1
|
Chong ZZ, Souayah N. Targeting Gene C9orf72 Pathogenesis for Amyotrophic Lateral Sclerosis. Int J Mol Sci 2025; 26:4276. [PMID: 40362512 PMCID: PMC12072292 DOI: 10.3390/ijms26094276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/23/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal adult neurodegenerative disorder. Since no cure has been found, finding effective therapeutic targets for ALS remains a major challenge. Gene C9orf72 mutations with the formation of hexanucleotide repeat (GGGGCC) expansion (HRE) have been considered the most common genetic pathogenesis of ALS. The literature review indicates that the C9orf72 HRE causes both the gain-of-function toxicity and loss of function of C9ORF72. The formation of RNA foci and dipeptide repeats (DPRs) resulting from HRE is responsible for toxic function gain. The RNA foci can interfere with RNA processing, while DPRs directly bind to and sequester associated proteins to disrupt processes of rRNA synthesis, mRNA translation, autophagy, and nucleocytoplasmic transport. The mutations of C9orf72 and HRE result in the loss of functional C9ORF72. Under physiological conditions, C9ORF72 binds to Smith-Magenis chromosome region 8 and WD repeat-containing protein and forms a protein complex. Loss of C9ORF72 leads to autophagic impairment, increased oxidative stress, nucleocytoplasmic transport impairment, and inflammatory response. The attempted treatments for ALS have been tried by targeting C9orf72 HRE; however, the outcomes are far from satisfactory yet. More studies should be performed on pharmacological and molecular modulators against C9orf72 HRE to evaluate their efficacy by targeting HRE.
Collapse
Affiliation(s)
- Zhao Zhong Chong
- Department of Neurology, New Jersey Medical School, Rutgers University, 185 S Orange, Newark, NJ 07103, USA
| | - Nizar Souayah
- Department of Neurology, New Jersey Medical School, Rutgers University, 90 Bergen Street DOC 8100, Newark, NJ 07101, USA
| |
Collapse
|
2
|
Song M, Ren J, Zhu Z, Yi Z, Wang C, Liang L, Tian J, Mao G, Mao G, Chen M. The STING Signaling: A Novel Target for Central Nervous System Diseases. Cell Mol Neurobiol 2025; 45:33. [PMID: 40195137 PMCID: PMC11977075 DOI: 10.1007/s10571-025-01550-4] [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: 01/11/2025] [Accepted: 03/19/2025] [Indexed: 04/09/2025]
Abstract
The canonical cyclic GMP-AMP (cGAMP) synthase (cGAS)-Stimulator of Interferon Genes (STING) pathway has been widely recognized as a crucial mediator of inflammation in many diseases, including tumors, infections, and tissue damage. STING signaling can also be activated in a cGAS- or cGAMP-independent manner, although the specific mechanisms remain unclear. In-depth studies on the structural and molecular biology of the STING pathway have led to the development of therapeutic strategies involving STING modulators and their targeted delivery. These strategies may effectively penetrate the blood-brain barrier (BBB) and target STING signaling in multiple central nervous system (CNS) diseases in humans. In this review, we outline both canonical and non-canonical pathways of STING activation and describe the general mechanisms and associations between STING activity and CNS diseases. Finally, we discuss the prospects for the targeted delivery and clinical application of STING agonists and inhibitors, highlighting the STING signaling pathway as a novel therapeutic target in CNS diseases.
Collapse
Affiliation(s)
- Min Song
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1 Minde Road, Nanchang, Jiangxi, China
| | - Jianxun Ren
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1 Minde Road, Nanchang, Jiangxi, China
| | - Zhipeng Zhu
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1 Minde Road, Nanchang, Jiangxi, China
- Department of Neurosurgery, Shangrao People's Hospital, Shangrao, Jiangxi Province, China
| | - Zhaohui Yi
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1 Minde Road, Nanchang, Jiangxi, China
| | - Chengyun Wang
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1 Minde Road, Nanchang, Jiangxi, China
| | - Lirong Liang
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1 Minde Road, Nanchang, Jiangxi, China
| | - Jiahui Tian
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1 Minde Road, Nanchang, Jiangxi, China
| | - Guofu Mao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1 Minde Road, Nanchang, Jiangxi, China
| | - Guohua Mao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1 Minde Road, Nanchang, Jiangxi, China.
| | - Min Chen
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1 Minde Road, Nanchang, Jiangxi, China.
| |
Collapse
|
3
|
Raas Q, Haouy G, de Calbiac H, Pasho E, Marian A, Guerrera IC, Rosello M, Oeckl P, Del Bene F, Catanese A, Ciura S, Kabashi E. TBK1 is involved in programmed cell death and ALS-related pathways in novel zebrafish models. Cell Death Discov 2025; 11:98. [PMID: 40075110 PMCID: PMC11903655 DOI: 10.1038/s41420-025-02374-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 01/27/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Pathogenic mutations within the TBK1 gene leading to haploinsufficiency are causative of amyotrophic lateral sclerosis (ALS). This gene is linked to autophagy and inflammation, two cellular mechanisms reported to be dysregulated in ALS patients, although its functional role in the pathogenesis could involve other players. We targeted the TBK1 ortholog in zebrafish, an optimal vertebrate model for investigating genetic defects in neurological disorders. We generated zebrafish models with invalidating tbk1 mutations using CRISPR-Cas9 or tbk1 knockdown models using antisense morpholino oligonucleotide (AMO). The early motor phenotype of zebrafish injected with tbk1 AMO beginning at 2 days post fertilization (dpf) is associated with the degeneration of motor neurons. In parallel, CRISPR-induced tbk1 mutants exhibit impaired motor function beginning at 5 dpf and increased lethality beginning at 9 dpf. A metabolomic analysis showed an association between tbk1 loss and severe dysregulation of nicotinamide metabolism, and incubation with nicotinamide riboside rescued the motor behavior of tbk1 mutant zebrafish. Furthermore, a proteomic analysis revealed increased levels of inflammatory markers and dysregulation of programmed cell death pathways. Necroptosis appeared to be strongly activated in TBK1 fish, and larvae treated with the necroptosis inhibitor necrosulfonamide exhibited improved survival. Finally, a combined analysis of mutant zebrafish and TBK1-mutant human motor neurons revealed dysregulation of the KEGG pathway "ALS", with disrupted nuclear-cytoplasmic transport and increased expression of STAT1. These findings point toward a major role for necroptosis in the degenerative features and premature lethality observed in tbk1 mutant zebrafish. Overall, the novel tbk1-deficient zebrafish models offer a great opportunity to better understand the cascade of events leading from the loss of tbk1 expression to the onset of motor deficits, with involvement of a metabolic defect and increased cell death, and for the development of novel therapeutic avenues for ALS and related neuromuscular diseases.
Collapse
Affiliation(s)
- Quentin Raas
- Laboratory of Translational Research for Neurological Disorders, Imagine Institute, INSERM UMR 1163, Université Paris Cité, 75015, Paris, France
| | - Gregoire Haouy
- Laboratory of Translational Research for Neurological Disorders, Imagine Institute, INSERM UMR 1163, Université Paris Cité, 75015, Paris, France
| | - Hortense de Calbiac
- Laboratory of Translational Research for Neurological Disorders, Imagine Institute, INSERM UMR 1163, Université Paris Cité, 75015, Paris, France
| | - Elena Pasho
- Laboratory of Translational Research for Neurological Disorders, Imagine Institute, INSERM UMR 1163, Université Paris Cité, 75015, Paris, France
| | - Anca Marian
- Laboratory of Translational Research for Neurological Disorders, Imagine Institute, INSERM UMR 1163, Université Paris Cité, 75015, Paris, France
| | - Ida Chiara Guerrera
- Proteomics Platform 3P5-Necker, - Structure Fédérative de Recherche Necker, Inserm US24/CNRS UAR 3633, Université Paris Cité, 75015, Paris, France
| | - Marion Rosello
- Sorbonne Université, INSERM U968, CNRS UMR 7210, Institut de la Vision, Paris, France
| | - Patrick Oeckl
- German Center for Neurodegenerative Diseases (DZNE) Ulm, Ulm, Germany
- Department of Neurology, Ulm University Hospital, Ulm, Germany
| | - Filippo Del Bene
- Sorbonne Université, INSERM U968, CNRS UMR 7210, Institut de la Vision, Paris, France
| | - Alberto Catanese
- Laboratory of Translational Research for Neurological Disorders, Imagine Institute, INSERM UMR 1163, Université Paris Cité, 75015, Paris, France
- Institute of Anatomy and Cell Biology, Ulm University School of Medicine, Ulm, Germany
| | - Sorana Ciura
- Laboratory of Translational Research for Neurological Disorders, Imagine Institute, INSERM UMR 1163, Université Paris Cité, 75015, Paris, France
| | - Edor Kabashi
- Laboratory of Translational Research for Neurological Disorders, Imagine Institute, INSERM UMR 1163, Université Paris Cité, 75015, Paris, France.
| |
Collapse
|
4
|
Ghezzi A, Gianferrari G, Baldassarri E, Zucchi E, Martinelli I, Vacchiano V, Bonan L, Zinno L, Nuredini A, Canali E, Gizzi M, Terlizzi E, Medici D, Sette E, Currò Dossi M, Morresi S, Santangelo M, Patuelli A, Longoni M, De Massis P, Ferro S, Fini N, Simonini C, Carra S, Zamboni G, Mandrioli J. Phenotypical Characterization of C9ALS Patients from the Emilia Romagna Registry of ALS: A Retrospective Case-Control Study. Genes (Basel) 2025; 16:309. [PMID: 40149460 PMCID: PMC11942173 DOI: 10.3390/genes16030309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 02/28/2025] [Accepted: 02/28/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES C9ORF72 expansion is associated with significant phenotypic heterogeneity. This study aimed to characterize the clinical features of C9ALS patients from the Emilia Romagna ALS registry (ERRALS) and compare them with non-mutated ALS (nmALS) patients matched for sex, age at onset, and diagnostic delay, sourced from the same register. METHODS In total, 67 C9ALS patients were compared to 201 nmALS. Clinical data, phenotype, and prognostic factors were analyzed in the two groups and within the C9ALS group after stratification by sex. RESULTS C9ALS patients displayed a higher disease progression rate and shorter times to gastrostomy and invasive ventilation, despite no differences in overall survival. Female C9ALS had a more severe bulbar and upper motor neuron involvement compared to males. Cognitive and behavioral symptoms were more common in the C9ALS group, and the former was an independent prognostic factor. Prevalences of, autoimmune diseases, and dyslipidemia were significantly higher among C9ALS patients. CONCLUSIONS In our dataset, we show an overall increased disease progression rate in C9ALS patients and hint at sex-specific discrepancies in some phenotypical characteristics. We also suggest a possible clinically relevant involvement of C9ORF72 expansion in metabolism and autoimmunity.
Collapse
Affiliation(s)
- Andrea Ghezzi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (A.G.); (E.B.); (E.Z.); (S.C.); (G.Z.); (J.M.)
- Neurology Unit, Azienda Ospedaliero Universitaria di Modena, 41126 Modena, Italy; (I.M.); (N.F.); (C.S.)
| | - Giulia Gianferrari
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (A.G.); (E.B.); (E.Z.); (S.C.); (G.Z.); (J.M.)
- Neurology Unit, Azienda Ospedaliero Universitaria di Modena, 41126 Modena, Italy; (I.M.); (N.F.); (C.S.)
| | - Elisa Baldassarri
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (A.G.); (E.B.); (E.Z.); (S.C.); (G.Z.); (J.M.)
| | - Elisabetta Zucchi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (A.G.); (E.B.); (E.Z.); (S.C.); (G.Z.); (J.M.)
- Neurology Unit, Azienda Ospedaliero Universitaria di Modena, 41126 Modena, Italy; (I.M.); (N.F.); (C.S.)
| | - Ilaria Martinelli
- Neurology Unit, Azienda Ospedaliero Universitaria di Modena, 41126 Modena, Italy; (I.M.); (N.F.); (C.S.)
| | - Veria Vacchiano
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, 40139 Bologna, Italy;
| | - Luigi Bonan
- Dipartimento di Scienze Biomediche e Neuromotorie, University of Bologna, 40126 Bologna, Italy;
| | - Lucia Zinno
- Department of Medicine and Surgery, University of Parma, 43121 Parma, Italy; (L.Z.); (A.N.)
| | - Andi Nuredini
- Department of Medicine and Surgery, University of Parma, 43121 Parma, Italy; (L.Z.); (A.N.)
| | - Elena Canali
- Neurology Unit, Arcispedale Santa Maria Nuova, AUSL-IRCCS Reggio Emilia, 42123 Reggio Emilia, Italy;
| | - Matteo Gizzi
- Department of Neurology, Faenza and Ravenna Hospital, 48121 Ravenna, Italy;
| | - Emilio Terlizzi
- Department of Neurology, G. Da Saliceto Hospital, 29121 Piacenza, Italy;
| | - Doriana Medici
- Department of Neurology, Fidenza Hospital, 43036 Fidenza, Italy;
| | - Elisabetta Sette
- Department of Neuroscience and Rehabilitation, St. Anna Hospital, 44124 Ferrara, Italy;
| | | | - Simonetta Morresi
- Department of Neurology and Stroke Unit, Bufalini Hospital, 47521 Cesena, Italy;
| | | | - Alberto Patuelli
- Department of Neurology and Stroke Unit, “Morgagni-Pierantoni” Hospital, 47121 Forlì, Italy; (A.P.); (M.L.)
| | - Marco Longoni
- Department of Neurology and Stroke Unit, “Morgagni-Pierantoni” Hospital, 47121 Forlì, Italy; (A.P.); (M.L.)
| | | | - Salvatore Ferro
- Department of Hospital Services, Emilia Romagna Regional Health Authority, 40127 Bologna, Italy;
| | - Nicola Fini
- Neurology Unit, Azienda Ospedaliero Universitaria di Modena, 41126 Modena, Italy; (I.M.); (N.F.); (C.S.)
| | - Cecilia Simonini
- Neurology Unit, Azienda Ospedaliero Universitaria di Modena, 41126 Modena, Italy; (I.M.); (N.F.); (C.S.)
| | - Serena Carra
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (A.G.); (E.B.); (E.Z.); (S.C.); (G.Z.); (J.M.)
| | - Giovanna Zamboni
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (A.G.); (E.B.); (E.Z.); (S.C.); (G.Z.); (J.M.)
- Neurology Unit, Azienda Ospedaliero Universitaria di Modena, 41126 Modena, Italy; (I.M.); (N.F.); (C.S.)
| | - Jessica Mandrioli
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (A.G.); (E.B.); (E.Z.); (S.C.); (G.Z.); (J.M.)
- Neurology Unit, Azienda Ospedaliero Universitaria di Modena, 41126 Modena, Italy; (I.M.); (N.F.); (C.S.)
| |
Collapse
|
5
|
Wang Y, Xu T, Wang W. C9orf72 Alleviates DSS‑Induced Ulcerative Colitis via the cGAS-STING Pathway. Immun Inflamm Dis 2025; 13:e70139. [PMID: 39873292 PMCID: PMC11774236 DOI: 10.1002/iid3.70139] [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: 07/08/2024] [Revised: 10/14/2024] [Accepted: 01/19/2025] [Indexed: 01/30/2025] Open
Abstract
PURPOSE C9orf72 deficiency contributes to severe inflammation in mice. Ulcerative colitis (UC) is a chronic inflammatory disorder with the shortage of clinical success. However, whether C9orf72 is involved in the progression of UC is not fully understood. This study investigated whether C9orf72 could alleviate dextran sulfate sodium (DSS)-induced colitis in mice and lipopolysaccharide (LPS)-induced colitis in Caco-2 cells. METHODS Mice were injected AAV9-C9orf72 lentivirus through tail vein and fed 3% DSS for a week. Caco-2 cells were cultured to establish C9orf723 overexpressed model. Histopathological examination, level of inflammation, cGAS-STING pathway, and gut barrier function were detected in mice and cells. RESULTS C9orf72 overexpression in mice attenuated DSS-induced colitis and intestinal epithelial barrier damage by stimulating ZO-1 and Occludin expression. In LPS-induced Caco-2 cells, C9orf72 overexpression increased cell viability and the expression of ZO-1 and Occludin. C9orf72 overexpression alleviated inflammation by inhibiting the cGAS-STING pathway in colonic tissue and Caco-2 cells. CONCLUSION C9orf72 overexpression attenuated DSS-induced colitis and intestinal epithelial barrier damage by inhibiting the cGAS-STING pathway. C9orf72 may present a target for mitigating UC.
Collapse
Affiliation(s)
- Yue Wang
- Department of GastroenterologyQingdao Municipal HospitalQingdaoShandongChina
| | - Ting Xu
- Department of GastroenterologyQingdao Municipal HospitalQingdaoShandongChina
| | - Wenjun Wang
- Department of Health CareQingdao Municipal HospitalQingdaoShandongChina
| |
Collapse
|
6
|
McKeever PM, Sababi AM, Sharma R, Xu Z, Xiao S, McGoldrick P, Ketela T, Sato C, Moreno D, Visanji N, Kovacs GG, Keith J, Zinman L, Rogaeva E, Goodarzi H, Bader GD, Robertson J. Single-nucleus transcriptome atlas of orbitofrontal cortex in amyotrophic lateral sclerosis with a deep learning-based decoding of alternative polyadenylation mechanisms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.22.573083. [PMID: 38187588 PMCID: PMC10769403 DOI: 10.1101/2023.12.22.573083] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) are two age-related and fatal neurodegenerative disorders that lie on a shared disease spectrum. While both disorders involve complex interactions between neuronal and glial cells, the specific cell-type alterations and their contributions to disease pathophysiology remain incompletely understood. Here, we applied single-nucleus RNA sequencing of the orbitofrontal cortex, a region affected in ALS-FTLD, to map cell-type specific transcriptional signatures in C9orf72-related ALS (with and without FTLD) and sporadic ALS cases. Our findings reveal disease- and cell-type-specific transcriptional changes, with neurons exhibiting the most pronounced alterations, primarily affecting mitochondrial function, protein homeostasis, and chromatin remodeling. A comparison with independent datasets from different cortical regions of C9orf72 and sporadic ALS cases showed concordance in several pathways, with neuronal STMN2 and NEFL showing consistent up-regulation between brain regions and disease subtypes. We also interrogated alternative polyadenylation (APA) as an additional layer of transcriptional regulation, demonstrating that APA events are not correlated with identified gene expression changes. To interpret these events, we developed APA-Net, a deep learning model that integrates transcript sequences with RNA-binding protein expression profiles, revealing cell type-specific patterns of APA regulation. Our atlas illuminates cell type-specific pathomechanisms of ALS/FTLD, providing a valuable resource for further investigation.
Collapse
|
7
|
Zhang Y, Zou M, Wu H, Zhu J, Jin T. The cGAS-STING pathway drives neuroinflammation and neurodegeneration via cellular and molecular mechanisms in neurodegenerative diseases. Neurobiol Dis 2024; 202:106710. [PMID: 39490400 DOI: 10.1016/j.nbd.2024.106710] [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: 07/03/2024] [Revised: 09/27/2024] [Accepted: 10/18/2024] [Indexed: 11/05/2024] Open
Abstract
Neurodegenerative diseases (NDs) are a type of common chronic progressive disorders characterized by progressive damage to specific cell populations in the nervous system, ultimately leading to disability or death. Effective treatments for these diseases are still lacking, due to a limited understanding of their pathogeneses, which involve multiple cellular and molecular pathways. The triggering of an immune response is a common feature in neurodegenerative disorders. A critical challenge is the intricate interplay between neuroinflammation, neurodegeneration, and immune responses, which are not yet fully characterized. In recent years, the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon gene (STING) pathway, a crucial immune response for intracellular DNA sensing, has gradually gained attention. However, the specific roles of this pathway within cellular types such as immune cells, glial and neuronal cells, and its contribution to ND pathogenesis, remain not fully elucidated. In this review, we systematically explore how the cGAS-STING signaling links various cell types with related cellular effector pathways under the context of NDs for multifaceted therapeutic directions. We emphasize the discovery of condition-dependent cellular heterogeneity in the cGAS-STING pathway, which is integral for understanding the diverse cellular responses and potential therapeutic targets. Additionally, we review the pathogenic role of cGAS-STING activation in Parkinson's disease, ataxia-telangiectasia, and amyotrophic lateral sclerosis. We focus on the complex bidirectional roles of the cGAS-STING pathway in Alzheimer's disease, Huntington's disease, and multiple sclerosis, revealing their double-edged nature in disease progression. The objective of this review is to elucidate the pivotal role of the cGAS-STING pathway in ND pathogenesis and catalyze new insights for facilitating the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Yuxin Zhang
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Meijuan Zou
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Hao Wu
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jie Zhu
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China; Department of Neurobiology, Care Sciences & Society, Karolinska Institute, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Tao Jin
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
8
|
Mirceta M, Schmidt MHM, Shum N, Prasolava TK, Meikle B, Lanni S, Mohiuddin M, Mckeever PM, Zhang M, Liang M, van der Werf I, Scheers S, Dion PA, Wang P, Wilson MD, Abell T, Philips EA, Sznajder ŁJ, Swanson MS, Mehkary M, Khan M, Yokoi K, Jung C, de Jong PJ, Freudenreich CH, McGoldrick P, Yuen RKC, Abrahão A, Keith J, Zinman L, Robertson J, Rogaeva E, Rouleau GA, Kooy RF, Pearson CE. C9orf72 expansion creates the unstable folate-sensitive fragile site FRA9A. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.26.620312. [PMID: 39569145 PMCID: PMC11577248 DOI: 10.1101/2024.10.26.620312] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
The hyper-unstable Chr9p21 locus, harbouring the interferon gene cluster, oncogenes and C9orf72, is linked to multiple diseases. C9orf72 (GGGGCC)n expansions ( C9orf72 Exp) are associated with incompletely penetrant amyotrophic lateral sclerosis, frontotemporal dementia and autoimmune disorders. C9orf72 Exp patients display hyperactive cGAS-STING-linked interferon immune and DNA damage responses, but the source of immuno-stimulatory or damaged DNA is unknown. Here, we show C9orf72 Exp in pre-symptomatic and ALS-FTD patient cells and brains cause the folate-sensitive chromosomal fragile site, FRA9A. FRA9A centers on >33kb of C9orf72 as highly-compacted chromatin embedded in an 8.2Mb fragility zone spanning 9p21, encompassing 46 genes, making FRA9A one of the largest fragile sites. C9orf72 Exp cells show chromosomal instability, heightened global- and Chr9p-enriched sister-chromatid exchanges, truncated-Chr9s, acentric-Chr9s and Chr9-containing micronuclei, providing endogenous sources of damaged and immunostimulatory DNA. Cells from one C9orf72 Exp patient contained highly-rearranged FRA9A-expressing Chr9 with Chr9-wide dysregulated gene expression. Somatic C9orf72 Exp repeat instability and chromosomal fragility are sensitive to folate-deficiency. Age-dependent repeat instability, chromosomal fragility, and chromosomal instability can be transferred to CNS and peripheral tissues of transgenic C9orf72 Exp mice, implicating C9orf72 Exp as the source. Our results highlight unappreciated effects of C9orf72 expansions that trigger vitamin-sensitive chromosome fragility, adding structural variations to the disease-enriched 9p21 locus, and likely elsewhere.
Collapse
|
9
|
Sirkis DW, Oddi AP, Jonson C, Bonham LW, Hoang PT, Yokoyama JS. The role of interferon signaling in neurodegeneration and neuropsychiatric disorders. Front Psychiatry 2024; 15:1480438. [PMID: 39421070 PMCID: PMC11484020 DOI: 10.3389/fpsyt.2024.1480438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 09/09/2024] [Indexed: 10/19/2024] Open
Abstract
Recent advances in transcriptomics research have uncovered heightened interferon (IFN) responses in neurodegenerative diseases including Alzheimer's disease, primary tauopathy, Parkinson's disease, TDP-43 proteinopathy, and related mouse models. Augmented IFN signaling is now relatively well established for microglia in these contexts, but emerging work has highlighted a novel role for IFN-responsive T cells in the brain and peripheral blood in some types of neurodegeneration. These findings complement a body of literature implicating dysregulated IFN signaling in neuropsychiatric disorders including major depression and post-traumatic stress disorder. In this review, we will characterize and integrate advances in our understanding of IFN responses in neurodegenerative and neuropsychiatric disease, discuss how sex and ancestry modulate the IFN response, and examine potential mechanistic explanations for the upregulation of antiviral-like IFN signaling pathways in these seemingly non-viral neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Daniel W. Sirkis
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States
| | - Alexis P. Oddi
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States
| | - Caroline Jonson
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States
- Center for Alzheimer’s and Related Dementias, National Institutes of Health, Bethesda, MD, United States
- DataTecnica LLC, Washington, DC, United States
| | - Luke W. Bonham
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
| | - Phuong T. Hoang
- Movement Disorders and Neuromodulation Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States
| | - Jennifer S. Yokoyama
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
- Global Brain Health Institute, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
10
|
Mirceta M, Schmidt MM, Shum N, Prasolava T, Meikle B, Lanni S, Mohiuddin M, McKeever P, Zhang M, Liang M, van der Werf I, Scheers S, Dion P, Wang P, Wilson M, Abell T, Philips E, Sznajder Ł, Swanson M, Mehkary M, Khan M, Yokoi K, Jung C, de Jong P, Freudenreich C, McGoldrick P, Yuen RC, Abrahão A, Keith J, Zinman L, Robertson J, Rogaeva E, Rouleau G, Kooy R, Pearson C. C9orf72 repeat expansion creates the unstable folate-sensitive fragile site FRA9A. NAR MOLECULAR MEDICINE 2024; 1:ugae019. [PMID: 39669124 PMCID: PMC11632612 DOI: 10.1093/narmme/ugae019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 11/11/2024] [Indexed: 12/14/2024]
Abstract
The hyper-unstable Chr9p21 locus, harbouring the interferon gene cluster, oncogenes and C9orf72, is linked to multiple diseases. C9orf72 (GGGGCC)n expansions (C9orf72Exp) are associated with incompletely penetrant amyotrophic lateral sclerosis, frontotemporal dementia and autoimmune disorders. C9orf72Exp patients display hyperactive cGAS-STING-linked interferon immune and DNA damage responses, but the source of immunostimulatory or damaged DNA is unknown. Here, we show C9orf72Exp in pre-symptomatic and amyotrophic lateral sclerosis-frontotemporal dementia patient cells and brains cause the folate-sensitive chromosomal fragile site, FRA9A. FRA9A centers on >33 kb of C9orf72 as highly compacted chromatin embedded in an 8.2 Mb fragility zone spanning 9p21, encompassing 46 genes, making FRA9A one of the largest fragile sites. C9orf72Exp cells show chromosomal instability, heightened global- and Chr9p-enriched sister-chromatid exchanges, truncated-Chr9s, acentric-Chr9s and Chr9-containing micronuclei, providing endogenous sources of damaged and immunostimulatory DNA. Cells from one C9orf72Exp patient contained a highly rearranged FRA9A-expressing Chr9 with Chr9-wide dysregulated gene expression. Somatic C9orf72Exp repeat instability and chromosomal fragility are sensitive to folate deficiency. Age-dependent repeat instability, chromosomal fragility and chromosomal instability can be transferred to CNS and peripheral tissues of transgenic C9orf72Exp mice, implicating C9orf72Exp as the source. Our results highlight unappreciated effects of C9orf72 expansions that trigger vitamin-sensitive chromosome fragility, adding structural variations to the disease-enriched 9p21 locus, and likely elsewhere.
Collapse
Affiliation(s)
- Mila Mirceta
- Program of Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON, M3S 1A8, Canada
| | - Monika H M Schmidt
- Program of Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON, M3S 1A8, Canada
| | - Natalie Shum
- Program of Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON, M3S 1A8, Canada
| | - Tanya K Prasolava
- Program of Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, M5G 0A4, Canada
| | - Bryanna Meikle
- Program of Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON, M3S 1A8, Canada
| | - Stella Lanni
- Program of Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, M5G 0A4, Canada
| | - Mohiuddin Mohiuddin
- Program of Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, M5G 0A4, Canada
| | - Paul M McKeever
- Tanz Centre for Research of Neurodegenerative Diseases, University of Toronto, 60 Leonard Avenue, Toronto, M5T 2S8, Canada
| | - Ming Zhang
- Tanz Centre for Research of Neurodegenerative Diseases, University of Toronto, 60 Leonard Avenue, Toronto, M5T 2S8, Canada
- The First Rehabilitation Hospital of Shanghai, Department of Medical Genetics, School of Medicine, Tongji University, Shanghai, 200090, China
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Advanced Study, Tongji University, Shanghai, 200092, China
| | - Minggao Liang
- Program of Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON, M3S 1A8, Canada
| | | | - Stefaan Scheers
- Department of Medical Genetics, University of Antwerp, Belgium
| | - Patrick A Dion
- Montreal Neurological Institute-Hospital, McGill University, 3801 University Avenue, Montreal, Quebec, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, 3801 University Avenue, Montreal, Quebec, H3A 2B4, Canada
| | - Peixiang Wang
- Program of Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, M5G 0A4, Canada
| | - Michael D Wilson
- Program of Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON, M3S 1A8, Canada
| | - Theresa Abell
- Department of Biology, Tufts University, 200 Boston Avenue, Medford, MA 02155, USA
| | - Elliot A Philips
- Department of Biology, Tufts University, 200 Boston Avenue, Medford, MA 02155, USA
| | - Łukasz J Sznajder
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, College of Medicine, University of Florida, 2033 Mowry Road, Gainesville, FL 32610-3610, USA
- Department of Chemistry and Biochemistry, University of Nevada, 4003-4505 South Maryland Parkway, Las Vegas, NV 89154, USA
| | - Maurice S Swanson
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, College of Medicine, University of Florida, 2033 Mowry Road, Gainesville, FL 32610-3610, USA
| | - Mustafa Mehkary
- Program of Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON, M3S 1A8, Canada
| | - Mahreen Khan
- Program of Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON, M3S 1A8, Canada
| | - Katsuyuki Yokoi
- Program of Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, M5G 0A4, Canada
| | - Christine Jung
- BACPAC Resource Center, Children’s Hospital Oakland Research Institute, 25129 NE 42nd Pl, Redmond, WA 98053, USA
| | - Pieter J de Jong
- BACPAC Resource Center, Children’s Hospital Oakland Research Institute, 25129 NE 42nd Pl, Redmond, WA 98053, USA
| | | | - Philip McGoldrick
- Tanz Centre for Research of Neurodegenerative Diseases, University of Toronto, 60 Leonard Avenue, Toronto, M5T 2S8, Canada
| | - Ryan K C Yuen
- Program of Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON, M3S 1A8, Canada
| | - Agessandro Abrahão
- Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, North York, Toronto, ON, M4N 3M5, Canada
| | - Julia Keith
- Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, North York, Toronto, ON, M4N 3M5, Canada
| | - Lorne Zinman
- Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, North York, Toronto, ON, M4N 3M5, Canada
| | - Janice Robertson
- Tanz Centre for Research of Neurodegenerative Diseases, University of Toronto, 60 Leonard Avenue, Toronto, M5T 2S8, Canada
| | - Ekaterina Rogaeva
- Tanz Centre for Research of Neurodegenerative Diseases, University of Toronto, 60 Leonard Avenue, Toronto, M5T 2S8, Canada
| | - Guy A Rouleau
- Montreal Neurological Institute-Hospital, McGill University, 3801 University Avenue, Montreal, Quebec, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, 3801 University Avenue, Montreal, Quebec, H3A 2B4, Canada
- Department of Human Genetics, McGill University, 3801 University Avenue, Montreal, Quebec, H3A 2B4, Canada
| | - R Frank Kooy
- Department of Medical Genetics, University of Antwerp, Belgium
| | - Christopher E Pearson
- Program of Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON, M3S 1A8, Canada
| |
Collapse
|
11
|
Nguyen L. Updates on Disease Mechanisms and Therapeutics for Amyotrophic Lateral Sclerosis. Cells 2024; 13:888. [PMID: 38891021 PMCID: PMC11172142 DOI: 10.3390/cells13110888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/08/2024] [Accepted: 05/15/2024] [Indexed: 06/20/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS), or Lou Gehrig's disease, is a motor neuron disease. In ALS, upper and lower motor neurons in the brain and spinal cord progressively degenerate during the course of the disease, leading to the loss of the voluntary movement of the arms and legs. Since its first description in 1869 by a French neurologist Jean-Martin Charcot, the scientific discoveries on ALS have increased our understanding of ALS genetics, pathology and mechanisms and provided novel therapeutic strategies. The goal of this review article is to provide a comprehensive summary of the recent findings on ALS mechanisms and related therapeutic strategies to the scientific audience. Several highlighted ALS research topics discussed in this article include the 2023 FDA approved drug for SOD1 ALS, the updated C9orf72 GGGGCC repeat-expansion-related mechanisms and therapeutic targets, TDP-43-mediated cryptic splicing and disease markers and diagnostic and therapeutic options offered by these recent discoveries.
Collapse
Affiliation(s)
- Lien Nguyen
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL 32610, USA;
- Center for NeuroGenetics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Genetics Institute, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
12
|
Fu RH, Chen HJ, Hong SY. Glycine-Alanine Dipeptide Repeat Protein from C9-ALS Interacts with Sulfide Quinone Oxidoreductase (SQOR) to Induce the Activity of the NLRP3 Inflammasome in HMC3 Microglia: Irisflorentin Reverses This Interaction. Antioxidants (Basel) 2023; 12:1896. [PMID: 37891975 PMCID: PMC10604625 DOI: 10.3390/antiox12101896] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/07/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal rare disease of progressive degeneration of motor neurons. The most common genetic mutation in ALS is the hexanucleotide repeat expansion (HRE) located in the first intron of the C9orf72 gene (C9-ALS). HRE can produce dipeptide repeat proteins (DPRs) such as poly glycine-alanine (GA) in a repeat-associated non-ATG (RAN) translation. GA-DPR has been shown to be toxic to motor neurons in various biological models. However, its effects on microglia involved in C9-ALS have not been reported. Here, we show that GA-DPR (GA50) activates the NLR family pyrin domain containing 3 (NLRP3) inflammasome in a human HMC3 microglia model. MCC950 (specific inhibitor of the NLRP3) treatment can abrogate this activity. Next, using yeast two-hybrid screening, we identified sulfide quinone oxidoreductase (SQOR) as a GA50 interacting protein. SQOR knockdown in HMC3 cells can significantly induce the activity of the NLRP3 inflammasome by upregulating the level of intracellular reactive oxygen species and the cytoplasmic escape of mitochondrial DNA. Furthermore, we obtained irisflorentin as an effective blocker of the interaction between SQOR and GA50, thus inhibiting NLRP3 inflammasome activity in GA50-expressing HMC3 cells. These results imply the association of GA-DPR, SQOR, and NLRP3 inflammasomes in microglia and establish a treatment strategy for C9-ALS with irisflorentin.
Collapse
Affiliation(s)
- Ru-Huei Fu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- Translational Medicine Research Center, China Medical University Hospital, Taichung 40447, Taiwan
- Ph.D. Program for Aging, China Medical University, Taichung 40402, Taiwan
| | - Hui-Jye Chen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
| | - Syuan-Yu Hong
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- Department of Medicine, School of Medicine, China Medical University, Taichung 40447, Taiwan
- Division of Pediatric Neurology, China Medical University Children’s Hospital, Taichung 40447, Taiwan
| |
Collapse
|