1
|
Wu J, Ye S, Liu X, Xu Y, Fan D. The burden of upper motor neuron involvement is correlated with the bilateral limb involvement interval in patients with amyotrophic lateral sclerosis: a retrospective observational study. Neural Regen Res 2025; 20:1505-1512. [PMID: 39075916 PMCID: PMC11624872 DOI: 10.4103/nrr.nrr-d-23-01359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 12/19/2023] [Accepted: 01/12/2024] [Indexed: 07/31/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202505000-00032/figure1/v/2024-07-28T173839Z/r/image-tiff Amyotrophic lateral sclerosis is a rare neurodegenerative disease characterized by the involvement of both upper and lower motor neurons. Early bilateral limb involvement significantly affects patients' daily lives and may lead them to be confined to bed. However, the effect of upper and lower motor neuron impairment and other risk factors on bilateral limb involvement is unclear. To address this issue, we retrospectively collected data from 586 amyotrophic lateral sclerosis patients with limb onset diagnosed at Peking University Third Hospital between January 2020 and May 2022. A univariate analysis revealed no significant differences in the time intervals of spread in different directions between individuals with upper motor neuron-dominant amyotrophic lateral sclerosis and those with classic amyotrophic lateral sclerosis. We used causal directed acyclic graphs for risk factor determination and Cox proportional hazards models to investigate the association between the duration of bilateral limb involvement and clinical baseline characteristics in amyotrophic lateral sclerosis patients. Multiple factor analyses revealed that higher upper motor neuron scores (hazard ratio [HR] = 1.05, 95% confidence interval [CI] = 1.01-1.09, P = 0.018), onset in the left limb (HR = 0.72, 95% CI = 0.58-0.89, P = 0.002), and a horizontal pattern of progression (HR = 0.46, 95% CI = 0.37-0.58, P < 0.001) were risk factors for a shorter interval until bilateral limb involvement. The results demonstrated that a greater degree of upper motor neuron involvement might cause contralateral limb involvement to progress more quickly in limb-onset amyotrophic lateral sclerosis patients. These findings may improve the management of amyotrophic lateral sclerosis patients with limb onset and the prediction of patient prognosis.
Collapse
Affiliation(s)
- Jieying Wu
- Department of Neurology, Peking University Third Hospital, Beijing, China
- Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
| | - Shan Ye
- Department of Neurology, Peking University Third Hospital, Beijing, China
- Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
| | - Xiangyi Liu
- Department of Neurology, Peking University Third Hospital, Beijing, China
- Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
| | - Yingsheng Xu
- Department of Neurology, Peking University Third Hospital, Beijing, China
- Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
| | - Dongsheng Fan
- Department of Neurology, Peking University Third Hospital, Beijing, China
- Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
- Key Laboratory for Neuroscience, National Health Commission/Ministry of Education, Peking University, Beijing, China
| |
Collapse
|
2
|
Olsen CG, Malmberg VN, Fahlström M, Alstadhaug KB, Bjørnå IK, Braathen GJ, Bråthen G, Demic N, Hallerstig E, Hogenesch I, Horn MA, Kampman MT, Kleveland G, Ljøstad U, Maniaol A, Morsund ÅH, Nakken O, Schlüter K, Schuler S, Seim E, Flemmen HØ, Tysnes OB, Holmøy T, Høyer H. Amyotrophic lateral sclerosis caused by the C9orf72 expansion in Norway - prevalence, ancestry, clinical characteristics and sociodemographic status. Amyotroph Lateral Scler Frontotemporal Degener 2025; 26:132-140. [PMID: 39316038 DOI: 10.1080/21678421.2024.2405118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/03/2024] [Accepted: 09/12/2024] [Indexed: 09/25/2024]
Abstract
OBJECTIVE The most common genetic cause of amyotrophic lateral sclerosis (ALS) is the C9orf72 expansion. A high incidence of this expansion has been detected in Sweden and Finland. This Norwegian population-based study aimed to identify the prevalence, geographic distribution, ancestry, and relatedness of ALS patients with a C9orf72 expansion (C9pos). Further, we compared C9pos and C9neg patients' clinical presentation, family history of ALS and other neurodegenerative disorders, and sociodemographic status. METHODS We recruited ALS patients from all 17 Departments of neurology in Norway. Blood samples and questionnaires regarding clinical characteristics, sociodemographic status and family history of ALS, and other neurodegenerative disorders were collected. The C9orf72 expansion was examined for all patients. RESULTS The study enrolled 500 ALS patients, 8.8% of whom were C9pos, with half being sporadic ALS cases. The proportion of C9pos cases differed between regions, ranging from 17.9% in the Northern region to 1.9% in the Western region. The majority of C9pos patients had non-Finnish European descent and were not closely related. C9pos patients exhibited a significantly shorter mean survival time, had a higher frequency of relatives with ALS or dementia, and were more often unmarried/single and childless than C9neg patients. CONCLUSION C9pos patients constitute a large portion of the Norwegian ALS population. Ancestry and relatedness do not adequately explain regional differences. Relying on clinical information to identify C9pos patients has proven to be challenging. Half of C9pos patients were reported as having sporadic ALS, underlining the importance of carefully assessing family history and the need for genetic testing.
Collapse
Affiliation(s)
- Cathrine Goberg Olsen
- Department of Medical Genetics, Telemark Hospital Trust, Skien, Norway
- Institute of Clinical Medicine, University of Oslo, Nordbyhagen, Norway
| | - Vetle Nilsen Malmberg
- Institute of Clinical Medicine, University of Oslo, Nordbyhagen, Norway
- Department of Neurology, Telemark Hospital Trust, Skien, Norway
| | - Maria Fahlström
- Department of Medical Genetics, Telemark Hospital Trust, Skien, Norway
| | | | | | | | - Geir Bråthen
- Department of Neurology and Clinical Neurophysiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Department of Neuromedicine and Movement Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Natasha Demic
- Department of Neurology, Vestfold Hospital Trust, Tønsberg, Norway
| | | | - Ineke Hogenesch
- Department of Neurology, Fonna Hospital Trust, Haugesund, Norway
| | | | - Margitta T Kampman
- Department of Neurology, University Hospital of North Norway, Tromsø, Norway
| | - Grethe Kleveland
- Department of Neurology, Innlandet Hospital Trust, Lillehammer, Norway
| | - Unn Ljøstad
- Department of Neurology, Sørlandet Hospital Trust, Kristiansand, Norway
- Department of Clinical Medicine, University of Bergen, Norway
| | | | - Åse Hagen Morsund
- Department of Neurology, Møre og Romsdal Hospital Trust, Molde, Norway
| | - Ola Nakken
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway
| | - Katrin Schlüter
- Department of Neurology, Stavanger University Hospital, Stavanger, Norway
| | - Stephan Schuler
- Department of Neurology, Nord-Trøndelag Hospital Trust, Namsos, Norway
| | - Elin Seim
- Department of Neurology, Førde Hospital Trust, Førde, Norway, and
| | | | - Ole-Bjørn Tysnes
- Department of Clinical Medicine, University of Bergen, Norway
- Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Trygve Holmøy
- Institute of Clinical Medicine, University of Oslo, Nordbyhagen, Norway
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway
| | - Helle Høyer
- Department of Medical Genetics, Telemark Hospital Trust, Skien, Norway
- Institute of Clinical Medicine, University of Oslo, Nordbyhagen, Norway
| |
Collapse
|
3
|
Lorenc F, Dupuis L, Cassel R. Impairments of inhibitory neurons in amyotrophic lateral sclerosis and frontotemporal dementia. Neurobiol Dis 2024; 203:106748. [PMID: 39592063 DOI: 10.1016/j.nbd.2024.106748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 11/28/2024] Open
Abstract
Amyotrophic lateral sclerosis and frontotemporal dementia are two fatal neurodegenerative disorders. They are part of a pathophysiological continuum, displaying clinical, neuropathological, and genetic overlaps. There is compelling evidence that neuronal circuit dysfunction is an early feature of both diseases. Impaired neuronal excitability, imbalanced excitatory and inhibitory influences, and altered functional connectivity have been reported. These phenomena are likely due to combined alterations in the various cellular components involved in the functioning of neuronal networks. This review focuses on one of these cellular components: inhibitory neurons. We assess the evidence for inhibitory neuron impairments in amyotrophic lateral sclerosis and frontotemporal dementia, as well as the mechanisms leading to the loss of inhibition. We also discuss the contributions of these alterations to symptoms, and the potential therapeutic strategies for targeting inhibitory neuron deficits.
Collapse
Affiliation(s)
- Félicie Lorenc
- Université de Strasbourg, INSERM, UMR-S 1329, Strasbourg Translational Neuroscience and Psychiatry, CRBS, Strasbourg, France.
| | - Luc Dupuis
- Université de Strasbourg, INSERM, UMR-S 1329, Strasbourg Translational Neuroscience and Psychiatry, CRBS, Strasbourg, France.
| | - Raphaelle Cassel
- Université de Strasbourg, INSERM, UMR-S 1329, Strasbourg Translational Neuroscience and Psychiatry, CRBS, Strasbourg, France.
| |
Collapse
|
4
|
Zhang Q, Lin J, Wang Y, Chen L, Ding Y, Zheng F, Song H, Lv A, Li Y, Guo Q, Lin M, Hu W, Xu L, Zhao W, Fang L, Cui M, Fu Z, Chen W, Zhang J, Wang Z, Wang N, Fu Y. Detection of pTDP-43 via routine muscle biopsy: A promising diagnostic biomarker for amyotrophic lateral sclerosis. Brain Pathol 2024; 34:e13261. [PMID: 38602336 PMCID: PMC11483524 DOI: 10.1111/bpa.13261] [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: 11/10/2023] [Accepted: 03/28/2024] [Indexed: 04/12/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease, pathologically characterized by TDP-43 aggregates. Recent evidence has been indicated that phosphorylated TDP-43 (pTDP-43) is present not only in motor neurons but also in muscle tissues. However, it is unclear whether testing pTDP-43 aggregation in muscle tissue would assist in the diagnosis of ALS. We propose three key questions: (i) Is aggregation of pTDP-43 detectable in routine biopsied muscles? (ii) Can detection of pTDP-43 aggregation discriminate between ALS and non-ALS patients? (iii) Can pTDP-43 aggregation be observed in the early stages of ALS? We conducted a diagnostic study comprising 2 groups: an ALS group in which 18 cases underwent muscle biopsy screened from a registered ALS cohort consisting of 802 patients and a non-ALS control group, in which we randomly selected 54 muscle samples from a biospecimen bank of 684 patients. Among the 18 ALS patients, 3 patients carried pathological GGGGCC repeats in the C9ORF72 gene, 2 patients carried SOD1 mutations, and 7 patients were at an early stage with only one body region clinically affected. The pTDP-43 accumulation could be detected in routine biopsied muscles, including biceps brachii, deltoid, tibialis anterior, and quadriceps. Abnormal aggregation of pTDP-43 was present in 94.4% of ALS patients (17/18) compared to 29.6% of non-ALS controls (16/54; p < 0.001). The pTDP-43 aggregates were mainly close to the sarcolemma. Using a semi-quantified pTDP-43 aggregates score, we applied a cut-off value of 3 as a diagnostic biomarker, resulting in a sensitivity of 94.4% and a specificity of 83.3%. Moreover, we observed that accumulation of pTDP-43 occurred in muscle tissues prior to clinical symptoms and electromyographic lesions. Our study provides proof-of-concept for the detection of pTDP-43 accumulation via routine muscle biopsy which may serve as a novel biomarker for diagnosis of ALS.
Collapse
Affiliation(s)
- Qi‐Jie Zhang
- Department of Neurology, Fujian Institute of Neurology, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Neurology, National Regional Medical Center, Binhai Campus of The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Fujian Key Laboratory of Molecular NeurologyFujian Medical UniversityFuzhouChina
| | - Jie Lin
- Department of Neurology, Fujian Institute of Neurology, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Fujian Key Laboratory of Molecular NeurologyFujian Medical UniversityFuzhouChina
| | - You‐Liang Wang
- Department of Neurology, Fujian Institute of Neurology, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Fujian Key Laboratory of Molecular NeurologyFujian Medical UniversityFuzhouChina
| | - Long Chen
- Department of Neurology, Fujian Institute of Neurology, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Fujian Key Laboratory of Molecular NeurologyFujian Medical UniversityFuzhouChina
| | - Ying Ding
- Department of Neurology, Fujian Institute of Neurology, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Fujian Key Laboratory of Molecular NeurologyFujian Medical UniversityFuzhouChina
| | - Fu‐Ze Zheng
- Department of Neurology, Fujian Institute of Neurology, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Fujian Key Laboratory of Molecular NeurologyFujian Medical UniversityFuzhouChina
| | - Huan‐Huan Song
- Department of Neurology, Fujian Institute of Neurology, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Fujian Key Laboratory of Molecular NeurologyFujian Medical UniversityFuzhouChina
| | - Ao‐Wei Lv
- Department of Neurology, Fujian Institute of Neurology, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Fujian Key Laboratory of Molecular NeurologyFujian Medical UniversityFuzhouChina
| | - Yu‐Ying Li
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of ChemistryBeijing Normal UniversityBeijingChina
| | - Qi‐Fu Guo
- Department of Neurology, Fujian Institute of Neurology, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Fujian Key Laboratory of Molecular NeurologyFujian Medical UniversityFuzhouChina
| | - Min‐Ting Lin
- Department of Neurology, Fujian Institute of Neurology, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Neurology, National Regional Medical Center, Binhai Campus of The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Fujian Key Laboratory of Molecular NeurologyFujian Medical UniversityFuzhouChina
| | - Wei Hu
- Department of Neurology, Fujian Institute of Neurology, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Neurology, National Regional Medical Center, Binhai Campus of The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Fujian Key Laboratory of Molecular NeurologyFujian Medical UniversityFuzhouChina
| | - Liu‐Qing Xu
- Department of Neurology, Fujian Institute of Neurology, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Neurology, National Regional Medical Center, Binhai Campus of The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Fujian Key Laboratory of Molecular NeurologyFujian Medical UniversityFuzhouChina
| | - Wen‐Long Zhao
- Department of Neurology, Fujian Institute of Neurology, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Neurology, National Regional Medical Center, Binhai Campus of The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Fujian Key Laboratory of Molecular NeurologyFujian Medical UniversityFuzhouChina
| | - Ling Fang
- Department of Neurology, Fujian Institute of Neurology, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Neurology, National Regional Medical Center, Binhai Campus of The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Fujian Key Laboratory of Molecular NeurologyFujian Medical UniversityFuzhouChina
| | - Meng‐Chao Cui
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of ChemistryBeijing Normal UniversityBeijingChina
| | - Zhi‐Fei Fu
- Public Technology Service CenterFujian Medical UniversityFuzhouChina
| | - Wan‐Jin Chen
- Department of Neurology, Fujian Institute of Neurology, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Neurology, National Regional Medical Center, Binhai Campus of The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Fujian Key Laboratory of Molecular NeurologyFujian Medical UniversityFuzhouChina
| | - Jing Zhang
- Department of Pathology, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- National Human Brain Bank for Health and DiseaseZhejiang UniversityHangzhouChina
| | - Zhi‐Qiang Wang
- Department of Neurology, Fujian Institute of Neurology, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Neurology, National Regional Medical Center, Binhai Campus of The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Fujian Key Laboratory of Molecular NeurologyFujian Medical UniversityFuzhouChina
| | - Ning Wang
- Department of Neurology, Fujian Institute of Neurology, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Neurology, National Regional Medical Center, Binhai Campus of The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Fujian Key Laboratory of Molecular NeurologyFujian Medical UniversityFuzhouChina
| | - Ying Fu
- Department of Neurology, Fujian Institute of Neurology, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Neurology, National Regional Medical Center, Binhai Campus of The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Fujian Key Laboratory of Molecular NeurologyFujian Medical UniversityFuzhouChina
| |
Collapse
|
5
|
Ghaderi S, Mohammadi S, Fatehi F. Calcium accumulation or iron deposition: Delving into the temporal sequence of amyotrophic lateral sclerosis pathophysiology in the primary motor cortex. IBRAIN 2024; 10:375-377. [PMID: 39346793 PMCID: PMC11427793 DOI: 10.1002/ibra.12168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/14/2024] [Accepted: 06/14/2024] [Indexed: 10/01/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) causes progressive motor neuron degeneration, but an in vivo understanding of its early pathology remains limited. A recent study used topographic layer imaging to investigate iron and calcium accumulation in the primary motor cortex (M1) of patients with ALS compared with controls. Despite the preserved cortical thickness, ALS patients showed increased iron in layer 6 and calcium accumulation in layer 5a and the superficial layer. Calcium accumulation was particularly prominent in the low-myelin borders, potentially preceding the demyelination. This study reveals a novel in vivo pathology in ALS, suggesting that calcium dysregulation may precede iron accumulation and contribute to early M1 cell degeneration. Further investigation using quantitative susceptibility mapping and complementary techniques, such as diffusion kurtosis imaging, along with ultrahigh-field magnetic resonance imaging, into the role of calcium and early intervention strategies is warranted.
Collapse
Affiliation(s)
- Sadegh Ghaderi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in MedicineTehran University of Medical SciencesTehranIran
- Neuromuscular Research Center, Department of Neurology, Shariati HospitalTehran University of Medical SciencesTehranIran
| | - Sana Mohammadi
- Neuromuscular Research Center, Department of Neurology, Shariati HospitalTehran University of Medical SciencesTehranIran
| | - Farzad Fatehi
- Neuromuscular Research Center, Department of Neurology, Shariati HospitalTehran University of Medical SciencesTehranIran
- Department of NeurologyUniversity Hospitals of Leicester NHS TrustLeicesterUK
| |
Collapse
|
6
|
Wang HLV, Xiang JF, Yuan C, Veire AM, Gendron TF, Murray ME, Tansey MG, Hu J, Gearing M, Glass JD, Jin P, Corces VG, McEachin ZT. pTDP-43 levels correlate with cell type specific molecular alterations in the prefrontal cortex of C9orf72 ALS/FTD patients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.01.12.523820. [PMID: 36711601 PMCID: PMC9882184 DOI: 10.1101/2023.01.12.523820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Repeat expansions in the C9orf72 gene are the most common genetic cause of amyotrophic lateral sclerosis and familial frontotemporal dementia (ALS/FTD). To identify molecular defects that take place in the dorsolateral frontal cortex of patients with C9orf72 ALS/FTD, we compared healthy controls with C9orf72 ALS/FTD donor samples staged based on the levels of cortical phosphorylated TAR DNA binding protein (pTDP-43), a neuropathological hallmark of disease progression. We identified distinct molecular changes in different cell types that take place during FTD development. Loss of neurosurveillance microglia and activation of the complement cascade take place early, when pTDP-43 aggregates are absent or very low, and become more pronounced in late stages, suggesting an initial involvement of microglia in disease progression. Reduction of layer 2-3 cortical projection neurons with high expression of CUX2/LAMP5 also occurs early, and the reduction becomes more pronounced as pTDP-43 accumulates. Several unique features were observed only in samples with high levels of pTDP-43, including global alteration of chromatin accessibility in oligodendrocytes, microglia, and astrocytes; higher ratios of premature oligodendrocytes; increased levels of the noncoding RNA NEAT1 in astrocytes and neurons, and higher amount of phosphorylated ribosomal protein S6. Our findings reveal previously unknown progressive functional changes in major cell types found in the frontal cortex of C9orf72 ALS/FTD patients that shed light on the mechanisms underlying the pathology of this disease.
Collapse
Affiliation(s)
- Hsiao-Lin V. Wang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
- Emory Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA 30322
| | - Jian-Feng Xiang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
| | - Chenyang Yuan
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, 30322, USA
| | - Austin M. Veire
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224
| | | | | | - Malú G. Tansey
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL 32607
- Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL 32607
| | - Jian Hu
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, 30322, USA
| | - Marla Gearing
- Emory Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA 30322
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322
| | - Jonathan D. Glass
- Emory Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA 30322
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322
| | - Peng Jin
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
- Emory Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA 30322
| | - Victor G. Corces
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
- Emory Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA 30322
| | - Zachary T. McEachin
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
- Emory Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA 30322
| |
Collapse
|
7
|
Lemon R. The Corticospinal System and Amyotrophic Lateral Sclerosis: IFCN handbook chapter. Clin Neurophysiol 2024; 160:56-67. [PMID: 38401191 DOI: 10.1016/j.clinph.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/23/2023] [Accepted: 02/03/2024] [Indexed: 02/26/2024]
Abstract
Corticospinal neurons located in motor areas of the cerebral neocortex project corticospinal axons which synapse with the spinal network; a parallel corticobulbar system projects to the cranial motor network and to brainstem motor pathways. The primate corticospinal system has a widespread cortical origin and an extensive range of different fibre diameters, including thick, fast-conducting axons. Direct cortico-motoneuronal (CM) projections from the motor cortex to arm and hand alpha motoneurons are a recent evolutionary feature, that is well developed in dexterous primates and particularly in humans. Many of these projections originate from the caudal subdivision of area 4 ('new' M1: primary motor cortex). They arise from corticospinal neurons of varied soma size, including those with fast- and relatively slow-conducting axons. This CM system has been shown to be involved in the control of skilled movements, carried out with fractionation of the distal extremities and at low force levels. During movement, corticospinal neurons are activated quite differently from 'lower' motoneurons, and there is no simple or fixed functional relationship between a so-called 'upper' motoneuron and its target lower motoneuron. There are key differences in the organisation and function of the corticospinal and CM system in primates versus non-primates, such as rodents. These differences need to be recognized when making the choice of animal model for understanding disorders such as amyotrophic lateral sclerosis (ALS). In this neurodegenerative brain disease there is a selective loss of fast-conducting corticospinal axons, and their synaptic connections, and this is reflected in responses to non-invasive cortical stimuli and measures of cortico-muscular coherence. The loss of CM connections influencing distal limb muscles results in a differential loss of muscle strength or 'split-hand' phenotype. Importantly, there is also a unique impairment in the coordination of skilled hand tasks that require fractionation of digit movement. Scores on validated tests of skilled hand function could be used to assess disease progression.
Collapse
Affiliation(s)
- Roger Lemon
- Department of Clinical and Movement Sciences, Queen Square Institute of Neurology, UCL, London WC1N 3BG, UK.
| |
Collapse
|
8
|
Tzeplaeff L, Jürs AV, Wohnrade C, Demleitner AF. Unraveling the Heterogeneity of ALS-A Call to Redefine Patient Stratification for Better Outcomes in Clinical Trials. Cells 2024; 13:452. [PMID: 38474416 PMCID: PMC10930688 DOI: 10.3390/cells13050452] [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: 01/31/2024] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Despite tremendous efforts in basic research and a growing number of clinical trials aiming to find effective treatments, amyotrophic lateral sclerosis (ALS) remains an incurable disease. One possible reason for the lack of effective causative treatment options is that ALS may not be a single disease entity but rather may represent a clinical syndrome, with diverse genetic and molecular causes, histopathological alterations, and subsequent clinical presentations contributing to its complexity and variability among individuals. Defining a way to subcluster ALS patients is becoming a central endeavor in the field. Identifying specific clusters and applying them in clinical trials could enable the development of more effective treatments. This review aims to summarize the available data on heterogeneity in ALS with regard to various aspects, e.g., clinical, genetic, and molecular.
Collapse
Affiliation(s)
- Laura Tzeplaeff
- Department of Neurology, Rechts der Isar Hospital, Technical University of Munich, 81675 München, Germany
| | - Alexandra V. Jürs
- Translational Neurodegeneration Section “Albrecht Kossel”, Department of Neurology, University Medical Center Rostock, 18057 Rostock, Germany
| | - Camilla Wohnrade
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany;
| | - Antonia F. Demleitner
- Department of Neurology, Rechts der Isar Hospital, Technical University of Munich, 81675 München, Germany
| |
Collapse
|
9
|
Zhou Z, Kim J, Huang AY, Nolan M, Park J, Doan R, Shin T, Miller MB, Chhouk B, Morillo K, Yeh RC, Kenny C, Neil JE, Lee CZ, Ohkubo T, Ravits J, Ansorge O, Ostrow LW, Lagier-Tourenne C, Lee EA, Walsh CA. Somatic Mosaicism in Amyotrophic Lateral Sclerosis and Frontotemporal Dementia Reveals Widespread Degeneration from Focal Mutations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.30.569436. [PMID: 38077003 PMCID: PMC10705414 DOI: 10.1101/2023.11.30.569436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
Although mutations in dozens of genes have been implicated in familial forms of amyotrophic lateral sclerosis (fALS) and frontotemporal degeneration (fFTD), most cases of these conditions are sporadic (sALS and sFTD), with no family history, and their etiology remains obscure. We tested the hypothesis that somatic mosaic mutations, present in some but not all cells, might contribute in these cases, by performing ultra-deep, targeted sequencing of 88 genes associated with neurodegenerative diseases in postmortem brain and spinal cord samples from 404 individuals with sALS or sFTD and 144 controls. Known pathogenic germline mutations were found in 20.6% of ALS, and 26.5% of FTD cases. Predicted pathogenic somatic mutations in ALS/FTD genes were observed in 2.7% of sALS and sFTD cases that did not carry known pathogenic or novel germline mutations. Somatic mutations showed low variant allele fraction (typically <2%) and were often restricted to the region of initial discovery, preventing detection through genetic screening in peripheral tissues. Damaging somatic mutations were preferentially enriched in primary motor cortex of sALS and prefrontal cortex of sFTD, mirroring regions most severely affected in each disease. Somatic mutation analysis of bulk RNA-seq data from brain and spinal cord from an additional 143 sALS cases and 23 controls confirmed an overall enrichment of somatic mutations in sALS. Two adult sALS cases were identified bearing pathogenic somatic mutations in DYNC1H1 and LMNA, two genes associated with pediatric motor neuron degeneration. Our study suggests that somatic mutations in fALS/fFTD genes, and in genes associated with more severe diseases in the germline state, contribute to sALS and sFTD, and that mosaic mutations in a small fraction of cells in focal regions of the nervous system can ultimately result in widespread degeneration.
Collapse
Affiliation(s)
- Zinan Zhou
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA
- Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Junho Kim
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA
- Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Biological Sciences, Sungkyunkwan University, Suwon, South Korea
| | - August Yue Huang
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA
- Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Matthew Nolan
- Department of Neurology, The Sean M. Healey and AMG Center for ALS at Mass General, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Junseok Park
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA
- Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Ryan Doan
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Taehwan Shin
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA
- Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Michael B. Miller
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Brian Chhouk
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA
- Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Katherine Morillo
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA
- Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Rebecca C. Yeh
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA
- Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Connor Kenny
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA
- Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Jennifer E. Neil
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA
- Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA, USA
| | - Chao-Zong Lee
- Department of Neurology, The Sean M. Healey and AMG Center for ALS at Mass General, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Takuya Ohkubo
- Department of Neurology, Yokohama City Minato Red Cross Hospital, Yokohama, Kanagawa, Japan
- Department of Neurosciences, School of Medicine, University of California at San Diego, La Jolla, CA, USA
| | - John Ravits
- Department of Neurosciences, School of Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Olaf Ansorge
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, Oxfordshire, UK
| | - Lyle W. Ostrow
- Department of Neurology, Lewis Katz School of Medicine at Temple University, Philadelphia, USA
| | - Clotilde Lagier-Tourenne
- Department of Neurology, The Sean M. Healey and AMG Center for ALS at Mass General, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Eunjung Alice Lee
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA
- Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Christopher A. Walsh
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA
- Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA, USA
| |
Collapse
|
10
|
Halkiadakis Y, Davidson N, Morgan KD. Time series modeling characterizes stride time variability to identify individuals with neurodegenerative disorders. Hum Mov Sci 2023; 92:103152. [PMID: 37898010 DOI: 10.1016/j.humov.2023.103152] [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: 03/20/2023] [Revised: 08/18/2023] [Accepted: 10/09/2023] [Indexed: 10/30/2023]
Abstract
The progressive death and dysfunction of neurons causes altered stride-to-stride variability in individuals with Amyotrophic Lateral Sclerosis (ALS) and Huntington's Disease (HD). Yet these altered gait dynamics can manifest differently in these populations based on how and where these neurodegenerative disorders attack the central nervous system. Time series analyses can quantify differences in stride time variability which can help contribute to the detection and identification of these disorders. Here, autoregressive modeling time series analysis was utilized to quantify differences in stride time variability amongst the Controls, the individuals with ALS, and the individuals with HD. For this study, fifteen Controls, 12 individuals with ALS and 15 individuals with HD walked up and down a hallway continuously for 5-min. Participants wore force sensitive resistors in their shoes to collect stride time data. A second order autoregressive (AR) model was fit to the time series created from the stride time data. The mean stride time and two AR model coefficients served as metrics to identify differences in stride time variability amongst the three groups. The individuals with HD walked with significantly greater stride time variability indicating a more chaotic gait while the individuals with ALS adopted more ordered, less variable stride time dynamics (p < 0.001). A plot of the stride time metrics illustrated how each group exhibited significantly different stride time dynamics. The stride time metrics successfully quantified differences in stride time variability amongst individuals with neurodegenerative disorders. This work provided valuable insight about how these neuromuscular disorders disrupt motor coordination leading to the adoption of new gait dynamics.
Collapse
Affiliation(s)
- Yannis Halkiadakis
- Biomedical Engineering, School of Engineering, University of Connecticut, Storrs, CT, USA
| | - Noah Davidson
- Biomedical Engineering, School of Engineering, University of Connecticut, Storrs, CT, USA
| | - Kristin D Morgan
- Biomedical Engineering, School of Engineering, University of Connecticut, Storrs, CT, USA.
| |
Collapse
|
11
|
Tan RH, McCann H, Shepherd CE, Pinkerton M, Mazumder S, Devenney EM, Adler GL, Rowe DB, Kril J, Halliday GM, Kiernan MC. Heterogeneity of cortical pTDP-43 inclusion morphologies in amyotrophic lateral sclerosis. Acta Neuropathol Commun 2023; 11:180. [PMID: 37957721 PMCID: PMC10642010 DOI: 10.1186/s40478-023-01670-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 10/10/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND Despite the presence of significant cortical pTDP-43 inclusions of heterogeneous morphologies in patients diagnosed with amyotrophic lateral sclerosis (ALS), pathological subclassification is routinely performed in the minority of patients with concomitant frontotemporal dementia (FTD). OBJECTIVE In order to improve current understanding of the presence and relevance of pathological pTDP-43 subtypes in ALS, the present study examined the pattern of cortical pTDP-43 aggregates in 61 ALS cases without FTD. RESULTS Based on the presence, morphology and composition of pTDP-43 pathology, three distinct ALS-TDP subtypes were delineated: (1) A predominant pattern of pTDP-43 granulofilamentous neuronal inclusions (GFNIs) and grains that were immuno-negative for p62 was identified in 18% of cases designated ALS-TDP type E; (2) neuronal cytoplasmic inclusions (NCIs) that were immuno-positive for both pTDP-43 and p62 were observed in 67% of cases assigned ALS-TDP type B; and (3) scarce cortical pTDP-43 and p62 aggregates were identified in 15% of cases coined ALS-TDP type SC (scarce cortical). Quantitative analyses revealed a significantly greater burden of pTDP-43 GFNI and grains in ALS-TDP type E. Principal component analysis demonstrated significant relationships between GFNIs, grains and ALS-TDP subtypes to support the distinction of subtypes E and B. No significant difference in age at death or disease duration was found between ALS-TDP subgroups to suggest that these subtypes represent earlier or later stages of the same disease process. Instead, a significantly higher ALS-TDP stage, indicating greater topographical spread of pTDP-43, was identified in ALS-TDP type E. Alzheimer's disease neuropathological change (ABC score ≥ intermediate) and Lewy body disease (Braak stage ≥ IV) was more prevalent in the ALS-TDP type SC cohort, which also demonstrated a significantly lower overall cognitive score. CONCLUSION In summary, the present study demonstrates that ALS-TDP does not represent a single homogenous neuropathology. We propose the subclassification of ALS-TDP into three distinct subtypes using standard immuno-stains for pTDP-43 and p62 in the motor cortex, which is routinely sampled and evaluated for diagnostic neuropathological characterisation of ALS. We propose that future studies specify both clinicopathological group and pTDP-43 subtype to advance current understanding of the pathogenesis of clinical phenotypes in pTDP-43 proteinopathies, which will have significant relevance to the development of targeted therapies for this heterogeneous disorder.
Collapse
Affiliation(s)
- Rachel H Tan
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia.
- Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Camperdown, NSW, Australia.
| | - Heather McCann
- Neuroscience Research Australia, Randwick, NSW, Australia
| | | | - Monica Pinkerton
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Camperdown, NSW, Australia
| | - Srestha Mazumder
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
| | - Emma M Devenney
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
| | - Gabrielle L Adler
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Camperdown, NSW, Australia
| | - Dominic B Rowe
- Macquarie University Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Jillian Kril
- Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Camperdown, NSW, Australia
- Dementia Research Centre, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Glenda M Halliday
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Camperdown, NSW, Australia
| | - Matthew C Kiernan
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
- Institute of Clinical Neurosciences, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| |
Collapse
|
12
|
Tsuboguchi S, Nakamura Y, Ishihara T, Kato T, Sato T, Koyama A, Mori H, Koike Y, Onodera O, Ueno M. TDP-43 differentially propagates to induce antero- and retrograde degeneration in the corticospinal circuits in mouse focal ALS models. Acta Neuropathol 2023; 146:611-629. [PMID: 37555859 DOI: 10.1007/s00401-023-02615-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 06/22/2023] [Accepted: 07/15/2023] [Indexed: 08/10/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease characterized by TDP-43 inclusions in the cortical and spinal motor neurons. It remains unknown whether and how pathogenic TDP-43 spreads across neural connections to progress degenerative processes in the cortico-spinal motor circuitry. Here we established novel mouse ALS models that initially induced mutant TDP-43 inclusions in specific neuronal or cell types in the motor circuits, and investigated whether TDP-43 and relevant pathological processes spread across neuronal or cellular connections. We first developed ALS models that primarily induced TDP-43 inclusions in the corticospinal neurons, spinal motor neurons, or forelimb skeletal muscle, by using adeno-associated virus (AAV) expressing mutant TDP-43. We found that TDP-43 induced in the corticospinal neurons was transported along the axons anterogradely and transferred to the oligodendrocytes along the corticospinal tract (CST), coinciding with mild axon degeneration. In contrast, TDP-43 introduced in the spinal motor neurons did not spread retrogradely to the cortical or spinal neurons; however, it induced an extreme loss of spinal motor neurons and subsequent degeneration of neighboring spinal neurons, suggesting a degenerative propagation in a retrograde manner in the spinal cord. The intraspinal degeneration further led to severe muscle atrophy. Finally, TDP-43 induced in the skeletal muscle did not propagate pathological events to spinal neurons retrogradely. Our data revealed that mutant TDP-43 spread across neuro-glial connections anterogradely in the corticospinal pathway, whereas it exhibited different retrograde degenerative properties in the spinal circuits. This suggests that pathogenic TDP-43 may induce distinct antero- and retrograde mechanisms of degeneration in the motor system in ALS.
Collapse
Affiliation(s)
- Shintaro Tsuboguchi
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Niigata, 951-8585, Japan
| | - Yuka Nakamura
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Japan
| | - Tomohiko Ishihara
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Niigata, 951-8585, Japan
| | - Taisuke Kato
- Department of Molecular Neuroscience, Brain Research Institute, Niigata University, Niigata, Japan
| | - Tokiharu Sato
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Japan
| | - Akihide Koyama
- Division of Legal Medicine, Graduate School of Medicine and Dental Sciences, Niigata University, Niigata, Japan
| | - Hideki Mori
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Niigata, 951-8585, Japan
| | - Yuka Koike
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Niigata, 951-8585, Japan
| | - Osamu Onodera
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Niigata, 951-8585, Japan.
- Department of Molecular Neuroscience, Brain Research Institute, Niigata University, Niigata, Japan.
| | - Masaki Ueno
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Japan.
| |
Collapse
|
13
|
Gao C, Jiang J, Tan Y, Chen S. Microglia in neurodegenerative diseases: mechanism and potential therapeutic targets. Signal Transduct Target Ther 2023; 8:359. [PMID: 37735487 PMCID: PMC10514343 DOI: 10.1038/s41392-023-01588-0] [Citation(s) in RCA: 192] [Impact Index Per Article: 96.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/11/2023] [Accepted: 08/03/2023] [Indexed: 09/23/2023] Open
Abstract
Microglia activation is observed in various neurodegenerative diseases. Recent advances in single-cell technologies have revealed that these reactive microglia were with high spatial and temporal heterogeneity. Some identified microglia in specific states correlate with pathological hallmarks and are associated with specific functions. Microglia both exert protective function by phagocytosing and clearing pathological protein aggregates and play detrimental roles due to excessive uptake of protein aggregates, which would lead to microglial phagocytic ability impairment, neuroinflammation, and eventually neurodegeneration. In addition, peripheral immune cells infiltration shapes microglia into a pro-inflammatory phenotype and accelerates disease progression. Microglia also act as a mobile vehicle to propagate protein aggregates. Extracellular vesicles released from microglia and autophagy impairment in microglia all contribute to pathological progression and neurodegeneration. Thus, enhancing microglial phagocytosis, reducing microglial-mediated neuroinflammation, inhibiting microglial exosome synthesis and secretion, and promoting microglial conversion into a protective phenotype are considered to be promising strategies for the therapy of neurodegenerative diseases. Here we comprehensively review the biology of microglia and the roles of microglia in neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, multiple system atrophy, amyotrophic lateral sclerosis, frontotemporal dementia, progressive supranuclear palsy, corticobasal degeneration, dementia with Lewy bodies and Huntington's disease. We also summarize the possible microglia-targeted interventions and treatments against neurodegenerative diseases with preclinical and clinical evidence in cell experiments, animal studies, and clinical trials.
Collapse
Affiliation(s)
- Chao Gao
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Jingwen Jiang
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Yuyan Tan
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| | - Shengdi Chen
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
- Lab for Translational Research of Neurodegenerative Diseases, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), Shanghai Tech University, 201210, Shanghai, China.
| |
Collapse
|
14
|
Li J, Jaiswal MK, Chien JF, Kozlenkov A, Jung J, Zhou P, Gardashli M, Pregent LJ, Engelberg-Cook E, Dickson DW, Belzil VV, Mukamel EA, Dracheva S. Divergent single cell transcriptome and epigenome alterations in ALS and FTD patients with C9orf72 mutation. Nat Commun 2023; 14:5714. [PMID: 37714849 PMCID: PMC10504300 DOI: 10.1038/s41467-023-41033-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 08/21/2023] [Indexed: 09/17/2023] Open
Abstract
A repeat expansion in the C9orf72 (C9) gene is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Here we investigate single nucleus transcriptomics (snRNA-seq) and epigenomics (snATAC-seq) in postmortem motor and frontal cortices from C9-ALS, C9-FTD, and control donors. C9-ALS donors present pervasive alterations of gene expression with concordant changes in chromatin accessibility and histone modifications. The greatest alterations occur in upper and deep layer excitatory neurons, as well as in astrocytes. In neurons, the changes imply an increase in proteostasis, metabolism, and protein expression pathways, alongside a decrease in neuronal function. In astrocytes, the alterations suggest activation and structural remodeling. Conversely, C9-FTD donors have fewer high-quality neuronal nuclei in the frontal cortex and numerous gene expression changes in glial cells. These findings highlight a context-dependent molecular disruption in C9-ALS and C9-FTD, indicating unique effects across cell types, brain regions, and diseases.
Collapse
Affiliation(s)
- Junhao Li
- Department of Cognitive Science, University of California San Diego, La Jolla, CA, 92037, US
| | - Manoj K Jaiswal
- Friedman Brain Institute and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, US
| | - Jo-Fan Chien
- Department of Physics, University of California San Diego, La Jolla, CA, 92037, US
| | - Alexey Kozlenkov
- Friedman Brain Institute and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, US
| | - Jinyoung Jung
- Friedman Brain Institute and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, US
| | - Ping Zhou
- Friedman Brain Institute and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, US
| | | | - Luc J Pregent
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, US
| | | | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, US
| | | | - Eran A Mukamel
- Department of Cognitive Science, University of California San Diego, La Jolla, CA, 92037, US.
| | - Stella Dracheva
- Friedman Brain Institute and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, US.
- Research & Development and VISN2 MIREC, James J, Peters VA Medical Center, Bronx, NY, 10468, US.
| |
Collapse
|
15
|
Swanson MEV, Mrkela M, Murray HC, Cao MC, Turner C, Curtis MA, Faull RLM, Walker AK, Scotter EL. Microglial CD68 and L-ferritin upregulation in response to phosphorylated-TDP-43 pathology in the amyotrophic lateral sclerosis brain. Acta Neuropathol Commun 2023; 11:69. [PMID: 37118836 PMCID: PMC10142752 DOI: 10.1186/s40478-023-01561-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 03/29/2023] [Indexed: 04/30/2023] Open
Abstract
Microglia, the innate immune cells of the brain, are activated by damage or disease. In mouse models of amyotrophic lateral sclerosis (ALS), microglia shift from neurotrophic to neurotoxic states with disease progression. It remains unclear how human microglia change relative to the TAR DNA-binding protein 43 (TDP-43) aggregation that occurs in 97% of ALS cases. Here we examine spatial relationships between microglial activation and TDP-43 pathology in brain tissue from people with ALS and from a TDP-43-driven ALS mouse model. Post-mortem human brain tissue from the Neurological Foundation Human Brain Bank was obtained from 10 control and 10 ALS cases in parallel with brain tissue from a bigenic NEFH-tTA/tetO-hTDP-43∆NLS (rNLS) mouse model of ALS at disease onset, early disease, and late disease stages. The spatiotemporal relationship between microglial activation and ALS pathology was determined by investigating microglial functional marker expression in brain regions with low and high TDP-43 burden at end-stage human disease: hippocampus and motor cortex, respectively. Sections were immunohistochemically labelled with a two-round multiplexed antibody panel against; microglial functional markers (L-ferritin, HLA-DR, CD74, CD68, and Iba1), a neuronal marker, an astrocyte marker, and pathological phosphorylated TDP-43 (pTDP-43). Single-cell levels of microglial functional markers were quantified using custom analysis pipelines and mapped to anatomical regions and ALS pathology. We identified a significant increase in microglial Iba1 and CD68 expression in the human ALS motor cortex, with microglial CD68 being significantly correlated with pTDP-43 pathology load. We also identified two subpopulations of microglia enriched in the ALS motor cortex that were defined by high L-ferritin expression. A similar pattern of microglial changes was observed in the rNLS mouse, with an increase first in CD68 and then in L-ferritin expression, with both occurring only after pTDP-43 inclusions were detectable. Our data strongly suggest that microglia are phagocytic at early-stage ALS but transition to a dysfunctional state at end-stage disease, and that these functional states are driven by pTDP-43 aggregation. Overall, these findings enhance our understanding of microglial phenotypes and function in ALS.
Collapse
Affiliation(s)
- Molly E V Swanson
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Miran Mrkela
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Helen C Murray
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Maize C Cao
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Department of Pharmacology and Clinical Pharmacology, University of Auckland, Auckland, New Zealand
| | - Clinton Turner
- Department of Anatomical Pathology, Pathology and Laboratory Medicine, Auckland City Hospital, Auckland, New Zealand
| | - Maurice A Curtis
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Richard L M Faull
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Adam K Walker
- Queensland Brain Institute, University of Queensland, Brisbane, Australia
| | - Emma L Scotter
- School of Biological Sciences, University of Auckland, Auckland, New Zealand.
- Centre for Brain Research, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
16
|
Filipi T, Matusova Z, Abaffy P, Vanatko O, Tureckova J, Benesova S, Kubiskova M, Kirdajova D, Zahumensky J, Valihrach L, Anderova M. Cortical glia in SOD1(G93A) mice are subtly affected by ALS-like pathology. Sci Rep 2023; 13:6538. [PMID: 37085528 PMCID: PMC10121704 DOI: 10.1038/s41598-023-33608-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 04/15/2023] [Indexed: 04/23/2023] Open
Abstract
The role of glia in amyotrophic lateral sclerosis (ALS) is undeniable. Their disease-related activity has been extensively studied in the spinal cord, but only partly in the brain. We present herein a comprehensive study of glia in the cortex of SOD1(G93A) mice-a widely used model of ALS. Using single-cell RNA sequencing (scRNA-seq) and immunohistochemistry, we inspected astrocytes, microglia, and oligodendrocytes, in four stages of the disease, respecting the factor of sex. We report minimal changes of glia throughout the disease progression and regardless of sex. Pseudobulk and single-cell analyses revealed subtle disease-related transcriptional alterations at the end-stage in microglia and oligodendrocytes, which were supported by immunohistochemistry. Therefore, our data support the hypothesis that the SOD1(G93A) mouse cortex does not recapitulate the disease in patients, and we recommend the use of a different model for future studies of the cortical ALS pathology.
Collapse
Affiliation(s)
- Tereza Filipi
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, 14220, Prague, Czech Republic
- Second Faculty of Medicine, Charles University, V Uvalu 84, 15006, Prague, Czech Republic
| | - Zuzana Matusova
- Laboratory of Gene Expression, Institute of Biotechnology CAS, BIOCEV, Prumyslova 595, 25250, Vestec, Czech Republic
- Faculty of Science, Charles University, Albertov 6, 12800, Prague, Czech Republic
| | - Pavel Abaffy
- Laboratory of Gene Expression, Institute of Biotechnology CAS, BIOCEV, Prumyslova 595, 25250, Vestec, Czech Republic
| | - Ondrej Vanatko
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, 14220, Prague, Czech Republic
- Second Faculty of Medicine, Charles University, V Uvalu 84, 15006, Prague, Czech Republic
| | - Jana Tureckova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, 14220, Prague, Czech Republic
| | - Sarka Benesova
- Laboratory of Gene Expression, Institute of Biotechnology CAS, BIOCEV, Prumyslova 595, 25250, Vestec, Czech Republic
- Department of Informatics and Chemistry, Faculty of Chemical Technology, University of Chemistry and Technology, Technicka 5, 16628, Prague, Czech Republic
| | - Monika Kubiskova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, 14220, Prague, Czech Republic
| | - Denisa Kirdajova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, 14220, Prague, Czech Republic
| | - Jakub Zahumensky
- Department of Functional Organization of Biomembranes, Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, 14220, Prague, Czech Republic
| | - Lukas Valihrach
- Laboratory of Gene Expression, Institute of Biotechnology CAS, BIOCEV, Prumyslova 595, 25250, Vestec, Czech Republic.
| | - Miroslava Anderova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, 14220, Prague, Czech Republic.
| |
Collapse
|
17
|
Canosa A, Calvo A, Moglia C, Vasta R, Palumbo F, Fuda G, Di Pede F, Cabras S, Arena V, Novara A, Salamone P, Matteoni E, Sbaiz L, Gallone S, Grassano M, Manera U, Chiò A, Pagani M. Brain 18fluorodeoxyglucose-positron emission tomography changes in amyotrophic lateral sclerosis with TARDBP mutations. J Neurol Neurosurg Psychiatry 2022; 93:1021-1023. [PMID: 35351820 DOI: 10.1136/jnnp-2021-328296] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/14/2022] [Indexed: 11/04/2022]
Affiliation(s)
- Antonio Canosa
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy .,SC Neurologia 1U, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Turin, Italy.,Institute of Cognitive Sciences and Technologies, C.N.R, Rome, Italy
| | - Andrea Calvo
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy.,SC Neurologia 1U, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Turin, Italy.,Neuroscience Institute of Turin (NIT), Turin, Italy
| | - Cristina Moglia
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy.,SC Neurologia 1U, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Turin, Italy
| | - Rosario Vasta
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
| | - Francesca Palumbo
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
| | - Giuseppe Fuda
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
| | - Francesca Di Pede
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
| | - Sara Cabras
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
| | - Vincenzo Arena
- Positron Emission Tomography Centre AFFIDEA-IRMET S.p.A, Turin, Italy
| | - Andrea Novara
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
| | - Paolina Salamone
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
| | - Enrico Matteoni
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
| | - Luca Sbaiz
- Laboratory of Genetics, Department of Clinical Pathology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Turin, Italy
| | - Salvatore Gallone
- SC Neurologia 1U, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Turin, Italy
| | - Maurizio Grassano
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
| | - Umberto Manera
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy.,SC Neurologia 1U, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Turin, Italy
| | - Adriano Chiò
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy.,SC Neurologia 1U, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Turin, Italy.,Institute of Cognitive Sciences and Technologies, C.N.R, Rome, Italy.,Neuroscience Institute of Turin (NIT), Turin, Italy
| | - Marco Pagani
- Institute of Cognitive Sciences and Technologies, C.N.R, Rome, Italy.,Department of Medical Radiation Physics and Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
18
|
Canosa A, Calvo A, Moglia C, Vasta R, Palumbo F, Solero L, Di Pede F, Cabras S, Arena V, Zocco G, Casale F, Brunetti M, Sbaiz L, Gallone S, Grassano M, Manera U, Pagani M, Chiò A. Amyotrophic lateral sclerosis with SOD1 mutations shows distinct brain metabolic changes. Eur J Nucl Med Mol Imaging 2022; 49:2242-2250. [PMID: 35076740 PMCID: PMC9165265 DOI: 10.1007/s00259-021-05668-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 12/20/2021] [Indexed: 12/11/2022]
Abstract
PURPOSE Neuropathological data suggest that ALS with SOD1 mutations (SOD1-ALS) is a distinct form of ALS. We evaluated brain metabolic changes characterizing SOD1-ALS as compared to sporadic ALS (sALS), employing 18fluorodeoxyglucose-positron-emission tomography (18F-FDG-PET). METHODS We included 18 SOD1-ALS patients, 40 healthy controls (HC), and 46 sALS patients without mutations in SOD1, TARDBP, FUS, and C9ORF72, randomly selected from 665 subjects who underwent brain 18F-FDG-PET at diagnosis between 2008 and 2019 at the ALS Centre of Turin. We excluded patients with frontotemporal dementia. We used the full factorial design in SPM12 to evaluate whether differences among groups exist overall. In case the hypothesis was confirmed, group comparisons were performed through the two-sample t-test model of SPM12. In all the analyses, the height threshold was P < 0.001 (P < 0.05 FWE-corrected at cluster level). RESULTS The full factorial design resulted in a significant main effect of groups. We identified a relative hypometabolism in sALS patients compared to SOD1-ALS cases in the right precentral and medial frontal gyrus, right paracentral lobule, and bilateral postcentral gyrus. SOD1 patients showed a relative hypermetabolism as compared to HC in the right precentral gyrus and paracentral lobule. As compared to HC, sALS patients showed relative hypometabolism in frontal, temporal, and occipital cortices. CONCLUSION SOD1-ALS was characterized by a relative hypermetabolism in the motor cortex as compared to sALS and HC. Since promising, targeted, therapeutic strategies are upcoming for SOD1-ALS, our data support the use of PET to study disease pathogenesis and to track its course in clinical trials, in both asymptomatic and symptomatic mutation carriers.
Collapse
Affiliation(s)
- Antonio Canosa
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy.
- Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, SC Neurologia 1U, Turin, Italy.
- Institute of Cognitive Sciences and Technologies, C.N.R., Rome, Italy.
| | - Andrea Calvo
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
- Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, SC Neurologia 1U, Turin, Italy
- Neuroscience Institute of Turin (NIT), Turin, Italy
| | - Cristina Moglia
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
- Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, SC Neurologia 1U, Turin, Italy
| | - Rosario Vasta
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
| | - Francesca Palumbo
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
| | - Luca Solero
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
| | - Francesca Di Pede
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
| | - Sara Cabras
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
| | - Vincenzo Arena
- Positron Emission Tomography Centre AFFIDEA-IRMET S.p.A., Turin, Italy
| | - Grazia Zocco
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
| | - Federico Casale
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
| | - Maura Brunetti
- Laboratory of Genetics, Department of Clinical Pathology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Turin, Italy
| | - Luca Sbaiz
- Laboratory of Genetics, Department of Clinical Pathology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Turin, Italy
| | - Salvatore Gallone
- Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, SC Neurologia 1U, Turin, Italy
| | - Maurizio Grassano
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
| | - Umberto Manera
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
- Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, SC Neurologia 1U, Turin, Italy
| | - Marco Pagani
- Institute of Cognitive Sciences and Technologies, C.N.R., Rome, Italy
- Department of Medical Radiation Physics and Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Adriano Chiò
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
- Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, SC Neurologia 1U, Turin, Italy
- Institute of Cognitive Sciences and Technologies, C.N.R., Rome, Italy
- Neuroscience Institute of Turin (NIT), Turin, Italy
| |
Collapse
|
19
|
Kon T, Mori F, Tanji K, Miki Y, Nishijima H, Nakamura T, Kinoshita I, Suzuki C, Kurotaki H, Tomiyama M, Wakabayashi K. Accumulation of Nonfibrillar TDP-43 in the Rough Endoplasmic Reticulum Is the Early-Stage Pathology in Amyotrophic Lateral Sclerosis. J Neuropathol Exp Neurol 2022; 81:271-281. [PMID: 35294549 DOI: 10.1093/jnen/nlac015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Transactivation response DNA-binding protein 43 (TDP-43)-immunoreactive neuronal cytoplasmic inclusions (NCIs) are the histopathological hallmarks of amyotrophic lateral sclerosis (ALS). They are classified as skein-like inclusions, round inclusions, dot-like inclusions, linear wisps, and diffuse punctate cytoplasmic staining (DPCS). We hypothesized that TDP-43-immunoreactive DPCS may form the early-stage pathology of ALS. Hence, we investigated phosphorylated TDP-43 pathology in the upper and lower motor neurons of patients with ALS and control participants. We designated patients whose disease duration was ≤1 year as short-duration ALS (n = 7) and those whose duration equaled 3-5 years as standard-duration ALS (n = 6). DPCS and skein-like inclusions were the most common NCIs in short-duration and standard-duration ALS, respectively. The density of DPCS was significantly higher in short-duration ALS than that in standard-duration ALS and was inversely correlated with disease duration. DPCS was not ubiquitinated and disappeared after proteinase K treatment, suggesting that it was not aggregated. Immunoelectron microscopy revealed that DPCS corresponded to nonfibrillar TDP-43 localized to the ribosomes of the rough endoplasmic reticulum (ER). These findings suggest that nonfibrillar TDP-43 accumulation in the rough ER is the earliest TDP-43 pathology in ALS, which may be helpful in developing future TDP-43 breakdown strategies for ALS.
Collapse
Affiliation(s)
- Tomoya Kon
- From the Department of Neurology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Fumiaki Mori
- Department of Neuropathology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Kunikazu Tanji
- Department of Neuropathology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Yasuo Miki
- Department of Neuropathology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Haruo Nishijima
- From the Department of Neurology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Takashi Nakamura
- From the Department of Neurology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Iku Kinoshita
- From the Department of Neurology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Chieko Suzuki
- From the Department of Neurology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Hidekachi Kurotaki
- Department of Pathology, Aomori Prefectural Central Hospital, Aomori, Japan
| | - Masahiko Tomiyama
- From the Department of Neurology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Koichi Wakabayashi
- Department of Neuropathology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| |
Collapse
|
20
|
Prater KE, Latimer CS, Jayadev S. Glial TDP-43 and TDP-43 induced glial pathology, focus on neurodegenerative proteinopathy syndromes. Glia 2022; 70:239-255. [PMID: 34558120 PMCID: PMC8722378 DOI: 10.1002/glia.24096] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 08/21/2021] [Accepted: 09/09/2021] [Indexed: 02/03/2023]
Abstract
Since its discovery in 2006, TAR DNA binding protein 43 (TDP-43) has driven rapidly evolving research in neurodegenerative diseases including amyotrophic lateral sclerosis (ALS), frontotemporal lobar degeneration (FTLD), and limbic predominant age-related TDP-43 encephalopathy (LATE). TDP-43 mislocalization or aggregation is the hallmark of TDP-43 proteinopathy and is associated with cognitive impairment that can be mapped to its regional deposition. Studies in human tissue and model systems demonstrate that TDP-43 may potentiate other proteinopathies such as the amyloid or tau pathology seen in Alzheimer's Disease (AD) in the combination of AD+LATE. Despite this growing body of literature, there remain gaps in our understanding of whether there is heterogeneity in TDP-43 driven mechanisms across cell types. The growing observations of correlation between TDP-43 proteinopathy and glial pathology suggest a relationship between the two, including pathogenic glial cell-autonomous dysfunction and dysregulated glial immune responses to neuronal TDP-43. In this review, we discuss the available data on TDP-43 in glia within the context of the neurodegenerative diseases ALS and FTLD and highlight the current lack of information about glial TDP-43 interaction in AD+LATE. TDP-43 has proven to be a significant modulator of cognitive and neuropathological outcomes. A deeper understanding of its role in diverse cell types may provide relevant insights into neurodegenerative syndromes.
Collapse
Affiliation(s)
| | - Caitlin S. Latimer
- Division of Neuropathology, Department of Pathology, University of Washington, Seattle, WA 98195
| | - Suman Jayadev
- Department of Neurology, University of Washington, Seattle, WA 98195,Division of Neuropathology, Department of Pathology, University of Washington, Seattle, WA 98195
| |
Collapse
|
21
|
Reyes-Leiva D, Dols-Icardo O, Sirisi S, Cortés-Vicente E, Turon-Sans J, de Luna N, Blesa R, Belbin O, Montal V, Alcolea D, Fortea J, Lleó A, Rojas-García R, Illán-Gala I. Pathophysiological Underpinnings of Extra-Motor Neurodegeneration in Amyotrophic Lateral Sclerosis: New Insights From Biomarker Studies. Front Neurol 2022; 12:750543. [PMID: 35115992 PMCID: PMC8804092 DOI: 10.3389/fneur.2021.750543] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 12/09/2021] [Indexed: 11/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) lie at opposing ends of a clinical, genetic, and neuropathological continuum. In the last decade, it has become clear that cognitive and behavioral changes in patients with ALS are more frequent than previously recognized. Significantly, these non-motor features can impact the diagnosis, prognosis, and management of ALS. Partially overlapping neuropathological staging systems have been proposed to describe the distribution of TAR DNA-binding protein 43 (TDP-43) aggregates outside the corticospinal tract. However, the relationship between TDP-43 inclusions and neurodegeneration is not absolute and other pathophysiological processes, such as neuroinflammation (with a prominent role of microglia), cortical hyperexcitability, and synaptic dysfunction also play a central role in ALS pathophysiology. In the last decade, imaging and biofluid biomarker studies have revealed important insights into the pathophysiological underpinnings of extra-motor neurodegeneration in the ALS-FTLD continuum. In this review, we first summarize the clinical and pathophysiological correlates of extra-motor neurodegeneration in ALS. Next, we discuss the diagnostic and prognostic value of biomarkers in ALS and their potential to characterize extra-motor neurodegeneration. Finally, we debate about how biomarkers could improve the diagnosis and classification of ALS. Emerging imaging biomarkers of extra-motor neurodegeneration that enable the monitoring of disease progression are particularly promising. In addition, a growing arsenal of biofluid biomarkers linked to neurodegeneration and neuroinflammation are improving the diagnostic accuracy and identification of patients with a faster progression rate. The development and validation of biomarkers that detect the pathological aggregates of TDP-43 in vivo are notably expected to further elucidate the pathophysiological underpinnings of extra-motor neurodegeneration in ALS. Novel biomarkers tracking the different aspects of ALS pathophysiology are paving the way to precision medicine approaches in the ALS-FTLD continuum. These are essential steps to improve the diagnosis and staging of ALS and the design of clinical trials testing novel disease-modifying treatments.
Collapse
Affiliation(s)
- David Reyes-Leiva
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Oriol Dols-Icardo
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Sonia Sirisi
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Elena Cortés-Vicente
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Janina Turon-Sans
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Noemi de Luna
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Rafael Blesa
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Olivia Belbin
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Victor Montal
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Daniel Alcolea
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Alberto Lleó
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Ricard Rojas-García
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Ignacio Illán-Gala
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
- *Correspondence: Ignacio Illán-Gala
| |
Collapse
|
22
|
Rossi S, Cozzolino M. Dysfunction of RNA/RNA-Binding Proteins in ALS Astrocytes and Microglia. Cells 2021; 10:cells10113005. [PMID: 34831228 PMCID: PMC8616248 DOI: 10.3390/cells10113005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/30/2021] [Accepted: 11/02/2021] [Indexed: 12/24/2022] Open
Abstract
Amyotrophic Lateral Sclerosis is a neurological disease that primarily affects motor neurons in the cortex, brainstem, and spinal cord. The process that leads to motor neuron degeneration is strongly influenced by non-motor neuronal events that occur in a variety of cell types. Among these, neuroinflammatory processes mediated by activated astrocytes and microglia play a relevant role. In recent years, it has become clear that dysregulation of essential steps of RNA metabolism, as a consequence of alterations in RNA-binding proteins (RBPs), is a central event in the degeneration of motor neurons. Yet, a causal link between dysfunctional RNA metabolism and the neuroinflammatory processes mediated by astrocytes and microglia in ALS has been poorly defined. In this review, we will discuss the available evidence showing that RBPs and associated RNA processing are affected in ALS astrocytes and microglia, and the possible mechanisms involved in these events.
Collapse
|
23
|
Gotkine M, de Majo M, Wong CH, Topp SD, Michaelson-Cohen R, Epsztejn-Litman S, Eiges R, Y YL, Kanaan M, Shaked HM, Alahmady N, Vance C, Newhouse SJ, Breen G, Nishimura AL, Shaw CE, Smith BN. A recessive S174X mutation in Optineurin causes amyotrophic lateral sclerosis through a loss of function via allele-specific nonsense-mediated decay. Neurobiol Aging 2021; 106:351.e1-351.e6. [PMID: 34272080 DOI: 10.1016/j.neurobiolaging.2021.05.009] [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: 11/15/2020] [Revised: 04/13/2021] [Accepted: 05/13/2021] [Indexed: 10/21/2022]
Abstract
Loss of function (LoF) mutations in Optineurin can cause recessive amyotrophic lateral sclerosis (ALS) with some heterozygous LoF mutations associated with dominant ALS. The molecular mechanisms underlying the variable inheritance pattern associated with OPTN mutations have remained elusive. We identified that affected members of a consanguineous Middle Eastern ALS kindred possessed a novel homozygous p.S174X OPTN mutation. Analysis of these primary fibroblast lines from family members identified that the p.S174X mutation reduces OPTN mRNA expression in an allele-dependent fashion by nonsense mediated decay. Western blotting correlated a reduced expression in heterozygote carriers but a complete absence of OPTN protein in the homozygous carrier. This data suggests that the p.S174X truncation mutation causes recessive ALS through LoF. However, functional analysis detected a significant increase in mitophagy markers TOM20 and COXIV, and higher rates of mitochondrial respiration and ATP levels in heterozygous carriers only. This suggests that heterozygous LoF OPTN mutations may not be causative in a Mendelian manner but may potentially behave as contributory ALS risk factors.
Collapse
Affiliation(s)
- Marc Gotkine
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Martina de Majo
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Chun Hao Wong
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Simon D Topp
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; United Kingdom Dementia Research Institute Centre, King's College London, London, UK
| | - Rachel Michaelson-Cohen
- Medical Genetics Institute, Department of Obstetrics & Gynecology, Shaare Zedek Medical Center affiliated with the Hebrew University School of Medicine, Jerusalem, Israel
| | - Silvina Epsztejn-Litman
- Medical Genetics Institute, Department of Obstetrics & Gynecology, Shaare Zedek Medical Center affiliated with the Hebrew University School of Medicine, Jerusalem, Israel
| | - Rachel Eiges
- Medical Genetics Institute, Department of Obstetrics & Gynecology, Shaare Zedek Medical Center affiliated with the Hebrew University School of Medicine, Jerusalem, Israel
| | - Yossef Lerner Y
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Moein Kanaan
- Hereditary Research Laboratory, Bethlehem University, Jerusalem, Israel
| | - Hagar Mor Shaked
- Department of Genetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Nada Alahmady
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Department of Biology, Imam Abdulrahman bin Faisal University, Dammam, Saudi Arabia
| | - Caroline Vance
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Stephen J Newhouse
- Department of Biostatistics and Health Informatics, King's College London, London, UK
| | - Gerome Breen
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Agnes L Nishimura
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Christopher E Shaw
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; United Kingdom Dementia Research Institute Centre, King's College London, London, UK
| | - Bradley N Smith
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Stem Cell Research Laboratory, Medical Genetics Institute, Shaare Zedek Medical Center affiliated with the Hebrew University School of Medicine, Jerusalem, Israel.
| |
Collapse
|
24
|
Valori CF, Neumann M. Contribution of RNA/DNA Binding Protein Dysfunction in Oligodendrocytes in the Pathogenesis of the Amyotrophic Lateral Sclerosis/Frontotemporal Lobar Degeneration Spectrum Diseases. Front Neurosci 2021; 15:724891. [PMID: 34539339 PMCID: PMC8440855 DOI: 10.3389/fnins.2021.724891] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 07/31/2021] [Indexed: 12/19/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) are two incurable neurodegenerative disorders, often considered as the extreme manifestations of a disease spectrum, as they share similar pathomechanisms. In support of this, pathological aggregation of the RNA/DNA binding proteins trans-activation response element DNA-binding protein 43 (TDP-43) or fused in sarcoma (FUS) is the pathological hallmark found in neurons and glial cells of subsets of patients affected by either condition (i.e., ALS/FTLD—TDP-43 or ALS/FTLD—FUS, respectively). Among glia, oligodendrocytes are the most abundant population, designated to ensheath the axons with myelin and to provide them with metabolic and trophic support. In this minireview, we recapitulate the neuropathological evidence for oligodendroglia impairment in ALS/FTLD. We then debate how TDP-43 and FUS target oligodendrocyte transcripts, thereby controlling their homeostatic abilities toward the axons. Finally, we discuss cellular and animal models aimed at investigating the functional consequences of manipulating TDP-43 and FUS in oligodendrocytes in vivo. Taken together, current data provide increasing evidence for an important role of TDP-43 and FUS-mediated oligodendroglia dysfunction in the pathogenesis of ALS/FTLD. Thus, targeting disrupted oligodendroglial functions may represent a new treatment approach for these conditions.
Collapse
Affiliation(s)
- Chiara F Valori
- Molecular Neuropathology of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Manuela Neumann
- Molecular Neuropathology of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases, Tübingen, Germany.,Department of Neuropathology, University Hospital of Tübingen, Tübingen, Germany
| |
Collapse
|
25
|
Blood-spinal cord barrier leakage is independent of motor neuron pathology in ALS. Acta Neuropathol Commun 2021; 9:144. [PMID: 34446086 PMCID: PMC8393479 DOI: 10.1186/s40478-021-01244-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 08/11/2021] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease involving progressive degeneration of upper and lower motor neurons. The pattern of lower motor neuron loss along the spinal cord follows the pattern of deposition of phosphorylated TDP-43 aggregates. The blood-spinal cord barrier (BSCB) restricts entry into the spinal cord parenchyma of blood components that can promote motor neuron degeneration, but in ALS there is evidence for barrier breakdown. Here we sought to quantify BSCB breakdown along the spinal cord axis, to determine whether BSCB breakdown displays the same patterning as motor neuron loss and TDP-43 proteinopathy. Cerebrospinal fluid hemoglobin was measured in living ALS patients (n = 87 control, n = 236 ALS) as a potential biomarker of BSCB and blood–brain barrier leakage. Cervical, thoracic, and lumbar post-mortem spinal cord tissue (n = 5 control, n = 13 ALS) were then immunolabelled and semi-automated imaging and analysis performed to quantify hemoglobin leakage, lower motor neuron loss, and phosphorylated TDP-43 inclusion load. Hemoglobin leakage was observed along the whole ALS spinal cord axis and was most severe in the dorsal gray and white matter in the thoracic spinal cord. In contrast, motor neuron loss and TDP-43 proteinopathy were seen at all three levels of the ALS spinal cord, with most abundant TDP-43 deposition in the anterior gray matter of the cervical and lumbar cord. Our data show that leakage of the BSCB occurs during life, but at end-stage disease the regions with most severe BSCB damage are not those where TDP-43 accumulation is most abundant. This suggests BSCB leakage and TDP-43 pathology are independent pathologies in ALS.
Collapse
|
26
|
Non-neuronal cells in amyotrophic lateral sclerosis - from pathogenesis to biomarkers. Nat Rev Neurol 2021; 17:333-348. [PMID: 33927394 DOI: 10.1038/s41582-021-00487-8] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2021] [Indexed: 02/04/2023]
Abstract
The prevailing motor neuron-centric view of amyotrophic lateral sclerosis (ALS) pathogenesis could be an important factor in the failure to identify disease-modifying therapy for this neurodegenerative disorder. Non-neuronal cells have crucial homeostatic functions within the CNS and evidence of involvement of these cells in the pathophysiology of several neurodegenerative disorders, including ALS, is accumulating. Microglia and astrocytes, in crosstalk with peripheral immune cells, can exert both neuroprotective and adverse effects, resulting in a highly nuanced range of neuronal and non-neuronal cell interactions. This Review provides an overview of the diverse roles of non-neuronal cells in relation to the pathogenesis of ALS and the emerging potential of non-neuronal cell biomarkers to advance therapeutic development.
Collapse
|
27
|
Srivastava S, Ahmad R, Khare SK. Alzheimer's disease and its treatment by different approaches: A review. Eur J Med Chem 2021; 216:113320. [PMID: 33652356 DOI: 10.1016/j.ejmech.2021.113320] [Citation(s) in RCA: 192] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 02/04/2021] [Accepted: 02/13/2021] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that impairs mental ability development and interrupts neurocognitive function. This neuropathological condition is depicted by neurodegeneration, neural loss, and development of neurofibrillary tangles and Aβ plaques. There is also a greater risk of developing AD at a later age for people with cardiovascular diseases, hypertension and diabetes. In the biomedical sciences, effective treatment for Alzheimer's disease is a severe obstacle. There is no such treatment to cure Alzheimer's disease. The drug present in the market show only symptomatic relief. The cause of Alzheimer's disease is not fully understood and the blood-brain barrier restricts drug efficacy are two main factors that hamper research. Stem cell-based therapy has been seen as an effective, secure, and creative therapeutic solution to overcoming AD because of AD's multifactorial nature and inadequate care. Current developments in nanotechnology often offer possibilities for the delivery of active drug candidates to address certain limitations. The key nanoformulations being tested against AD include polymeric nanoparticles (NP), inorganic NPs and lipid-based NPs. Nano drug delivery systems are promising vehicles for targeting several therapeutic moieties by easing drug molecules' penetration across the CNS and improving their bioavailability. In this review, we focus on the causes of the AD and their treatment by different approaches.
Collapse
Affiliation(s)
- Sukriti Srivastava
- Enzyme and Microbial Biochemistry Laboratory, Department of Chemistry, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Razi Ahmad
- Enzyme and Microbial Biochemistry Laboratory, Department of Chemistry, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Sunil Kumar Khare
- Enzyme and Microbial Biochemistry Laboratory, Department of Chemistry, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India.
| |
Collapse
|
28
|
Human ALS/FTD brain organoid slice cultures display distinct early astrocyte and targetable neuronal pathology. Nat Neurosci 2021; 24:1542-1554. [PMID: 34675437 PMCID: PMC8553627 DOI: 10.1038/s41593-021-00923-4] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 08/16/2021] [Indexed: 12/09/2022]
Abstract
Amyotrophic lateral sclerosis overlapping with frontotemporal dementia (ALS/FTD) is a fatal and currently untreatable disease characterized by rapid cognitive decline and paralysis. Elucidating initial cellular pathologies is central to therapeutic target development, but obtaining samples from presymptomatic patients is not feasible. Here, we report the development of a cerebral organoid slice model derived from human induced pluripotent stem cells (iPSCs) that recapitulates mature cortical architecture and displays early molecular pathology of C9ORF72 ALS/FTD. Using a combination of single-cell RNA sequencing and biological assays, we reveal distinct transcriptional, proteostasis and DNA repair disturbances in astroglia and neurons. We show that astroglia display increased levels of the autophagy signaling protein P62 and that deep layer neurons accumulate dipeptide repeat protein poly(GA), DNA damage and undergo nuclear pyknosis that could be pharmacologically rescued by GSK2606414. Thus, patient-specific iPSC-derived cortical organoid slice cultures are a reproducible translational platform to investigate preclinical ALS/FTD mechanisms as well as novel therapeutic approaches.
Collapse
|
29
|
Crabé R, Aimond F, Gosset P, Scamps F, Raoul C. How Degeneration of Cells Surrounding Motoneurons Contributes to Amyotrophic Lateral Sclerosis. Cells 2020; 9:cells9122550. [PMID: 33260927 PMCID: PMC7760029 DOI: 10.3390/cells9122550] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/19/2020] [Accepted: 11/24/2020] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurological disorder characterized by the progressive degeneration of upper and lower motoneurons. Despite motoneuron death being recognized as the cardinal event of the disease, the loss of glial cells and interneurons in the brain and spinal cord accompanies and even precedes motoneuron elimination. In this review, we provide striking evidence that the degeneration of astrocytes and oligodendrocytes, in addition to inhibitory and modulatory interneurons, disrupt the functionally coherent environment of motoneurons. We discuss the extent to which the degeneration of glial cells and interneurons also contributes to the decline of the motor system. This pathogenic cellular network therefore represents a novel strategic field of therapeutic investigation.
Collapse
Affiliation(s)
- Roxane Crabé
- The Neuroscience Institute of Montpellier, INSERM, UMR1051, University of Montpellier, 34091 Montpellier, France; (R.C.); (F.A.); (P.G.); (F.S.)
| | - Franck Aimond
- The Neuroscience Institute of Montpellier, INSERM, UMR1051, University of Montpellier, 34091 Montpellier, France; (R.C.); (F.A.); (P.G.); (F.S.)
| | - Philippe Gosset
- The Neuroscience Institute of Montpellier, INSERM, UMR1051, University of Montpellier, 34091 Montpellier, France; (R.C.); (F.A.); (P.G.); (F.S.)
| | - Frédérique Scamps
- The Neuroscience Institute of Montpellier, INSERM, UMR1051, University of Montpellier, 34091 Montpellier, France; (R.C.); (F.A.); (P.G.); (F.S.)
| | - Cédric Raoul
- The Neuroscience Institute of Montpellier, INSERM, UMR1051, University of Montpellier, 34091 Montpellier, France; (R.C.); (F.A.); (P.G.); (F.S.)
- Laboratory of Neurobiology, Kazan Federal University, 420008 Kazan, Russia
- Correspondence:
| |
Collapse
|