even as disease progresses and until the end of life. For the informal caregivers, this balancing act is the key element of care provision to pwALS and therefore guides decisions surrounding caregiving. On this understanding, those caregivers that succeed in holding the balance can provide care at home until death. The balance is heavily influenced by contextual factors of caregiving, for example relating to personal characteristics of the caregiver, or activities of caregiving where the goal is to ensure the quality of life of the pwALS. As there is a heterogeneity of speed and subtype of progression of ALS, our work accounts for multiple caregiving trajectories.
Collapse
Affiliation(s)
| | - Martine Verwey
- Patient Association ALS Patients ConnectedBilthovenThe Netherlands
| | - Tenzin Wangmo
- Institute for Biomedical EthicsUniversity of BaselBaselSwitzerland
| |
Collapse
|
43
|
Proaño B, Casani-Cubel J, Benlloch M, Rodriguez-Mateos A, Navarro-Illana E, Lajara-Romance JM, de la Rubia Ortí JE. Is Dutasteride a Therapeutic Alternative for Amyotrophic Lateral Sclerosis? Biomedicines 2022; 10:biomedicines10092084. [PMID: 36140184 PMCID: PMC9495995 DOI: 10.3390/biomedicines10092084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/17/2022] [Accepted: 08/23/2022] [Indexed: 11/24/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease that is characterized by the loss of upper and lower motor neurons (MNs) in the cerebral cortex, brainstem and spinal cord, with consequent weakness, atrophy and the progressive paralysis of all muscles. There is currently no medical cure, and riluzole and edaravone are the only two known approved drugs for treating this condition. However, they have limited efficacy, and hence there is a need to find new molecules. Dutasteride, a dual inhibitor of type 1 and type 2 5α-reductase (5AR) enzymes, the therapeutic purposes of which, to date, are the treatment of benign prostatic hyperplasia and androgenic alopecia, shows great anti-ALS properties by the molecular-topology methodology. Based on this evidence, this review aims to assess the effects of dutasteride on testosterone (T), progesterone (PROG) and 17β-estradiol (17BE) as a therapeutic alternative for the clinical improvement of ALS, based on the hormonal, metabolic and molecular pathways related to the pathogenesis of the disease. According to the evidence found, dutasteride shows great neuroprotective, antioxidant and anti-inflammatory effects. It also appears effective against glutamate toxicity, and it is capable of restoring altered dopamine activity (DA). These effects are achieved both directly and through steroid hormones. Therefore, dutasteride seems to be a promising molecule for the treatment of ALS, although clinical studies are required for confirmation.
Collapse
Affiliation(s)
- Belén Proaño
- Doctoral Degree School, Health Sciences, Catholic University of Valencia San Vicente Mártir, 46001 Valencia, Spain
| | - Julia Casani-Cubel
- School of Medicine and Health Sciences, Catholic University San Vicente Mártir, 46001 Valencia, Spain
- Correspondence: (J.C.-C.); (M.B.)
| | - María Benlloch
- Department Nursing, Catholic University San Vicente Mártir, 46001 Valencia, Spain
- Correspondence: (J.C.-C.); (M.B.)
| | - Ana Rodriguez-Mateos
- Department of Nutritional Sciences, King’s College London, Franklin Wilkins Building, London SE1 9NH, UK
| | | | | | | |
Collapse
|
44
|
Trentini A, Manfrinato MC, Castellazzi M, Bellini T. Sex-Related Differences of Matrix Metalloproteinases (MMPs): New Perspectives for These Biomarkers in Cardiovascular and Neurological Diseases. J Pers Med 2022; 12:jpm12081196. [PMID: 35893290 PMCID: PMC9331234 DOI: 10.3390/jpm12081196] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 11/16/2022] Open
Abstract
It is now established that sex differences occur in clinical manifestation, disease progression, and prognosis for both cardiovascular (CVDs) and central nervous system (CNS) disorders. As such, a great deal of effort is now being put into understanding these differences and turning them into “advantages”: (a) for the discovery of new sex-specific biomarkers and (b) through a review of old biomarkers from the perspective of the “newly” discovered sex/gender medicine. This is also true for matrix metalloproteinases (MMPs), enzymes involved in extracellular matrix (ECM) remodelling, which play a role in both CVDs and CNS disorders. However, most of the studies conducted up to now relegated sex to a mere confounding variable used for statistical model correction rather than a determining factor that can influence MMP levels and, in turn, disease prognosis. Consistently, this approach causes a loss of information that might help clinicians in identifying novel patterns and improve the applicability of MMPs in clinical practice by providing sex-specific threshold values. In this scenario, the current review aims to gather the available knowledge on sex-related differences in MMPs levels in CVDs and CNS conditions, hoping to shed light on their use as sex-specific biomarkers of disease prognosis or progression.
Collapse
Affiliation(s)
- Alessandro Trentini
- Department of Environmental and Prevention Sciences, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy;
- University Center for Studies on Gender Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Maria Cristina Manfrinato
- University Center for Studies on Gender Medicine, University of Ferrara, 44121 Ferrara, Italy
- Department of Neuroscience and Rehabilitation, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy; (M.C.M.); (T.B.)
| | - Massimiliano Castellazzi
- University Center for Studies on Gender Medicine, University of Ferrara, 44121 Ferrara, Italy
- Department of Neuroscience and Rehabilitation, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy; (M.C.M.); (T.B.)
- Interdepartmental Research Center for the Study of Multiple Sclerosis and Inflammatory and Degenerative Diseases of the Nervous System, University of Ferrara, 44121 Ferrara, Italy
- Correspondence:
| | - Tiziana Bellini
- University Center for Studies on Gender Medicine, University of Ferrara, 44121 Ferrara, Italy
- Department of Neuroscience and Rehabilitation, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy; (M.C.M.); (T.B.)
| |
Collapse
|
45
|
Shen H, Yanas A, Owens MC, Zhang C, Fritsch C, Fare CM, Copley KE, Shorter J, Goldman YE, Liu KF. Sexually dimorphic RNA helicases DDX3X and DDX3Y differentially regulate RNA metabolism through phase separation. Mol Cell 2022; 82:2588-2603.e9. [PMID: 35588748 PMCID: PMC9308757 DOI: 10.1016/j.molcel.2022.04.022] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 04/09/2022] [Accepted: 04/19/2022] [Indexed: 02/05/2023]
Abstract
Sex differences are pervasive in human health and disease. One major key to sex-biased differences lies in the sex chromosomes. Although the functions of the X chromosome proteins are well appreciated, how they compare with their Y chromosome homologs remains elusive. Herein, using ensemble and single-molecule techniques, we report that the sex chromosome-encoded RNA helicases DDX3X and DDX3Y are distinct in their propensities for liquid-liquid phase separation (LLPS), dissolution, and translation repression. We demonstrate that the N-terminal intrinsically disordered region of DDX3Y more strongly promotes LLPS than the corresponding region of DDX3X and that the weaker ATPase activity of DDX3Y, compared with DDX3X, contributes to the slower disassembly dynamics of DDX3Y-positive condensates. Interestingly, DDX3Y-dependent LLPS represses mRNA translation and enhances aggregation of FUS more strongly than DDX3X-dependent LLPS. Our study provides a platform for future comparisons of sex chromosome-encoded protein homologs, providing insights into sex differences in RNA metabolism and human disease.
Collapse
Affiliation(s)
- Hui Shen
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amber Yanas
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Graduate Group in Biochemistry and Molecular Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael C Owens
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Graduate Group in Biochemistry and Molecular Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Celia Zhang
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Clark Fritsch
- Graduate Group in Cellular and Molecular Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Charlotte M Fare
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Graduate Group in Biochemistry and Molecular Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Katie E Copley
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Graduate Group in Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - James Shorter
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Graduate Group in Biochemistry and Molecular Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Graduate Group in Cellular and Molecular Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Graduate Group in Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yale E Goldman
- Graduate Group in Biochemistry and Molecular Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kathy Fange Liu
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Graduate Group in Biochemistry and Molecular Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
46
|
Li W, Pandya D, Pasternack N, Garcia-Montojo M, Henderson L, Kozak CA, Nath A. Retroviral Elements in Pathophysiology and as Therapeutic Targets for Amyotrophic Lateral Sclerosis. Neurotherapeutics 2022; 19:1085-1101. [PMID: 35415778 PMCID: PMC9587200 DOI: 10.1007/s13311-022-01233-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2022] [Indexed: 10/18/2022] Open
Abstract
The study of the role of retroviruses in amyotrophic lateral sclerosis (ALS) dates back to the 1960s shortly after transposable elements themselves were first discovered. It was quickly realized that in wild mice both horizontal and vertical transmissions of retroviral elements were key to the development of an ALS-like syndrome leading to the postulate that endogenous retroviruses (ERVs) contribute significantly to the pathogenicity of this disease. Subsequent studies identified retroviral reverse transcriptase activity in brains of individuals with ALS from Guam. However, except for a single study from the former Soviet Union, ALS could not be transmitted to rhesus macaques. The discovery of an ALS-like syndrome in human immunodeficiency virus (HIV) and human T cell leukemia virus infected individuals led to renewed interest in the field and reverse transcriptase activity was found in the blood and cerebrospinal fluid of individuals with sporadic ALS. However, exogenous retroviruses could not be found in individuals with ALS which further reinforced the possibility of involvement of a human ERV (HERV). The first demonstration of the involvement of a HERV was the discovery of the activation of human endogenous retrovirus-K subtype HML-2 in the brains of individuals with ALS. The envelope protein of HML-2 is neurotoxic and transgenic animals expressing the envelope protein develop an ALS-like syndrome. Activation of HML-2 occurs in the context of generalized transposable element activation and is not specific for ALS. Individuals with HIV-associated ALS show a remarkable response to antiretroviral therapy; however, antiretroviral trials in ALS down-regulate HML-2 without ameliorating the disease. This highlights the need for specific drugs to be developed against HML-2 as a novel therapeutic target for ALS. Other approaches might include antisense oligonucleotides, shRNA targeted against the envelope gene or antibodies that can target the extracellular envelope protein. Future clinical trials in ALS should consider combination therapies to control these ERVs.
Collapse
Affiliation(s)
- Wenxue Li
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Darshan Pandya
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Nicholas Pasternack
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Marta Garcia-Montojo
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Lisa Henderson
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Christine A Kozak
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Avindra Nath
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA.
| |
Collapse
|
47
|
Goutman SA, Hardiman O, Al-Chalabi A, Chió A, Savelieff MG, Kiernan MC, Feldman EL. Recent advances in the diagnosis and prognosis of amyotrophic lateral sclerosis. Lancet Neurol 2022; 21:480-493. [PMID: 35334233 PMCID: PMC9513753 DOI: 10.1016/s1474-4422(21)00465-8] [Citation(s) in RCA: 229] [Impact Index Per Article: 76.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/24/2021] [Accepted: 12/16/2021] [Indexed: 12/14/2022]
Abstract
The diagnosis of amyotrophic lateral sclerosis can be challenging due to its heterogeneity in clinical presentation and overlap with other neurological disorders. Diagnosis early in the disease course can improve outcomes as timely interventions can slow disease progression. An evolving awareness of disease genotypes and phenotypes and new diagnostic criteria, such as the recent Gold Coast criteria, could expedite diagnosis. Improved prognosis, such as that achieved with the survival model from the European Network for the Cure of ALS, could inform the patient and their family about disease course and improve end-of-life planning. Novel staging and scoring systems can help monitor disease progression and might potentially serve as clinical trial outcomes. Lastly, new tools, such as fluid biomarkers, imaging modalities, and neuromuscular electrophysiological measurements, might increase diagnostic and prognostic accuracy.
Collapse
Affiliation(s)
| | - Orla Hardiman
- Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Ammar Al-Chalabi
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, and Department of Neurology, King's College London, London, UK
| | - Adriano Chió
- Rita Levi Montalcini Department of Neurosciences, University of Turin, Turin, Italy
| | | | - Matthew C Kiernan
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia; Department of Neurology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
48
|
Re DB, Yan B, Calderón-Garcidueñas L, Andrew AS, Tischbein M, Stommel EW. A perspective on persistent toxicants in veterans and amyotrophic lateral sclerosis: identifying exposures determining higher ALS risk. J Neurol 2022; 269:2359-2377. [PMID: 34973105 PMCID: PMC9021134 DOI: 10.1007/s00415-021-10928-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 02/07/2023]
Abstract
Multiple studies indicate that United States veterans have an increased risk of developing amyotrophic lateral sclerosis (ALS) compared to civilians. However, the responsible etiological factors are unknown. In the general population, specific occupational (e.g. truck drivers, airline pilots) and environmental exposures (e.g. metals, pesticides) are associated with an increased ALS risk. As such, the increased prevalence of ALS in veterans strongly suggests that there are exposures experienced by military personnel that are disproportionate to civilians. During service, veterans may encounter numerous neurotoxic exposures (e.g. burn pits, engine exhaust, firing ranges). So far, however, there is a paucity of studies investigating environmental factors contributing to ALS in veterans and even fewer assessing their exposure using biomarkers. Herein, we discuss ALS pathogenesis in relation to a series of persistent neurotoxicants (often emitted as mixtures) including: chemical elements, nanoparticles and lipophilic toxicants such as dioxins, polycyclic aromatic hydrocarbons and polychlorinated biphenyls. We propose these toxicants should be directly measured in veteran central nervous system tissue, where they may have accumulated for decades. Specific toxicants (or mixtures thereof) may accelerate ALS development following a multistep hypothesis or act synergistically with other service-linked exposures (e.g. head trauma/concussions). Such possibilities could explain the lower age of onset observed in veterans compared to civilians. Identifying high-risk exposures within vulnerable populations is key to understanding ALS etiopathogenesis and is urgently needed to act upon modifiable risk factors for military personnel who deserve enhanced protection during their years of service, not only for their short-term, but also long-term health.
Collapse
Affiliation(s)
- Diane B Re
- Department of Environmental Health Science, Center for Motor Neuron Biology and Disease, Columbia University, New York, NY, USA
| | - Beizhan Yan
- Department of Geochemistry, Lamont-Doherty Earth Observatory of Columbia University, Palisades, NY, USA
| | - Lilian Calderón-Garcidueñas
- Department Biomedical Sciences, College of Health, University of Montana, Missoula, MT, USA
- Universidad del Valle de México, Mexico City, Mexico
| | - Angeline S Andrew
- Department of Neurology, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - Maeve Tischbein
- Department of Neurology, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - Elijah W Stommel
- Department of Neurology, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA.
| |
Collapse
|
49
|
Newell ME, Adhikari S, Halden RU. Systematic and state-of the science review of the role of environmental factors in Amyotrophic Lateral Sclerosis (ALS) or Lou Gehrig's Disease. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 817:152504. [PMID: 34971691 DOI: 10.1016/j.scitotenv.2021.152504] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 06/14/2023]
Abstract
The etiology of sporadic amyotrophic lateral sclerosis (ALS) is still unclear. We evaluate environmental factors suspected to be associated with ALS for their potential linkage to disease causality and to model geographic distributions of susceptible populations and expected cases worldwide. A PRISMA systematic literature review was performed 2021. Bradford Hill criteria were used to identify and rank environmental factors and a secondary review of ALS diagnoses in population studies and ALS case or cohort studies was conducted. Prevalence rate projection informed estimates of impacted regions and populations. Among 1710 papers identified, 258 met the inclusion criteria, of which 173 responded to at least one of nine Bradford Hill criteria among 83 literature-identified ALS environmental factors. Environmental determinants of ALS in order of decreasing significance were β-N-methylamino-L-alanine (BMAA), formaldehyde, selenium, and heavy metals including manganese, mercury, zinc, and copper. Murine animal models were the most common methodology for exploring environmental factors. Another line of investigation of 62 population exposure studies implicated the same group of environmental agents (mean odds ratios): BMAA (2.32), formaldehyde (1.54), heavy metals (2.99), manganese (3.85), mercury (2.74), and zinc (2.78). An age-adjusted incidence model estimated current total ALS cases globally at ~85,000 people compared to only ~1600 cases projected from the reported ALS incidence in the literature. Modeling with the prevalence microscope equation forecasted an increase in U.S. ALS cases from 16,707 confirmed in 2015 to ~22,650 projected for 2040. Two orthogonal methods employed implicate BMAA, formaldehyde, manganese, mercury, and zinc as environmental factors with strong ALS associations. ALS cases likely are significantly underreported globally, and high vulnerability exists in regions with large aging populations. Recent studies on other diseases with environmental determinants suggest the need to consider additional potential triggers and mechanisms, including exposures to microbial agents and epigenetic modifications.
Collapse
Affiliation(s)
- Melanie Engstrom Newell
- Biodesign Center for Environmental Health Engineering, Biodesign Institute, Building B, Arizona State University, 1001 S McAllister Ave, Tempe, AZ 85281-8101, USA; School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, AZ, USA.
| | - Sangeet Adhikari
- Biodesign Center for Environmental Health Engineering, Biodesign Institute, Building B, Arizona State University, 1001 S McAllister Ave, Tempe, AZ 85281-8101, USA; School of Sustainable Engineering and the Built Environment, Arizona State University, Tempe, AZ, USA.
| | - Rolf U Halden
- Biodesign Center for Environmental Health Engineering, Biodesign Institute, Building B, Arizona State University, 1001 S McAllister Ave, Tempe, AZ 85281-8101, USA; School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, AZ, USA; School of Sustainable Engineering and the Built Environment, Arizona State University, Tempe, AZ, USA; OneWaterOneHealth, Arizona State University Foundation, 1001 S. McAllister Avenue, Tempe, AZ 85287-8101, USA; Global Futures Laboratory, Arizona State University, 800 S. Cady Mall, Tempe, AZ 85281, USA.
| |
Collapse
|
50
|
Boddy S, Islam M, Moll T, Kurz J, Burrows D, McGown A, Bhargava A, Julian TH, Harvey C, Marshall JNG, Hall BPC, Allen SP, Kenna KP, Sanderson E, Zhang S, Ramesh T, Snyder MP, Shaw PJ, McDermott C, Cooper-Knock J. Unbiased metabolome screen leads to personalized medicine strategy for amyotrophic lateral sclerosis. Brain Commun 2022; 4:fcac069. [PMID: 35441136 PMCID: PMC9010771 DOI: 10.1093/braincomms/fcac069] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 11/29/2021] [Accepted: 03/15/2022] [Indexed: 11/17/2022] Open
Abstract
Amyotrophic lateral sclerosis is a rapidly progressive neurodegenerative disease that affects 1/350 individuals in the United Kingdom. The cause of amyotrophic lateral sclerosis is unknown in the majority of cases. Two-sample Mendelian randomization enables causal inference between an exposure, such as the serum concentration of a specific metabolite, and disease risk. We obtained genome-wide association study summary statistics for serum concentrations of 566 metabolites which were population matched with a genome-wide association study of amyotrophic lateral sclerosis. For each metabolite, we performed Mendelian randomization using an inverse variance weighted estimate for significance testing. After stringent Bonferroni multiple testing correction, our unbiased screen revealed three metabolites that were significantly linked to the risk of amyotrophic lateral sclerosis: Estrone-3-sulphate and bradykinin were protective, which is consistent with literature describing a male preponderance of amyotrophic lateral sclerosis and a preventive effect of angiotensin-converting enzyme inhibitors which inhibit the breakdown of bradykinin. Serum isoleucine was positively associated with amyotrophic lateral sclerosis risk. All three metabolites were supported by robust Mendelian randomization measures and sensitivity analyses; estrone-3-sulphate and isoleucine were confirmed in a validation amyotrophic lateral sclerosis genome-wide association study. Estrone-3-sulphate is metabolized to the more active estradiol by the enzyme 17β-hydroxysteroid dehydrogenase 1; further, Mendelian randomization demonstrated a protective effect of estradiol and rare variant analysis showed that missense variants within HSD17B1, the gene encoding 17β-hydroxysteroid dehydrogenase 1, modify risk for amyotrophic lateral sclerosis. Finally, in a zebrafish model of C9ORF72-amyotrophic lateral sclerosis, we present evidence that estradiol is neuroprotective. Isoleucine is metabolized via methylmalonyl-CoA mutase encoded by the gene MMUT in a reaction that consumes vitamin B12. Multivariable Mendelian randomization revealed that the toxic effect of isoleucine is dependent on the depletion of vitamin B12; consistent with this, rare variants which reduce the function of MMUT are protective against amyotrophic lateral sclerosis. We propose that amyotrophic lateral sclerosis patients and family members with high serum isoleucine levels should be offered supplementation with vitamin B12.
Collapse
Affiliation(s)
- Sarah Boddy
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Mahjabin Islam
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Tobias Moll
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Julian Kurz
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - David Burrows
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Alexander McGown
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Anushka Bhargava
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Thomas H Julian
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Calum Harvey
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Jack NG Marshall
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Benjamin PC Hall
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Scott P Allen
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Kevin P Kenna
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Eleanor Sanderson
- Medical Research Council (MRC) Integrative Epidemiology Unit (IEU), University of Bristol, Bristol, UK
| | - Sai Zhang
- Center for Genomics and Personalized Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Tennore Ramesh
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Michael P Snyder
- Center for Genomics and Personalized Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Christopher McDermott
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Johnathan Cooper-Knock
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|