1
|
Xie Q, Li K, Chen Y, Li Y, Jiang W, Cao W, Yu H, Fan D, Deng B. Gene therapy breakthroughs in ALS: a beacon of hope for 20% of ALS patients. Transl Neurodegener 2025; 14:19. [PMID: 40234983 PMCID: PMC12001736 DOI: 10.1186/s40035-025-00477-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 03/05/2025] [Indexed: 04/17/2025] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal motor neuron disease that remains incurable. Although the etiologies of ALS are diverse and the precise pathogenic mechanisms are not fully understood, approximately 20% of ALS cases are caused by genetic factors. Therefore, advancing targeted gene therapies holds significant promise, at least for the 20% of ALS patients with genetic etiologies. In this review, we summarize the main strategies and techniques of current ALS gene therapies based on ALS risk genes, and review recent findings from animal studies and clinical trials. Additionally, we highlight ALS-related genes with well-understood pathogenic mechanisms and the potential of numerous emerging gene-targeted therapeutic approaches for ALS.
Collapse
Affiliation(s)
- Qingjian Xie
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 32500, China
- First School of Clinical Medicine, Wenzhou Medical University, Wenzhou, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kezheng Li
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 32500, China
- First School of Clinical Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yinuo Chen
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 32500, China
- First School of Clinical Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yaojia Li
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 32500, China
| | - Wenhua Jiang
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 32500, China
| | - Wen Cao
- Department of Neurology, Peking University Third Hospital, Beijing, China
| | - Huan Yu
- Department of Pediatrics, Second Affiliated Hospital and Yuying Children'S Hospital of Wenzhou Medical University, Wenzhou, China
| | - Dongsheng Fan
- Department of Neurology, Peking University Third Hospital, Beijing, China
| | - Binbin Deng
- Department of Rehabilitation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 32500, China.
| |
Collapse
|
2
|
Jiang SS, Nie HB, Hua S, Xie M, Xu RS. Preliminary Analysis of Potentially Overlapping Differentially Expressed Proteins in Both the Spinal Cord and Brain of SOD1 G93A Mice. Curr Protein Pept Sci 2025; 26:57-75. [PMID: 38984582 DOI: 10.2174/0113892037293525240621120033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 05/23/2024] [Accepted: 05/27/2024] [Indexed: 07/11/2024]
Abstract
OBJECTIVE Proteomic elucidation is an essential step in improving our understanding of the biological properties of proteins in amyotrophic lateral sclerosis (ALS). METHODS Preliminary proteomic analysis was performed on the spinal cord and brain of SOD1 G93A (TG) and wild-type (WT) mice using isobaric tags for relative and absolute quantitation. RESULTS Partial up- and downregulated proteins showing significant differences between TG and WT mice were identified, of which 105 proteins overlapped with differentially expressed proteins in both the spinal cord and brain of progression mice. Bioinformatic analyses using Gene Ontology, a cluster of orthologous groups, and Kyoto Encyclopedia of Genes and Genomes pathway revealed that the significantly up- and downregulated proteins represented multiple biological functions closely related to ALS, with 105 overlapping differentially expressed proteins in the spinal cord and brain at the progression stage of TG mice closely related to 122 pathways. Differentially expressed proteins involved in a set of molecular functions play essential roles in maintaining neural cell survival. CONCLUSION This study provides additional proteomic profiles of TG mice, including potential overlapping proteins in both the spinal cord and brain that participate in pathogenesis, as well as novel insights into the up- and downregulation of proteins involved in the pathogenesis of ALS.
Collapse
Affiliation(s)
- Shi-Shi Jiang
- Department of Neurology, Jiangxi Provincial People's Hospital, Clinical College of Nanchang Medical College, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, Jiangxi, China
| | - Hong-Bing Nie
- Department of Neurology, Jiangxi Provincial People's Hospital, Clinical College of Nanchang Medical College, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, Jiangxi, China
| | - Shan Hua
- Department of Ultrasonography, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, Jiangxi, China
| | - Meng Xie
- Health Management Center, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, Jiangxi, China
| | - Ren-Shi Xu
- Department of Neurology, Jiangxi Provincial People's Hospital, Clinical College of Nanchang Medical College, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, Jiangxi, China
| |
Collapse
|
3
|
Ma D, Stocks J, Rosen H, Kantarci K, Lockhart SN, Bateman JR, Craft S, Gurcan MN, Popuri K, Beg MF, Wang L, on behalf of the ALLFTD consortium. Differential diagnosis of frontotemporal dementia subtypes with explainable deep learning on structural MRI. Front Neurosci 2024; 18:1331677. [PMID: 38384484 PMCID: PMC10879283 DOI: 10.3389/fnins.2024.1331677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 01/08/2024] [Indexed: 02/23/2024] Open
Abstract
Background Frontotemporal dementia (FTD) represents a collection of neurobehavioral and neurocognitive syndromes that are associated with a significant degree of clinical, pathological, and genetic heterogeneity. Such heterogeneity hinders the identification of effective biomarkers, preventing effective targeted recruitment of participants in clinical trials for developing potential interventions and treatments. In the present study, we aim to automatically differentiate patients with three clinical phenotypes of FTD, behavioral-variant FTD (bvFTD), semantic variant PPA (svPPA), and nonfluent variant PPA (nfvPPA), based on their structural MRI by training a deep neural network (DNN). Methods Data from 277 FTD patients (173 bvFTD, 63 nfvPPA, and 41 svPPA) recruited from two multi-site neuroimaging datasets: the Frontotemporal Lobar Degeneration Neuroimaging Initiative and the ARTFL-LEFFTDS Longitudinal Frontotemporal Lobar Degeneration databases. Raw T1-weighted MRI data were preprocessed and parcellated into patch-based ROIs, with cortical thickness and volume features extracted and harmonized to control the confounding effects of sex, age, total intracranial volume, cohort, and scanner difference. A multi-type parallel feature embedding framework was trained to classify three FTD subtypes with a weighted cross-entropy loss function used to account for unbalanced sample sizes. Feature visualization was achieved through post-hoc analysis using an integrated gradient approach. Results The proposed differential diagnosis framework achieved a mean balanced accuracy of 0.80 for bvFTD, 0.82 for nfvPPA, 0.89 for svPPA, and an overall balanced accuracy of 0.84. Feature importance maps showed more localized differential patterns among different FTD subtypes compared to groupwise statistical mapping. Conclusion In this study, we demonstrated the efficiency and effectiveness of using explainable deep-learning-based parallel feature embedding and visualization framework on MRI-derived multi-type structural patterns to differentiate three clinically defined subphenotypes of FTD: bvFTD, nfvPPA, and svPPA, which could help with the identification of at-risk populations for early and precise diagnosis for intervention planning.
Collapse
Affiliation(s)
- Da Ma
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Jane Stocks
- Department of Psychiatry and Behavioral Health, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Howard Rosen
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, United States
| | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN, United States
| | - Samuel N. Lockhart
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - James R. Bateman
- Department of Neurology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Suzanne Craft
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Metin N. Gurcan
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Karteek Popuri
- Department of Computer Science, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Mirza Faisal Beg
- School of Engineering Science, Simon Fraser University, Burnaby, BC, Canada
| | - Lei Wang
- Department of Psychiatry and Behavioral Health, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Department of Psychiatry and Behavioral Health, Ohio State University Wexner Medical Center, Columbus, OH, United States
| | | |
Collapse
|
4
|
Genin EC, Abou-Ali M, Paquis-Flucklinger V. Mitochondria, a Key Target in Amyotrophic Lateral Sclerosis Pathogenesis. Genes (Basel) 2023; 14:1981. [PMID: 38002924 PMCID: PMC10671245 DOI: 10.3390/genes14111981] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/19/2023] [Accepted: 10/21/2023] [Indexed: 11/26/2023] Open
Abstract
Mitochondrial dysfunction occurs in numerous neurodegenerative diseases, particularly amyotrophic lateral sclerosis (ALS), where it contributes to motor neuron (MN) death. Of all the factors involved in ALS, mitochondria have been considered as a major player, as secondary mitochondrial dysfunction has been found in various models and patients. Abnormal mitochondrial morphology, defects in mitochondrial dynamics, altered activities of respiratory chain enzymes and increased production of reactive oxygen species have been described. Moreover, the identification of CHCHD10 variants in ALS patients was the first genetic evidence that a mitochondrial defect may be a primary cause of MN damage and directly links mitochondrial dysfunction to the pathogenesis of ALS. In this review, we focus on the role of mitochondria in ALS and highlight the pathogenic variants of ALS genes associated with impaired mitochondrial functions. The multiple pathways demonstrated in ALS pathogenesis suggest that all converge to a common endpoint leading to MN loss. This may explain the disappointing results obtained with treatments targeting a single pathological process. Fighting against mitochondrial dysfunction appears to be a promising avenue for developing combined therapies in the future.
Collapse
Affiliation(s)
- Emmanuelle C. Genin
- Institute for Research on Cancer and Aging, Nice (IRCAN), Université Côte d’Azur, Inserm U1081, CNRS UMR7284, Centre Hospitalier Universitaire (CHU) de Nice, 06200 Nice, France; (M.A.-A.); (V.P.-F.)
| | | | | |
Collapse
|
5
|
Cheng A, Liu C, Ye W, Huang D, She W, Liu X, Fung CP, Xu N, Suen MC, Ye W, Sung HHY, Williams ID, Zhu G, Qian PY. Selective C9orf72 G-Quadruplex-Binding Small Molecules Ameliorate Pathological Signatures of ALS/FTD Models. J Med Chem 2022; 65:12825-12837. [PMID: 36226410 PMCID: PMC9574859 DOI: 10.1021/acs.jmedchem.2c00654] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
The G-quadruplex (G4) forming C9orf72 GGGGCC (G4C2) expanded hexanucleotide repeat (EHR)
is the predominant genetic cause of amyotrophic lateral sclerosis
(ALS) and frontotemporal dementia (FTD). Developing selective G4-binding
ligands is challenging due to the conformational polymorphism and
similarity of G4 structures. We identified three first-in-class marine
natural products, chrexanthomycin A (cA), chrexanthomycin
B (cB), and chrexanthomycin C (cC), with
remarkable bioactivities. Thereinto, cA shows the highest
permeability and lowest cytotoxicity to live cells. NMR titration
experiments and in silico analysis demonstrate that cA, cB, and cC selectively bind
to DNA and RNA G4C2 G4s. Notably, cA and cC dramatically reduce G4C2 EHR-caused cell death, diminish G4C2 RNA
foci in (G4C2)29-expressing Neuro2a cells, and significantly
eliminate ROS in HT22 cells. In (G4C2)29-expressing Drosophila, cA and cC significantly
rescue eye degeneration and improve locomotor deficits. Overall, our
findings reveal that cA and cC are potential
therapeutic agents deserving further clinical study.
Collapse
Affiliation(s)
- Aifang Cheng
- Department of Ocean Science and Hong Kong Branch of Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), The Hong Kong University of Science and Technology, Hong Kong 999077, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China
| | - Changdong Liu
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong 999077, China
- Shenzhen Research Institute, The Hong Kong University of Science and Technology, Hi-Tech Park, Nanshan, Shenzhen 518057, China
| | - Wenkang Ye
- Department of Ocean Science and Hong Kong Branch of Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), The Hong Kong University of Science and Technology, Hong Kong 999077, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China
- SZU-HKUST Joint Ph.D. Program in Marine Environmental Science, Shenzhen University, Shenzhen 518060, China
| | - Duli Huang
- Department of Ocean Science and Hong Kong Branch of Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), The Hong Kong University of Science and Technology, Hong Kong 999077, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China
| | - Weiyi She
- Department of Ocean Science and Hong Kong Branch of Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), The Hong Kong University of Science and Technology, Hong Kong 999077, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China
- SZU-HKUST Joint Ph.D. Program in Marine Environmental Science, Shenzhen University, Shenzhen 518060, China
| | - Xin Liu
- Department of Ocean Science and Hong Kong Branch of Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), The Hong Kong University of Science and Technology, Hong Kong 999077, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China
| | - Chun Po Fung
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong 999077, China
| | - Naining Xu
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong 999077, China
- Department of Oral and Maxillofacial Surgery, Stomatological Center, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
| | - Monica Ching Suen
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong 999077, China
| | - Wei Ye
- Department of Ocean Science and Hong Kong Branch of Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), The Hong Kong University of Science and Technology, Hong Kong 999077, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China
| | - Herman Ho Yung Sung
- Department of Chemistry, The Hong Kong University of Science and Technology, Hong Kong 999077, China
| | - Ian Duncan Williams
- Department of Chemistry, The Hong Kong University of Science and Technology, Hong Kong 999077, China
| | - Guang Zhu
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong 999077, China
- Shenzhen Research Institute, The Hong Kong University of Science and Technology, Hi-Tech Park, Nanshan, Shenzhen 518057, China
| | - Pei-Yuan Qian
- Department of Ocean Science and Hong Kong Branch of Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), The Hong Kong University of Science and Technology, Hong Kong 999077, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China
| |
Collapse
|
6
|
Synucleinopathy in Amyotrophic Lateral Sclerosis: A Potential Avenue for Antisense Therapeutics? Int J Mol Sci 2022; 23:ijms23169364. [PMID: 36012622 PMCID: PMC9409035 DOI: 10.3390/ijms23169364] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 01/02/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is the most common adult-onset motor neuron disease classified as both a neurodegenerative and neuromuscular disorder. With a complex aetiology and no current cure for ALS, broadening the understanding of disease pathology and therapeutic avenues is required to progress with patient care. Alpha-synuclein (αSyn) is a hallmark for disease in neurodegenerative disorders, such as Parkinson's disease, Lewy body dementia, and multiple system atrophy. A growing body of evidence now suggests that αSyn may also play a pathological role in ALS, with αSyn-positive Lewy bodies co-aggregating alongside known ALS pathogenic proteins, such as SOD1 and TDP-43. This review endeavours to capture the scope of literature regarding the aetiology and development of ALS and its commonalities with "synucleinopathy disorders". We will discuss the involvement of αSyn in ALS and motor neuron disease pathology, and the current theories and strategies for therapeutics in ALS treatment, as well as those targeting αSyn for synucleinopathies, with a core focus on small molecule RNA technologies.
Collapse
|
7
|
Kirola L, Mukherjee A, Mutsuddi M. Recent Updates on the Genetics of Amyotrophic Lateral Sclerosis and Frontotemporal Dementia. Mol Neurobiol 2022; 59:5673-5694. [PMID: 35768750 DOI: 10.1007/s12035-022-02934-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/16/2022] [Indexed: 10/17/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) primarily affect the motor and frontotemporal areas of the brain, respectively. These disorders share clinical, genetic, and pathological similarities, and approximately 10-15% of ALS-FTD cases are considered to be multisystemic. ALS-FTD overlaps have been linked to families carrying an expansion in the intron of C9orf72 along with inclusions of TDP-43 in the brain. Other overlapping genes (VCP, FUS, SQSTM1, TBK1, CHCHD10) are also involved in similar functions that include RNA processing, autophagy, proteasome response, protein aggregation, and intracellular trafficking. Recent advances in genome sequencing have identified new genes that are involved in these disorders (TBK1, CCNF, GLT8D1, KIF5A, NEK1, C21orf2, TBP, CTSF, MFSD8, DNAJC7). Additional risk factors and modifiers have been also identified in genome-wide association studies and array-based studies. However, the newly identified genes show higher disease frequencies in combination with known genes that are implicated in pathogenesis, thus indicating probable digenetic/polygenic inheritance models, along with epistatic interactions. Studies suggest that these genes play a pleiotropic effect on ALS-FTD and other diseases such as Alzheimer's disease, Ataxia, and Parkinsonism. Besides, there have been numerous improvements in the genotype-phenotype correlations as well as clinical trials on stem cell and gene-based therapies. This review discusses the possible genetic models of ALS and FTD, the latest therapeutics, and signaling pathways involved in ALS-FTD.
Collapse
Affiliation(s)
- Laxmi Kirola
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Ashim Mukherjee
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Mousumi Mutsuddi
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India.
| |
Collapse
|
8
|
Berthold N, Pytte J, Bulik CM, Tschochner M, Medland SE, Akkari PA. Bridging the gap: Short structural variants in the genetics of anorexia nervosa. Int J Eat Disord 2022; 55:747-753. [PMID: 35470453 PMCID: PMC9545787 DOI: 10.1002/eat.23716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 11/07/2022]
Abstract
Anorexia nervosa (AN) is a devastating disorder with evidence of underexplored heritability. Twin and family studies estimate heritability (h2 ) to be 57%-64%, and genome-wide association studies (GWAS) reveal significant genetic correlations with psychiatric and anthropometric traits and a total of nine genome-wide significant loci. Whether significantly associated single nucleotide polymorphisms identified by GWAS are causal or tag true causal variants, remains to be elucidated. We propose a novel method for bridging this knowledge gap by fine-mapping short structural variants (SSVs) in and around GWAS-identified loci. SSV fine-mapping of loci associated with complex disorders such as schizophrenia, amyotrophic lateral sclerosis, and Alzheimer's disease has uncovered genetic risk markers, phenotypic variability between patients, new pathological mechanisms, and potential therapeutic targets. We analyze previous investigations' methods and propose utilizing an evaluation algorithm to prioritize 10 SSVs for each of the top two AN GWAS-identified loci followed by Sanger sequencing and fragment analysis via capillary electrophoresis to characterize these SSVs for case/control association studies. Success of previous SSV analyses in complex disorders and effective utilization of similar methodologies supports our proposed method. Furthermore, the structural and spatial properties of the 10 SSVs identified for each of the top two AN GWAS-associated loci, cell adhesion molecule 1 (CADM1) and NCK interacting protein with SH3 domain (NCKIPSD), are similar to previous studies. We propose SSV fine-mapping of AN-associated loci will identify causal genetic architecture. Deepening understandings of AN may lead to novel therapeutic targets and subsequently increase quality-of-life for individuals living with the illness. PUBLIC SIGNIFICANCE STATEMENT: Anorexia nervosa is a severe and complex illness, arising from a combination of environmental and genetic factors. Recent studies estimate the contribution of genetic variability; however, the specific DNA sequences and how they contribute remain unknown. We present a novel approach, arguing that the genetic variant class, short structural variants, could answer this knowledge gap and allow development of biologically targeted therapeutics, improving quality-of-life and patient outcomes for affected individuals.
Collapse
Affiliation(s)
- Natasha Berthold
- School of Nursing, Midwifery, Health Sciences & PhysiotherapyUniversity of Notre Dame AustraliaFremantleWestern AustraliaAustralia
- Perron Institute for Neurological and Translational ScienceNedlandsWestern AustraliaAustralia
- School of Human Sciences, University of Western AustraliaCrawleyWestern AustraliaAustralia
| | - Julia Pytte
- Perron Institute for Neurological and Translational ScienceNedlandsWestern AustraliaAustralia
- School of Human Sciences, University of Western AustraliaCrawleyWestern AustraliaAustralia
| | - Cynthia M. Bulik
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
- Department of PsychiatryUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Department of NutritionUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Monika Tschochner
- School of Nursing, Midwifery, Health Sciences & PhysiotherapyUniversity of Notre Dame AustraliaFremantleWestern AustraliaAustralia
| | - Sarah E. Medland
- QIMR Berghofer Medical Research InstituteBrisbaneQueenslandAustralia
| | - Patrick Anthony Akkari
- Perron Institute for Neurological and Translational ScienceNedlandsWestern AustraliaAustralia
- Centre for Molecular Medicine and Innovative TherapeuticsMurdoch UniversityPerthWestern AustraliaAustralia
- Centre for Neuromuscular and Neurological DisordersUniversity of Western AustraliaNedlandsWestern AustraliaAustralia
- Department of NeurologyDuke UniversityDurhamNorth Carolina
| |
Collapse
|
9
|
Xu Z, Jiang J, Xu S, Xie Z, He P, Jiang S, Xu R. Nerve Growth Factor is a Potential Treated Target in Tg(SOD1*G93A)1Gur Mice. Cell Mol Neurobiol 2022; 42:1035-1046. [PMID: 33236288 PMCID: PMC11441269 DOI: 10.1007/s10571-020-00993-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/28/2020] [Indexed: 12/11/2022]
Abstract
Nerve growth factor (NGF) is a protective factor of neural cells; the possible relationship between the NGF and the pathogenesis of amyotrophic lateral sclerosis (ALS) hasn't been completely known. In this study, we observed and analyzed the expression and distribution of NGF, as well as the possible relationship between the NGF expression and distribution and the neural cell death in both SOD1 wild-type (WT) and Tg(SOD1*G93A)1Gur (TG) mice applying the fluorescence immunohistochemistry method. The results showed that the expression and distribution of NGF in the anterior horn (AH), the lateral horn (LH), and the surrounding central canal (CC) significantly increased at the supper early stage of ALS (Pre-onset stage) and the early stage (Onset stage), but the NGF expression and distribution in the AH, the LH, and the surrounding CC significantly reduced at the progression stage. The astrocyte, neuron, and oligodendrocyte produced the NGF and the neural precursor cells (NPCs) produced the NGF. The neural cell death gradually increased accompanying with the reduction of NGF expression and distribution. Our data suggested that the NGF was a protective factor of neural cells, because the neural cells in the AH, the LH, and the surrounding CC produced more NGF at the supper early and early stage of ALS; moreover, the NPCs produced the NGF. It implied that the NGF exerted the protective effect of neural cells, prevented from the neural cell death and aroused the potential of self-repair in the development of ALS.
Collapse
Affiliation(s)
- Zhenzhen Xu
- Department of Neurology, Jiangxi Provincial People's Hospital, Affiliated People's Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- Department of Neurology, First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Jianxiang Jiang
- Department of Neurology, Jiangxi Provincial People's Hospital, Affiliated People's Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Shengyuan Xu
- Department of Neurology, Jiangxi Provincial People's Hospital, Affiliated People's Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Zunchun Xie
- Department of Neurology, First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Pei He
- Department of Neurology, Jiangxi Provincial People's Hospital, Affiliated People's Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Shishi Jiang
- Department of Neurology, Jiangxi Provincial People's Hospital, Affiliated People's Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Renshi Xu
- Department of Neurology, Jiangxi Provincial People's Hospital, Affiliated People's Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
10
|
Ursu A, Baisden JT, Bush JA, Taghavi A, Choudhary S, Zhang YJ, Gendron TF, Petrucelli L, Yildirim I, Disney MD. A Small Molecule Exploits Hidden Structural Features within the RNA Repeat Expansion That Causes c9ALS/FTD and Rescues Pathological Hallmarks. ACS Chem Neurosci 2021; 12:4076-4089. [PMID: 34677935 DOI: 10.1021/acschemneuro.1c00470] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The hexanucleotide repeat expansion GGGGCC [r(G4C2)exp] within intron 1 of C9orf72 causes genetically defined amyotrophic lateral sclerosis and frontotemporal dementia, collectively named c9ALS/FTD. , the repeat expansion causes neurodegeneration via deleterious phenotypes stemming from r(G4C2)exp RNA gain- and loss-of-function mechanisms. The r(G4C2)exp RNA folds into both a hairpin structure with repeating 1 × 1 nucleotide GG internal loops and a G-quadruplex structure. Here, we report the identification of a small molecule (CB253) that selectively binds the hairpin form of r(G4C2)exp. Interestingly, the small molecule binds to a previously unobserved conformation in which the RNA forms 2 × 2 nucleotide GG internal loops, as revealed by a series of binding and structural studies. NMR and molecular dynamics simulations suggest that the r(G4C2)exp hairpin interconverts between 1 × 1 and 2 × 2 internal loops through the process of strand slippage. We provide experimental evidence that CB253 binding indeed shifts the equilibrium toward the 2 × 2 GG internal loop conformation, inhibiting mechanisms that drive c9ALS/FTD pathobiology, such as repeat-associated non-ATG translation formation of stress granules and defective nucleocytoplasmic transport in various cellular models of c9ALS/FTD.
Collapse
Affiliation(s)
- Andrei Ursu
- Department of Chemistry, The Scripps Research Institute, 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Jared T. Baisden
- Department of Chemistry, The Scripps Research Institute, 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Jessica A. Bush
- Department of Chemistry, The Scripps Research Institute, 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Amirhossein Taghavi
- Department of Chemistry and Biochemistry, Florida Atlantic University, Jupiter, Florida 33458, United States
| | - Shruti Choudhary
- Department of Chemistry, The Scripps Research Institute, 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Yong-Jie Zhang
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Rd., Jacksonville, Florida 32224, United States
| | - Tania F. Gendron
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Rd., Jacksonville, Florida 32224, United States
| | - Leonard Petrucelli
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Rd., Jacksonville, Florida 32224, United States
| | - Ilyas Yildirim
- Department of Chemistry and Biochemistry, Florida Atlantic University, Jupiter, Florida 33458, United States
| | - Matthew D. Disney
- Department of Chemistry, The Scripps Research Institute, 130 Scripps Way, Jupiter, Florida 33458, United States
| |
Collapse
|
11
|
Theunissen F, Anderton RS, Mastaglia FL, Flynn LL, Winter SJ, James I, Bedlack R, Hodgetts S, Fletcher S, Wilton SD, Laing NG, MacShane M, Needham M, Saunders A, Mackay-Sim A, Melamed Z, Ravits J, Cleveland DW, Akkari PA. Novel STMN2 Variant Linked to Amyotrophic Lateral Sclerosis Risk and Clinical Phenotype. Front Aging Neurosci 2021; 13:658226. [PMID: 33841129 PMCID: PMC8033025 DOI: 10.3389/fnagi.2021.658226] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/02/2021] [Indexed: 12/19/2022] Open
Abstract
Objective There is a critical need to establish genetic markers that explain the complex phenotypes and pathogenicity of ALS. This study identified a polymorphism in the Stathmin-2 gene and investigated its association with sporadic ALS (sALS) disease risk, age-of onset and survival duration. Methods The candidate CA repeat was systematically analyzed using PCR, Sanger sequencing and high throughput capillary separation for genotyping. Stathmin-2 expression was investigated using RT-PCR in patient olfactory neurosphere-derived (ONS) cells and RNA sequencing in laser-captured spinal motor neurons. Results In a case-control analysis of a combined North American sALS cohort (n = 321) and population control group (n = 332), long/long CA genotypes were significantly associated with disease risk (p = 0.042), and most strongly when one allele was a 24 CA repeat (p = 0.0023). In addition, longer CA allele length was associated with earlier age-of-onset (p = 0.039), and shorter survival duration in bulbar-onset cases (p = 0.006). In an Australian longitudinal sALS cohort (n = 67), ALS functional rating scale scores were significantly lower in carriers of the long/long genotype (p = 0.034). Stathmin-2 mRNA expression was reduced in sporadic patient ONS cells. Additionally, sALS patients and controls exhibited variable expression of Stathmin-2 mRNA according to CA genotype in laser-captured spinal motor neurons. Conclusions We report a novel non-coding CA repeat in Stathmin-2 which is associated with sALS disease risk and has disease modifying effects. The potential value of this variant as a disease marker and tool for cohort enrichment in clinical trials warrants further investigation.
Collapse
Affiliation(s)
- Frances Theunissen
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
| | - Ryan S Anderton
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, University of Western Australia, Nedlands, WA, Australia.,School of Health Sciences, Institute for Health Research, The University of Notre Dame Australia, Fremantle, WA, Australia
| | - Frank L Mastaglia
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, University of Western Australia, Nedlands, WA, Australia
| | - Loren L Flynn
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, University of Western Australia, Nedlands, WA, Australia
| | - Samantha J Winter
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,School of Health Sciences, Institute for Health Research, The University of Notre Dame Australia, Fremantle, WA, Australia
| | - Ian James
- Institute for Immunology and Infectious Disease, Murdoch University, Perth, WA, Australia
| | - Richard Bedlack
- Department of Neurology, Duke University, Durham, NC, United States
| | - Stuart Hodgetts
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,School of Human Sciences, University of Western Australia, Nedlands, WA, Australia
| | - Sue Fletcher
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, University of Western Australia, Nedlands, WA, Australia
| | - Steve D Wilton
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, University of Western Australia, Nedlands, WA, Australia
| | - Nigel G Laing
- Centre for Medical Research, Harry Perkins Institute of Medical Research, The University of Western Australia, Perth, WA, Australia
| | - Mandi MacShane
- Centre for Medical Research, Harry Perkins Institute of Medical Research, The University of Western Australia, Perth, WA, Australia
| | - Merrilee Needham
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia.,Faculty of Medicine, The University of Notre Dame Australia, Fremantle, WA, Australia.,Department of Neurology, Fiona Stanley Hospital, Murdoch, WA, Australia
| | - Ann Saunders
- Zinfandel Pharmaceuticals, Chapel Hill, NC, United States
| | - Alan Mackay-Sim
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| | - Ze'ev Melamed
- Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA, United States.,Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, United States
| | - John Ravits
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Don W Cleveland
- Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA, United States.,Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, United States.,Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States
| | - P Anthony Akkari
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, University of Western Australia, Nedlands, WA, Australia.,Department of Neurology, Duke University, Durham, NC, United States
| |
Collapse
|
12
|
Beckers J, Tharkeshwar AK, Van Damme P. C9orf72 ALS-FTD: recent evidence for dysregulation of the autophagy-lysosome pathway at multiple levels. Autophagy 2021; 17:3306-3322. [PMID: 33632058 PMCID: PMC8632097 DOI: 10.1080/15548627.2021.1872189] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are two clinically distinct classes of neurodegenerative disorders. Yet, they share a range of genetic, cellular, and molecular features. Hexanucleotide repeat expansions (HREs) in the C9orf72 gene and the accumulation of toxic protein aggregates in the nervous systems of the affected individuals are among such common features. Though the mechanisms by which HREs cause toxicity is not clear, the toxic gain of function due to transcribed HRE RNA or dipeptide repeat proteins (DPRs) produced by repeat-associated non-AUG translation together with a reduction in C9orf72 expression are proposed as the contributing factors for disease pathogenesis in ALS and FTD. In addition, several recent studies point toward alterations in protein homeostasis as one of the root causes of the disease pathogenesis. In this review, we discuss the effects of the C9orf72 HRE in the autophagy-lysosome pathway based on various recent findings. We suggest that dysfunction of the autophagy-lysosome pathway synergizes with toxicity from C9orf72 repeat RNA and DPRs to drive disease pathogenesis. Abbreviation: ALP: autophagy-lysosome pathway; ALS: amyotrophic lateral sclerosis; AMPK: AMP-activated protein kinase; ATG: autophagy-related; ASO: antisense oligonucleotide; C9orf72: C9orf72-SMCR8 complex subunit; DENN: differentially expressed in normal and neoplastic cells; DPR: dipeptide repeat protein; EIF2A/eIF2α: eukaryotic translation initiation factor 2A; ER: endoplasmic reticulum; FTD: frontotemporal dementia; GAP: GTPase-activating protein; GEF: guanine nucleotide exchange factor; HRE: hexanucleotide repeat expansion; iPSC: induced pluripotent stem cell; ISR: integrated stress response; M6PR: mannose-6-phosphate receptor, cation dependent; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MN: motor neuron; MTORC1: mechanistic target of rapamycin kinase complex 1; ND: neurodegenerative disorder; RAN: repeat-associated non-ATG; RB1CC1/FIP200: RB1 inducible coiled-coil 1; SLC66A1/PQLC2: solute carrier family 66 member 1; SMCR8: SMCR8-C9orf72 complex subunit; SQSTM1/p62: sequestosome 1; STX17: syntaxin 17; TARDBP/TDP-43: TAR DNA binding protein; TBK1: TANK binding kinase 1; TFEB: transcription factor EB; ULK1: unc-51 like autophagy activating kinase 1; UPS: ubiquitin-proteasome system; WDR41: WD repeat domain 41.
Collapse
Affiliation(s)
- Jimmy Beckers
- Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium.,VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | - Arun Kumar Tharkeshwar
- Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium.,VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | - Philip Van Damme
- Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium.,VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium.,University Hospitals Leuven, Department of Neurology, Leuven, Belgium
| |
Collapse
|
13
|
Ursu A, Wang KW, Bush JA, Choudhary S, Chen JL, Baisden JT, Zhang YJ, Gendron TF, Petrucelli L, Yildirim I, Disney MD. Structural Features of Small Molecules Targeting the RNA Repeat Expansion That Causes Genetically Defined ALS/FTD. ACS Chem Biol 2020; 15:3112-3123. [PMID: 33196168 DOI: 10.1021/acschembio.0c00049] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Genetically defined amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), collectively named c9ALS/FTD, are triggered by hexanucleotide GGGGCC repeat expansions [r(G4C2)exp] within the C9orf72 gene. In these diseases, neuronal loss occurs through an interplay of deleterious phenotypes, including r(G4C2)exp RNA gain-of-function mechanisms. Herein, we identified a benzimidazole derivative, CB096, that specifically binds to a repeating 1 × 1 GG internal loop structure, 5'CGG/3'GGC, that is formed when r(G4C2)exp folds. Structure-activity relationship (SAR) studies and molecular dynamics (MD) simulations were used to define the molecular interactions formed between CB096 and r(G4C2)exp that results in the rescue of disease-associated pathways. Overall, this study reveals a unique structural feature within r(G4C2)exp that can be exploited for the development of lead medicines and chemical probes.
Collapse
Affiliation(s)
- Andrei Ursu
- Department of Chemistry, The Scripps Research Institute, 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Kye Won Wang
- Department of Chemistry and Biochemistry, Florida Atlantic University, Jupiter, Florida 33458, United States
| | - Jessica A. Bush
- Department of Chemistry, The Scripps Research Institute, 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Shruti Choudhary
- Department of Chemistry, The Scripps Research Institute, 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Jonathan L. Chen
- Department of Chemistry, The Scripps Research Institute, 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Jared T. Baisden
- Department of Chemistry, The Scripps Research Institute, 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Yong-Jie Zhang
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Rd., Jacksonville, Florida 32224, United States
| | - Tania F. Gendron
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Rd., Jacksonville, Florida 32224, United States
| | - Leonard Petrucelli
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Rd., Jacksonville, Florida 32224, United States
| | - Ilyas Yildirim
- Department of Chemistry and Biochemistry, Florida Atlantic University, Jupiter, Florida 33458, United States
| | - Matthew D. Disney
- Department of Chemistry, The Scripps Research Institute, 130 Scripps Way, Jupiter, Florida 33458, United States
| |
Collapse
|
14
|
Current Status and Challenges Associated with CNS-Targeted Gene Delivery across the BBB. Pharmaceutics 2020; 12:pharmaceutics12121216. [PMID: 33334049 PMCID: PMC7765480 DOI: 10.3390/pharmaceutics12121216] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/19/2020] [Accepted: 12/11/2020] [Indexed: 12/21/2022] Open
Abstract
The era of the aging society has arrived, and this is accompanied by an increase in the absolute numbers of patients with neurological disorders, such as Alzheimer’s disease (AD) and Parkinson’s disease (PD). Such neurological disorders are serious costly diseases that have a significant impact on society, both globally and socially. Gene therapy has great promise for the treatment of neurological disorders, but only a few gene therapy drugs are currently available. Delivery to the brain is the biggest hurdle in developing new drugs for the central nervous system (CNS) diseases and this is especially true in the case of gene delivery. Nanotechnologies such as viral and non-viral vectors allow efficient brain-targeted gene delivery systems to be created. The purpose of this review is to provide a comprehensive review of the current status of the development of successful drug delivery to the CNS for the treatment of CNS-related disorders especially by gene therapy. We mainly address three aspects of this situation: (1) blood-brain barrier (BBB) functions; (2) adeno-associated viral (AAV) vectors, currently the most advanced gene delivery vector; (3) non-viral brain targeting by non-invasive methods.
Collapse
|
15
|
Yang Q, Jiao B, Shen L. The Development of C9orf72-Related Amyotrophic Lateral Sclerosis and Frontotemporal Dementia Disorders. Front Genet 2020; 11:562758. [PMID: 32983232 PMCID: PMC7492664 DOI: 10.3389/fgene.2020.562758] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 08/12/2020] [Indexed: 12/12/2022] Open
Abstract
The expanded GGGGCC hexanucleotide repeat in the non-coding region of the C9orf72 gene is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). There are three main disease mechanisms: loss of function of C9ORF72 protein, gain of function from the accumulation of sense and antisense (GGGGCC)n in RNA, and from the production of toxic dipeptides repeat proteins (DPRs) by non-AUG initiated translation. While many of the downstream mechanisms have been identified, the specific pathogenic pathway is still unclear. In this article, we provide an overview on the currently available literature and propose several hypotheses: (1) The pathogenesis of C9orf72-associated ALS/FTD, which cannot be explained by a single mechanism, involves a dual mechanism of both loss and gain of function. (2) The loss of function and gain of function can cause TDP-43 aggregation and damage nucleocytoplasmic transport. (3) Neurodegeneration can be caused by an accumulation of toxic substances in neurons themselves. In addition, we suggest that microglia may cause neurodegeneration by releasing inflammatory factors to neurons. Finally, we summarize several of the most promising treatment strategies.
Collapse
Affiliation(s)
- Qijie Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Bin Jiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
| | - Lu Shen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
| |
Collapse
|
16
|
Liscic RM, Alberici A, Cairns NJ, Romano M, Buratti E. From basic research to the clinic: innovative therapies for ALS and FTD in the pipeline. Mol Neurodegener 2020; 15:31. [PMID: 32487123 PMCID: PMC7268618 DOI: 10.1186/s13024-020-00373-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 03/27/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and Frontotemporal Degeneration (FTD) are neurodegenerative disorders, related by deterioration of motor and cognitive functions and short survival. Aside from cases with an inherited pathogenic mutation, the causes of the disorders are still largely unknown and no effective treatment currently exists. It has been shown that FTD may coexist with ALS and this overlap occurs at clinical, genetic, and molecular levels. In this work, we review the main pathological aspects of these complex diseases and discuss how the integration of the novel pathogenic molecular insights and the analysis of molecular interaction networks among all the genetic players represents a critical step to shed light on discovering novel therapeutic strategies and possibly tailoring personalized medicine approaches to specific ALS and FTD patients.
Collapse
Affiliation(s)
- Rajka Maria Liscic
- Department of Neurology, Johannes Kepler University, Linz, Austria
- School of Medicine, University of Osijek, Osijek, Croatia
| | - Antonella Alberici
- Neurology Unit, Department of Neurological Sciences and Vision, ASST-Spedali Civili-University of Brescia, Brescia, Italy
| | - Nigel John Cairns
- College of Medicine and Health and Living Systems Institute, University of Exeter, Exeter, UK
| | - Maurizio Romano
- Department of Life Sciences, Via Valerio 28, University of Trieste, 34127, Trieste, Italy
| | - Emanuele Buratti
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano 99, 34149, Trieste, Italy.
| |
Collapse
|
17
|
Theunissen F, Flynn LL, Anderton RS, Mastaglia F, Pytte J, Jiang L, Hodgetts S, Burns DK, Saunders A, Fletcher S, Wilton SD, Akkari PA. Structural Variants May Be a Source of Missing Heritability in sALS. Front Neurosci 2020; 14:47. [PMID: 32082115 PMCID: PMC7005198 DOI: 10.3389/fnins.2020.00047] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 01/13/2020] [Indexed: 12/11/2022] Open
Abstract
The underlying genetic and molecular mechanisms that drive amyotrophic lateral sclerosis (ALS) remain poorly understood. Structural variants within the genome can play a significant role in neurodegenerative disease risk, such as the repeat expansion in C9orf72 and the tri-nucleotide repeat in ATXN2, both of which are associated with familial and sporadic ALS. Many such structural variants reside in uncharacterized regions of the human genome, and have been under studied. Therefore, characterization of structural variants located in and around genes associated with ALS could provide insight into disease pathogenesis, and lead to the discovery of highly informative genetic tools for stratification in clinical trials. Such genomic variants may provide a deeper understanding of how gene expression can affect disease etiology, disease severity and trajectory, patient response to treatment, and may hold the key to understanding the genetics of sporadic ALS. This article outlines the current understanding of amyotrophic lateral sclerosis genetics and how structural variations may underpin some of the missing heritability of this disease.
Collapse
Affiliation(s)
- Frances Theunissen
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,School of Human Sciences, University of Western Australia, Nedlands, WA, Australia
| | - Loren L Flynn
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, University of Western Australia, Nedlands, WA, Australia.,Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
| | - Ryan S Anderton
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, University of Western Australia, Nedlands, WA, Australia.,School of Health Sciences, Institute for Health Research, University of Notre Dame Australia, Fremantle, WA, Australia
| | - Frank Mastaglia
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, University of Western Australia, Nedlands, WA, Australia
| | - Julia Pytte
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,School of Human Sciences, University of Western Australia, Nedlands, WA, Australia
| | - Leanne Jiang
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,School of Biological Sciences, University of Western Australia, Nedlands, WA, Australia
| | - Stuart Hodgetts
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,School of Human Sciences, University of Western Australia, Nedlands, WA, Australia
| | - Daniel K Burns
- Zinfandel Pharmaceuticals, Chapel Hill, NC, United States
| | - Ann Saunders
- Zinfandel Pharmaceuticals, Chapel Hill, NC, United States
| | - Sue Fletcher
- Centre for Neuromuscular and Neurological Disorders, University of Western Australia, Nedlands, WA, Australia.,Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
| | - Steve D Wilton
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, University of Western Australia, Nedlands, WA, Australia.,Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
| | - Patrick Anthony Akkari
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, University of Western Australia, Nedlands, WA, Australia.,Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
| |
Collapse
|
18
|
Querin G, Bede P, El Mendili MM, Li M, Pélégrini-Issac M, Rinaldi D, Catala M, Saracino D, Salachas F, Camuzat A, Marchand-Pauvert V, Cohen-Adad J, Colliot O, Le Ber I, Pradat PF. Presymptomatic spinal cord pathology in c9orf72 mutation carriers: A longitudinal neuroimaging study. Ann Neurol 2019; 86:158-167. [PMID: 31177556 DOI: 10.1002/ana.25520] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 06/03/2019] [Accepted: 06/06/2019] [Indexed: 12/11/2022]
Abstract
OBJECTIVE C9orf72 hexanucleotide repeats expansions account for almost half of familial amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) cases. Recent imaging studies in asymptomatic C9orf72 carriers have demonstrated cerebral white (WM) and gray matter (GM) degeneration before the age of 40 years. The objective of this study was to characterize cervical spinal cord (SC) changes in asymptomatic C9orf72 hexanucleotide carriers. METHODS Seventy-two asymptomatic individuals were enrolled in a prospective study of first-degree relatives of ALS and FTD patients carrying the c9orf72 hexanucleotide expansion. Forty of them carried the pathogenic mutation (C9+ ). Each subject underwent quantitative cervical cord imaging. Structural GM and WM metrics and diffusivity parameters were evaluated at baseline and 18 months later. Data were analyzed in C9+ and C9- subgroups, and C9+ subjects were further stratified by age. RESULTS At baseline, significant WM atrophy was detected at each cervical vertebral level in C9+ subjects older than 40 years without associated changes in GM and diffusion tensor imaging parameters. At 18-month follow-up, WM atrophy was accompanied by significant corticospinal tract (CST) fractional anisotropy (FA) reductions. Intriguingly, asymptomatic C9+ subjects older than 40 years with family history of ALS (as opposed to FTD) also exhibited significant CST FA reduction at baseline. INTERPRETATION Cervical SC imaging detects WM atrophy exclusively in C9+ subjects older than 40 years, and progressive CST FA reductions can be identified on 18-month follow-up. Cervical SC magnetic resonance imaging readily captures presymptomatic pathological changes and disease propagation in c9orf72-associated conditions. ANN NEUROL 2019;86:158-167.
Collapse
Affiliation(s)
- Giorgia Querin
- Department of Neurology, SLA Reference Center, Pitié-Salpêtrière Hospital, Public Hospital Network of Paris, Paris, France.,Laboratory of Biomedical Imaging, National Center for Scientific Research, National Institute of Health and Medical Research, Sorbonne University, Paris, France
| | - Peter Bede
- Department of Neurology, SLA Reference Center, Pitié-Salpêtrière Hospital, Public Hospital Network of Paris, Paris, France.,Laboratory of Biomedical Imaging, National Center for Scientific Research, National Institute of Health and Medical Research, Sorbonne University, Paris, France.,Computational Neuroimaging Group, Academic Unit of Neurology, Trinity College Dublin, Dublin, Ireland
| | - Mohamed Mounir El Mendili
- Laboratory of Biomedical Imaging, National Center for Scientific Research, National Institute of Health and Medical Research, Sorbonne University, Paris, France.,Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Menghan Li
- Laboratory of Biomedical Imaging, National Center for Scientific Research, National Institute of Health and Medical Research, Sorbonne University, Paris, France
| | - Mélanie Pélégrini-Issac
- Laboratory of Biomedical Imaging, National Center for Scientific Research, National Institute of Health and Medical Research, Sorbonne University, Paris, France
| | - Daisy Rinaldi
- Brain and Spinal Cord Institute, Sorbonne University, National Institute of Health and Medical Research U1127, National Center for Scientific Research Mixed Unit of Research 7225, Pitié-Salpêtrière Hospital, Paris, France.,Reference Center for Rare or Early Dementia, Pitié-Salpêtrière Hospital, Paris, France
| | - Martin Catala
- Department of Neurology, Pitié-Salpêtrière Hospital, Public Hospital Network of Paris, Sorbonne University, National Center for Scientific Research Mixed Unit of Research 7622, National Institute of Health and Medical Research Accademic Research Unit 1156, Biology Institute Paris-Seine, Paris, France
| | - Dario Saracino
- Brain and Spinal Cord Institute, Sorbonne University, National Institute of Health and Medical Research U1127, National Center for Scientific Research Mixed Unit of Research 7225, Pitié-Salpêtrière Hospital, Paris, France
| | - François Salachas
- Department of Neurology, SLA Reference Center, Pitié-Salpêtrière Hospital, Public Hospital Network of Paris, Paris, France
| | - Agnes Camuzat
- Brain and Spinal Cord Institute, Sorbonne University, National Institute of Health and Medical Research U1127, National Center for Scientific Research Mixed Unit of Research 7225, Pitié-Salpêtrière Hospital, Paris, France
| | - Véronique Marchand-Pauvert
- Laboratory of Biomedical Imaging, National Center for Scientific Research, National Institute of Health and Medical Research, Sorbonne University, Paris, France
| | - Julien Cohen-Adad
- NeuroPoly Laboratory, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, Quebec, Canada.,Functional Neuroimaging Unit, Research Center of the University Institute of Geriatrics of Montreal, University of Montreal, Montreal, Quebec, Canada
| | - Olivier Colliot
- Brain and Spinal Cord Institute, Sorbonne University, National Institute of Health and Medical Research U1127, National Center for Scientific Research Mixed Unit of Research 7225, Pitié-Salpêtrière Hospital, Paris, France.,Aramis Project Team, Inria Research Center of Paris, Paris, France.,Center for Image Acquisition and Processing, Brain and Spinal Cord Institute, Paris, France
| | - Isabelle Le Ber
- Brain and Spinal Cord Institute, Sorbonne University, National Institute of Health and Medical Research U1127, National Center for Scientific Research Mixed Unit of Research 7225, Pitié-Salpêtrière Hospital, Paris, France.,Reference Center for Rare or Early Dementia, Pitié-Salpêtrière Hospital, Paris, France.,Institute of Memory and Alzheimer's Disease, Center of Excellence of Neurodegenerative Disease, Department of Neurology, SLA Reference Center, Pitié-Salpêtrière Hospital, Public Hospital Network of Paris, Paris, France
| | - Pierre-François Pradat
- Department of Neurology, SLA Reference Center, Pitié-Salpêtrière Hospital, Public Hospital Network of Paris, Paris, France.,Laboratory of Biomedical Imaging, National Center for Scientific Research, National Institute of Health and Medical Research, Sorbonne University, Paris, France.,Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, Ulster University, Clinical-Translational Research and Innovation Center, Altnagelvin Hospital, Londonderry, United Kingdom
| | | |
Collapse
|
19
|
Martier R, Liefhebber JM, Miniarikova J, van der Zon T, Snapper J, Kolder I, Petry H, van Deventer SJ, Evers MM, Konstantinova P. Artificial MicroRNAs Targeting C9orf72 Can Reduce Accumulation of Intra-nuclear Transcripts in ALS and FTD Patients. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 14:593-608. [PMID: 30776581 PMCID: PMC6378669 DOI: 10.1016/j.omtn.2019.01.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/22/2019] [Accepted: 01/22/2019] [Indexed: 12/13/2022]
Abstract
The most common pathogenic mutation in amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) is an intronic GGGGCC (G4C2) repeat in the chromosome 9 open reading frame 72 (C9orf72) gene. Cellular toxicity due to RNA foci and dipeptide repeat (DPR) proteins produced by the sense and antisense repeat-containing transcripts is thought to underlie the pathogenesis of both diseases. RNA sequencing (RNA-seq) data of C9orf72-ALS patients and controls were analyzed to better understand the sequence conservation of C9orf72 in patients. MicroRNAs were developed in conserved regions to silence C9orf72 (miC), and the feasibility of different silencing approaches was demonstrated in reporter overexpression systems. In addition, we demonstrated the feasibility of a bidirectional targeting approach by expressing two concatenated miC hairpins. The efficacy of miC was confirmed by the reduction of endogenously expressed C9orf72 mRNA, in both nucleus and cytoplasm, and an ∼50% reduction of nuclear RNA foci in (G4C2)44-expressing cells. Ultimately, two miC candidates were incorporated in adeno-associated virus vector serotype 5 (AAV5), and silencing of C9orf72 was demonstrated in HEK293T cells and induced pluripotent stem cell (iPSC)-derived neurons. These data support the feasibility of microRNA (miRNA)-based and AAV-delivered gene therapy that could alleviate the gain of toxicity seen in ALS and FTD patients.
Collapse
Affiliation(s)
- Raygene Martier
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, the Netherlands; Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Jolanda M Liefhebber
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, the Netherlands
| | - Jana Miniarikova
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, the Netherlands
| | - Tom van der Zon
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, the Netherlands
| | - Jolanda Snapper
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, the Netherlands
| | - Iris Kolder
- BaseClear B.V., Sylviusweg 74, 2333 BE, Leiden, the Netherlands
| | - Harald Petry
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, the Netherlands
| | - Sander J van Deventer
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, the Netherlands; Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Melvin M Evers
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, the Netherlands
| | - Pavlina Konstantinova
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, the Netherlands.
| |
Collapse
|
20
|
Liao TC, Ma TZ, Liang Z, Zhang XT, Luo CY, Liu L, Zhou CQ. A Comparative Study on High Selectivities of Human Telomeric Dimeric G-Quadruplexes by Dimeric G-Quadruplex Binders. Chemistry 2018; 24:15840-15851. [DOI: 10.1002/chem.201802796] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 07/18/2018] [Indexed: 01/02/2023]
Affiliation(s)
- Ting-Cong Liao
- Guangdong Provincial Key Laboratory of New Drug Screening; School of Pharmaceutical Sciences; Southern Medical University; Guangzhou 510515 P. R. China
| | - Tian-Zhu Ma
- Guangdong Provincial Key Laboratory of New Drug Screening; School of Pharmaceutical Sciences; Southern Medical University; Guangzhou 510515 P. R. China
| | - Zhi Liang
- Guangdong Provincial Key Laboratory of New Drug Screening; School of Pharmaceutical Sciences; Southern Medical University; Guangzhou 510515 P. R. China
| | - Xin-Tong Zhang
- Guangdong Provincial Key Laboratory of New Drug Screening; School of Pharmaceutical Sciences; Southern Medical University; Guangzhou 510515 P. R. China
| | - Chun-Yin Luo
- Guangdong Provincial Key Laboratory of New Drug Screening; School of Pharmaceutical Sciences; Southern Medical University; Guangzhou 510515 P. R. China
| | - Lihong Liu
- Guangdong Provincial Key Laboratory of New Drug Screening; School of Pharmaceutical Sciences; Southern Medical University; Guangzhou 510515 P. R. China
| | - Chun-Qiong Zhou
- Guangdong Provincial Key Laboratory of New Drug Screening; School of Pharmaceutical Sciences; Southern Medical University; Guangzhou 510515 P. R. China
| |
Collapse
|
21
|
Deverman BE, Ravina BM, Bankiewicz KS, Paul SM, Sah DWY. Gene therapy for neurological disorders: progress and prospects. Nat Rev Drug Discov 2018; 17:641-659. [DOI: 10.1038/nrd.2018.110] [Citation(s) in RCA: 215] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
22
|
Kumar V, Hasan GM, Hassan MI. Unraveling the Role of RNA Mediated Toxicity of C9orf72 Repeats in C9-FTD/ALS. Front Neurosci 2017; 11:711. [PMID: 29326544 PMCID: PMC5736982 DOI: 10.3389/fnins.2017.00711] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 12/05/2017] [Indexed: 12/12/2022] Open
Abstract
The most frequent genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) is intronic hexanucleotide (G4C2) repeat expansions (HRE) in the C9orf72 gene. The non-exclusive pathogenic mechanisms by which C9orf72 repeat expansions contribute to these neurological disorders include loss of C9orf72 function and gain-of-function determined by toxic RNA molecules and dipeptides repeats protein toxicity. The expanded repeats are transcribed bidirectionally and forms RNA foci in the central nervous system, and sequester key RNA-binding proteins (RBPs) leading to impairment in RNA processing events. Many studies report widespread transcriptome changes in ALS carrying a C9orf72 repeat expansion. Here we review the contribution of RNA foci interaction with RBPs as well as transcriptome changes involved in the pathogenesis of C9orf72- associated FTD/ALS. These informations are essential to elucidate the pathology and therapeutic intervention of ALS and/or FTD.
Collapse
Affiliation(s)
- Vijay Kumar
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, India
| | - Gulam M Hasan
- Department of Biochemistry, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, India
| |
Collapse
|
23
|
Kim BJ, Irwin DJ, Song D, Daniel E, Leveque JD, Raquib AR, Pan W, Ying GS, Aleman TS, Dunaief JL, Grossman M. Optical coherence tomography identifies outer retina thinning in frontotemporal degeneration. Neurology 2017; 89:1604-1611. [PMID: 28887373 DOI: 10.1212/wnl.0000000000004500] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 07/21/2017] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE Whereas Alzheimer disease (AD) is associated with inner retina thinning visualized by spectral-domain optical coherence tomography (SD-OCT), we sought to determine if the retina has a distinguishing biomarker for frontotemporal degeneration (FTD). METHODS Using a cross-sectional design, we examined retinal structure in 38 consecutively enrolled patients with FTD and 44 controls using a standard SD-OCT protocol. Retinal layers were segmented with the Iowa Reference Algorithm. Subgroups of highly predictive molecular pathology (tauopathy, TAR DNA-binding protein 43, unknown) were determined by clinical criteria, genetic markers, and a CSF biomarker (total tau: β-amyloid) to exclude presumed AD. We excluded eyes with poor image quality or confounding diseases. SD-OCT measures of patients (n = 46 eyes) and controls (n = 69 eyes) were compared using a generalized linear model accounting for intereye correlation, and correlations between retinal layer thicknesses and Mini-Mental State Examination (MMSE) were evaluated. RESULTS Adjusting for age, sex, and race, patients with FTD had a thinner outer retina than controls (132 vs 142 μm, p = 0.004). Patients with FTD also had a thinner outer nuclear layer (ONL) (88.5 vs 97.9 μm, p = 0.003) and ellipsoid zone (EZ) (14.5 vs 15.1 μm, p = 0.009) than controls, but had similar thicknesses for inner retinal layers. The outer retina thickness of patients correlated with MMSE (Spearman r = 0.44, p = 0.03). The highly predictive tauopathy subgroup (n = 31 eyes) also had a thinner ONL (88.7 vs 97.4 μm, p = 0.01) and EZ (14.4 vs 15.1 μm, p = 0.01) than controls. CONCLUSIONS FTD is associated with outer retina thinning, and this thinning correlates with disease severity.
Collapse
Affiliation(s)
- Benjamin J Kim
- From the Scheie Eye Institute, Department of Ophthalmology (B.J.K., D.S., E.D., J.D.L., A.R.R., W.P., G.-S.Y., T.S.A., J.L.D.), and Frontotemporal Lobar Degeneration Center, Department of Neurology (D.J.I., M.G.), Perelman School of Medicine, University of Pennsylvania, Philadelphia.
| | - David J Irwin
- From the Scheie Eye Institute, Department of Ophthalmology (B.J.K., D.S., E.D., J.D.L., A.R.R., W.P., G.-S.Y., T.S.A., J.L.D.), and Frontotemporal Lobar Degeneration Center, Department of Neurology (D.J.I., M.G.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Delu Song
- From the Scheie Eye Institute, Department of Ophthalmology (B.J.K., D.S., E.D., J.D.L., A.R.R., W.P., G.-S.Y., T.S.A., J.L.D.), and Frontotemporal Lobar Degeneration Center, Department of Neurology (D.J.I., M.G.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Ebenezer Daniel
- From the Scheie Eye Institute, Department of Ophthalmology (B.J.K., D.S., E.D., J.D.L., A.R.R., W.P., G.-S.Y., T.S.A., J.L.D.), and Frontotemporal Lobar Degeneration Center, Department of Neurology (D.J.I., M.G.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Jennifer D Leveque
- From the Scheie Eye Institute, Department of Ophthalmology (B.J.K., D.S., E.D., J.D.L., A.R.R., W.P., G.-S.Y., T.S.A., J.L.D.), and Frontotemporal Lobar Degeneration Center, Department of Neurology (D.J.I., M.G.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Aaishah R Raquib
- From the Scheie Eye Institute, Department of Ophthalmology (B.J.K., D.S., E.D., J.D.L., A.R.R., W.P., G.-S.Y., T.S.A., J.L.D.), and Frontotemporal Lobar Degeneration Center, Department of Neurology (D.J.I., M.G.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Wei Pan
- From the Scheie Eye Institute, Department of Ophthalmology (B.J.K., D.S., E.D., J.D.L., A.R.R., W.P., G.-S.Y., T.S.A., J.L.D.), and Frontotemporal Lobar Degeneration Center, Department of Neurology (D.J.I., M.G.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Gui-Shuang Ying
- From the Scheie Eye Institute, Department of Ophthalmology (B.J.K., D.S., E.D., J.D.L., A.R.R., W.P., G.-S.Y., T.S.A., J.L.D.), and Frontotemporal Lobar Degeneration Center, Department of Neurology (D.J.I., M.G.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Tomas S Aleman
- From the Scheie Eye Institute, Department of Ophthalmology (B.J.K., D.S., E.D., J.D.L., A.R.R., W.P., G.-S.Y., T.S.A., J.L.D.), and Frontotemporal Lobar Degeneration Center, Department of Neurology (D.J.I., M.G.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Joshua L Dunaief
- From the Scheie Eye Institute, Department of Ophthalmology (B.J.K., D.S., E.D., J.D.L., A.R.R., W.P., G.-S.Y., T.S.A., J.L.D.), and Frontotemporal Lobar Degeneration Center, Department of Neurology (D.J.I., M.G.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Murray Grossman
- From the Scheie Eye Institute, Department of Ophthalmology (B.J.K., D.S., E.D., J.D.L., A.R.R., W.P., G.-S.Y., T.S.A., J.L.D.), and Frontotemporal Lobar Degeneration Center, Department of Neurology (D.J.I., M.G.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| |
Collapse
|
24
|
Murphy NA, Arthur KC, Tienari PJ, Houlden H, Chiò A, Traynor BJ. Age-related penetrance of the C9orf72 repeat expansion. Sci Rep 2017; 7:2116. [PMID: 28522837 PMCID: PMC5437033 DOI: 10.1038/s41598-017-02364-1] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 04/10/2017] [Indexed: 12/11/2022] Open
Abstract
A pathogenic hexanucleotide repeat expansion within the C9orf72 gene has been identified as the major cause of two neurodegenerative syndromes, amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). This mutation is known to have incomplete penetrance, with some patients developing disease in their twenties and a small portion of carriers surviving to their ninth decade without developing symptoms. Describing penetrance by age among C9orf72 carriers and identifying parameters that alter onset age are essential to better understanding this locus and to enhance predictive counseling. To do so, data from 1,170 individuals were used to model penetrance. Our analysis showed that the penetrance was incomplete and age-dependent. Additionally, familial and sporadic penetrance did not significantly differ from one another; ALS cases exhibited earlier age of onset than FTD cases; and individuals with spinal-onset exhibited earlier age of onset than those with bulbar-onset. The older age of onset among female cases in general, and among female bulbar-onset cases in particular, was the most striking finding, and there may be an environmental, lifestyle, or hormonal factor that is influencing these penetrance patterns. These results will have important applications for future clinical research, the identification of disease modifiers, and genetic counseling.
Collapse
Affiliation(s)
- Natalie A Murphy
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Karissa C Arthur
- Geisinger Commonwealth School of Medicine, Scranton, Pennsylvania, 18509, USA
| | - Pentti J Tienari
- Department of Neurology, Helsinki University Hospital, Helsinki, FIN-02900, Finland.,Molecular Neurology Research Program Unit, University of Helsinki, Helsinki, FIN-02900, Finland
| | - Henry Houlden
- Department of Molecular Neurosciences, Institute of Neurology, University College London, Queen Square House, London, WC1N 3BG, UK
| | - Adriano Chiò
- Department of Neuroscience, University of Turin, 10126, Turin, Italy
| | - Bryan J Traynor
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, 20892, USA. .,Brain Science Institute, Department of Neurology, Johns Hopkins University, Baltimore, Maryland, 21205, USA.
| |
Collapse
|
25
|
Corcia P, Couratier P, Blasco H, Andres C, Beltran S, Meininger V, Vourc’h P. Genetics of amyotrophic lateral sclerosis. Rev Neurol (Paris) 2017; 173:254-262. [DOI: 10.1016/j.neurol.2017.03.030] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 03/27/2017] [Indexed: 12/12/2022]
|
26
|
Liang H, Wu C, Deng Y, Zhu L, Zhang J, Gan W, Tang C, Xu R. Aldehyde Dehydrogenases 1A2 Expression and Distribution are Potentially Associated with Neuron Death in Spinal Cord of Tg(SOD1*G93A)1Gur Mice. Int J Biol Sci 2017; 13:574-587. [PMID: 28539831 PMCID: PMC5441175 DOI: 10.7150/ijbs.19150] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 02/20/2017] [Indexed: 12/11/2022] Open
Abstract
The pathogenesis of amyotrophic lateral sclerosis (ALS) has not been unclear yet, it might be associated with the abnormal expression and distribution of certain proteins. Aldehyde dehydrogenases 1A2 (ALDH1A2) was thought to be one of potential candidates. Therefore, in this study we observed and analyzed the alteration of the expression and distribution of ALDH1A2 in the spinal cord of wild-type (WT) and Tg(SOD1*G93A)1Gur mice. We compared the expression and distribution of ALDH1A2 in the different segments, anatomic regions and neural cells of spinal cord at the different stages of WT and Tg(SOD1*G93A)1Gur mice applied the methods of fluorescent immunohistochemistry and western blot. Results revealed that ALDH1A2 extensively expressed and distributed in the spinal cord of adult WT and Tg(SOD1*G93A)1Gur mice. The expression and distribution of ALDH1A2 in the white matter including the anterior, posterior and lateral funiculus were more than that in the gray matter including the central canal, the anterior and dorsal horn. ALDH1A2 majorly expressed and distributed in the astrocyte, microglial, oligodendrocyte and neuron cells. The ALDH1A2 expression significantly decreased and redistributed in some anatomic regions of spinal cord at the onset and progression stages of Tg(SOD1*G93A)1Gur mice. The expression decrease of ALDH1A2 followed with the increase of neuron cells death. This study suggested that the alteration of expression and distribution of ALDH1A2 was potentially associated with the pathogenesis of ALS.
Collapse
Affiliation(s)
- Huiting Liang
- Department of Neurology, First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Chengsi Wu
- Department of Neurology, First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Youqing Deng
- Department of Neurology, Third Affiliated Hospital of Nanchang University, Nanchang 330008, Jiangxi, China
| | - Lei Zhu
- Department of Neurology, First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Jie Zhang
- Department of Neurology, First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Weiming Gan
- Department of Neurology, First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Chunyan Tang
- Department of Neurology, First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Renshi Xu
- Department of Neurology, First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| |
Collapse
|
27
|
Li ZQ, Liao TC, Dong C, Yang JW, Chen XJ, Liu L, Luo Y, Liang YY, Chen WH, Zhou CQ. Specifically targeting mixed-type dimeric G-quadruplexes using berberine dimers. Org Biomol Chem 2017; 15:10221-10229. [DOI: 10.1039/c7ob02326j] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Berberine dimer (1a) with the shortest polyether linker demonstrates highest binding affinity, selectivity and thermal stabilization towards mixed-type dimeric quadruplexes.
Collapse
|
28
|
Tan WH, Bird LM. Angelman syndrome: Current and emerging therapies in 2016. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2016; 172:384-401. [PMID: 27860204 DOI: 10.1002/ajmg.c.31536] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Angelman syndrome (AS) is a severe neurodevelopmental disorder caused by a loss of the maternally-inherited UBE3A; the paternal UBE3A is silenced in neurons by a mechanism involving an antisense transcript (UBE3A-AS) at the unmethylated paternal locus. We reviewed all published information on the clinical trials that have been completed as well as the publicly available information on ongoing trials of therapies in AS. To date, all clinical trials that strove to improve neurodevelopment in AS have been unsuccessful. Attempts at hypermethylating the maternal locus through dietary compounds were ineffective. The results of an 8-week open-label trial using minocycline as a matrix metalloproteinase-9 inhibitor were inconclusive, while a subsequent randomized placebo-controlled trial suggested that treatment with minocycline for 8 weeks did not result in any neurodevelopmental gains. A 1-year randomized placebo-controlled trial using levodopa to alter the phosphorylation of calcium/calmodulin-dependent kinase II did not lead to any improvement in neurodevelopment. Topoisomerase inhibitors and antisense oligonucleotides are being developed to directly inhibit UBE3A-AS. Artificial transcription factors are being developed to "super activate" UBE3A or inhibit UBE3A-AS. Other strategies targeting specific pathways are briefly discussed. We also reviewed the medications that are currently used to treat seizures and sleep disturbances, which are two of the more common complications of AS. © 2016 Wiley Periodicals, Inc.
Collapse
|