Glial Cell Dysfunction in C9orf72-Related Amyotrophic Lateral Sclerosis and Frontotemporal Dementia
Abstract
:1. Introduction
2. Overview of Pathogenic Mechanisms Underlying C9orf72 Repeat Expansion Mutation in ALS/FTD
2.1. Loss of Function Mechanisms
2.2. Challenges in Loss-of-Function Theory
2.3. Gain-of-Function Mechanisms
- Binding of RNA foci to RNA-binding proteins (RBPs), forming neurotoxic aggregates [26];
- Impairment of liquid–liquid phase separation (LLPS) through interaction with low complexity domain (LCD) proteins in nucleoli and stress granules [97,106,107,108]. LLPS of key protein and nucleic acid scaffolds play an important role in the biogenesis of diverse membrane-less organelles (e.g., P granules and stress granules in the cytoplasm and nucleoli and paraspeckles in the nucleus) that are essential organizers of subcellular biochemistry, controlling the information processing from genotype to phenotype [109];
- Impairment of ribosomal RNA maturation and abnormal splicing. A pioneering study by Kwon et al. (2014) indicated that exogenous administration of synthetic poly-(GR)20 and poly-(PR)20 to human astrocytes led to their accumulation in the nucleus and binding to the LCD of hnRNPA2, causing aberrant pre-mRNA splicing and impaired rRNA biogenesis [104];
3. Neuroinflammation in C9orf72 ALS/FTD: Glial Cells Dysfunction
3.1. Glial Cells in the Central Nervous System
3.2. A Role for Glial Cells in C9orf72 ALS/FTD
3.2.1. Evidence for Gliosis in C9orf72 ALS/FTD
3.2.2. C9orf72 Pathologic Hallmarks in Glial Cells
3.2.3. Toxic Effects of C9orf72 Glial Cells
4. Biomarkers in C9orf72 ALS/FTD
4.1. Non-Inflammatory Biomarkers
4.2. Inflammatory/Glial Biomarkers
4.3. Imaging-Based Markers
5. Therapeutic Approaches: Focus on Glial Cells
6. Conclusions and Perspectives
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Zucchi, E.; Ticozzi, N.; Mandrioli, J. Psychiatric Symptoms in Amyotrophic Lateral Sclerosis: Beyond a Motor Neuron Disorder. Front. Neurosci. 2019, 13, 175. [Google Scholar] [CrossRef] [Green Version]
- Ghasemi, M.; Brown, R.H., Jr. Genetics of Amyotrophic Lateral Sclerosis. Cold Spring Harb. Perspect. Med. 2018, 8, a024125. [Google Scholar] [CrossRef] [PubMed]
- Al-Sarraj, S.; King, A.; Troakes, C.; Smith, B.; Maekawa, S.; Bodi, I.; Rogelj, B.; Al-Chalabi, A.; Hortobágyi, T.; Shaw, C.E. p62 positive, TDP-43 negative, neuronal cytoplasmic and intranuclear inclusions in the cerebellum and hippocampus define the pathology of C9orf72-linked FTLD and MND/ALS. Acta Neuropathol. 2011, 122, 691–702. [Google Scholar] [CrossRef] [PubMed]
- Cooper-Knock, J.; Hewitt, C.; Highley, J.R.; Brockington, A.; Milano, A.; Man, S.; Martindale, J.; Hartley, J.; Walsh, T.; Gelsthorpe, C.; et al. Clinico-pathological features in amyotrophic lateral sclerosis with expansions in C9ORF72. Brain 2012, 135, 751–764. [Google Scholar] [CrossRef] [PubMed]
- Schipper, L.J.; Raaphorst, J.; Aronica, E.; Baas, F.; De Haan, R.J.; De Visser, M.; Troost, D. Prevalence of brain and spinal cord inclusions, including dipeptide repeat proteins, in patients with the C9ORF72 hexanucleotide repeat expansion: A systematic neuropathological review. Neuropathol. Appl. Neurobiol. 2016, 42, 547–560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Simón-Sánchez, J.; Dopper, E.G.P.; Cohn-Hokke, P.E.; Hukema, R.K.; Nicolaou, N.; Seelaar, H.; De Graaf, J.R.A.; De Koning, I.; Van Schoor, N.M.; Deeg, D.J.H.; et al. The clinical and pathological phenotype of C9ORF72 hexanucleotide repeat expansions. Brain 2012, 135, 723–735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jiang, J.; Ravits, J.M. Pathogenic Mechanisms and Therapy Development for C9orf72 Amyotrophic Lateral Sclerosis/Frontotemporal Dementia. Neurotherapeutics 2019, 16, 1115–1132. [Google Scholar] [CrossRef] [PubMed]
- DeJesus-Hernandez, M.; MacKenzie, I.R.; Boeve, B.F.; Boxer, A.L.; Baker, M.; Rutherford, N.J.; Nicholson, A.M.; Finch, N.A.; Flynn, H.; Adamson, J.; et al. Expanded GGGGCC Hexanucleotide Repeat in Noncoding Region of C9ORF72 Causes Chromosome 9p-Linked FTD and ALS. Neuron 2011, 72, 245–256. [Google Scholar] [CrossRef] [Green Version]
- Renton, A.E.; Majounie, E.; Waite, A.; Simón-Sánchez, J.; Rollinson, S.; Gibbs, J.R.; Schymick, J.C.; Laaksovirta, H.; Van Swieten, J.C.; Myllykangas, L.; et al. A Hexanucleotide Repeat Expansion in C9ORF72 Is the Cause of Chromosome 9p21-Linked ALS-FTD. Neuron 2011, 72, 257–268. [Google Scholar] [CrossRef] [Green Version]
- Gijselinck, I.; Van Langenhove, T.; Van Der Zee, J.; Sleegers, K.; Philtjens, S.; Kleinberger, G.; Janssens, J.; Bettens, K.; Van Cauwenberghe, C.; Pereson, S.; et al. A C9orf72 promoter repeat expansion in a Flanders-Belgian cohort with disorders of the frontotemporal lobar degeneration-amyotrophic lateral sclerosis spectrum: A gene identification study. Lancet Neurol. 2012, 11, 54–65. [Google Scholar] [CrossRef]
- Nguyen, H.P.; Van Broeckhoven, C.; Van Der Zee, J. ALS Genes in the Genomic Era and their Implications for FTD. Trends Genet. 2018, 34, 404–423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Haeusler, A.R.; Donnelly, C.J.; Rothstein, J.D. The expanding biology of the C9orf72 nucleotide repeat expansion in neurodegenerative disease. Nat. Rev. Neurosci. 2016, 17, 383–395. [Google Scholar] [CrossRef] [PubMed]
- Cammack, A.J.; Atassi, N.; Hyman, T.; Van Den Berg, L.H.; Harms, M.; Baloh, R.H.; Brown, R.H.; Van Es, M.A.; Veldink, J.H.; De Vries, B.S.; et al. Prospective natural history study of C9orf72 ALS clinical characteristics and biomarkers. Neurology 2019, 93, e1605–e1617. [Google Scholar] [CrossRef] [PubMed]
- Byrne, S.C.; Elamin, M.; Bede, P.; Shatunov, A.; Walsh, C.; Corr, B.; Heverin, M.; Jordan, N.; Kenna, K.; Lynch, C.; et al. Cognitive and clinical characteristics of patients with amyotrophic lateral sclerosis carrying a C9orf72 repeat expansion: A population-based cohort study. Lancet Neurol. 2012, 11, 232–240. [Google Scholar] [CrossRef] [Green Version]
- Millecamps, S.; Boillée, S.; Le Ber, I.; Seilhean, D.; Teyssou, E.; Giraudeau, M.; Moigneu, C.; Vandenberghe, N.; Danel-Brunaud, V.; Corcia, P.; et al. Phenotype difference between ALS patients with expanded repeats in C9ORF72 and patients with mutations in other ALS-related genes. J. Med. Genet. 2012, 49, 258–263. [Google Scholar] [CrossRef]
- Chiò, A.; Borghero, G.; Restagno, G.; Mora, G.; Drepper, C.; Traynor, B.J.; Sendtner, M.; Brunetti, M.; Ossola, I.; Calvo, A.; et al. Clinical characteristics of patients with familial amyotrophic lateral sclerosis carrying the pathogenic GGGGCC hexanucleotide repeat expansion of C9ORF72. Brain 2012, 135, 784–793. [Google Scholar] [CrossRef]
- Irwin, D.J.; McMillan, C.T.; Brettschneider, J.; Libon, D.J.; Powers, J.; Rascovsky, K.; Toledo, J.B.; Boller, A.; Bekisz, J.; Chandrasekaran, K.; et al. Cognitive decline and reduced survival in C9orf72 expansion frontotemporal degeneration and amyotrophic lateral sclerosis. J. Neurol. Neurosurg. Psychiatry 2013, 84, 163–169. [Google Scholar] [CrossRef] [Green Version]
- Snowden, J.S.; Adams, J.; Harris, J.; Thompson, J.C.; Rollinson, S.; Richardson, A.; Jones, M.J.; Neary, D.; Mann, D.M.; Pickering-Brown, S. Distinct clinical and pathological phenotypes in frontotemporal dementia associated with MAPT, PGRN and C9orf72 mutations. Amyotroph. Lateral Scler. Front. Degener. 2015, 16, 497–505. [Google Scholar] [CrossRef]
- Picher-Martel, V.; Valdmanis, P.N.; Gould, P.V.; Julien, J.-P.; Dupre, N. From animal models to human disease: A genetic approach for personalized medicine in ALS. Acta Neuropathol. Commun. 2016, 4, 1–29. [Google Scholar] [CrossRef]
- Ciura, S.; Lattante, S.; Le Ber, I.; Latouche, M.; Tostivint, H.; Brice, A.; Kabashi, E. Loss of function of C9orf72 causes motor deficits in a zebrafish model of Amyotrophic Lateral Sclerosis. Ann. Neurol. 2013, 74, 180–187. [Google Scholar] [CrossRef]
- Fratta, P.; Poulter, M.; Lashley, T.; Rohrer, J.D.; Polke, J.M.; Beck, J.; Ryan, N.; Hensman, D.; Mizielinska, S.; Waite, A.J.; et al. Homozygosity for the C9orf72 GGGGCC repeat expansion in frontotemporal dementia. Acta Neuropathol. 2013, 126, 401–409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Belzil, V.V.; Bauer, P.O.; Prudencio, M.; Gendron, T.F.; Stetler, C.T.; Yan, I.K.; Pregent, L.; Daughrity, L.; Baker, M.C.; Rademakers, R.; et al. Reduced C9orf72 gene expression in c9FTD/ALS is caused by histone trimethylation, an epigenetic event detectable in blood. Acta Neuropathol. 2013, 126, 895–905. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Waite, A.J.; Bäumer, D.; East, S.; Neal, J.; Morris, H.R.; Ansorge, O.; Blake, D.J. Reduced C9orf72 protein levels in frontal cortex of amyotrophic lateral sclerosis and frontotemporal degeneration brain with the C9ORF72 hexanucleotide repeat expansion. Neurobiol. Aging 2014, 35, 1779.e5–1779.e13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Van Blitterswijk, M.; Gendron, T.F.; Baker, M.C.; DeJesus-Hernandez, M.; Finch, N.A.; Brown, P.H.; Daughrity, L.M.; Murray, M.E.; Heckman, M.G.; Jiang, J.; et al. Novel clinical associations with specific C9ORF72 transcripts in patients with repeat expansions in C9ORF72. Acta Neuropathol. 2015, 130, 863–876. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Donnelly, C.J.; Zhang, P.-W.; Pham, J.T.; Haeusler, A.R.; Mistry, N.A.; Vidensky, S.; Daley, E.L.; Poth, E.M.; Hoover, B.; Fines, D.M.; et al. RNA Toxicity from the ALS/FTD C9ORF72 Expansion Is Mitigated by Antisense Intervention. Neuron 2013, 80, 415–428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peters, O.M.; Ghasemi, M.; Brown, R.H., Jr. Emerging mechanisms of molecular pathology in ALS. J. Clin. Investig. 2015, 125, 1767–1779. [Google Scholar] [CrossRef]
- Tran, H.; Almeida, S.; Moore, J.M.; Gendron, T.F.; Chalasani, U.; Lu, Y.; Du, X.; Nickerson, J.A.; Petrucelli, L.; Weng, Z.; et al. Differential Toxicity of Nuclear RNA Foci versus Dipeptide Repeat Proteins in a Drosophila Model of C9ORF72 FTD/ALS. Neuron 2015, 87, 1207–1214. [Google Scholar] [CrossRef] [Green Version]
- Rizzu, P.; Blauwendraat, C.; Heetveld, S.; Lynes, E.M.; Castillo-Lizardo, M.; Dhingra, A.; Pyz, E.; Hobert, M.; Synofzik, M.; Simón-Sánchez, J.; et al. C9orf72 is differentially expressed in the central nervous system and myeloid cells and consistently reduced in C9orf72, MAPT and GRN mutation carriers. Acta Neuropathol. Commun. 2016, 4, 37. [Google Scholar] [CrossRef] [Green Version]
- Frick, P.; Sellier, C.; MacKenzie, I.R.A.; Cheng, C.-Y.; Tahraoui-Bories, J.; Martinat, C.; Pasterkamp, R.J.; Prudlo, J.; Edbauer, D.; Oulad-Abdelghani, M.; et al. Novel antibodies reveal presynaptic localization of C9orf72 protein and reduced protein levels in C9orf72 mutation carriers. Acta Neuropathol. Commun. 2018, 6, 72. [Google Scholar] [CrossRef] [Green Version]
- Xiao, S.; Macnair, L.; McGoldrick, P.; McKeever, P.M.; McLean, J.R.; Zhang, M.; Keith, J.; Zinman, L.; Rogaeva, E.; Robertson, J. Isoform-specific antibodies reveal distinct subcellular localizations of C9orf72 in amyotrophic lateral sclerosis. Ann. Neurol. 2015, 78, 568–583. [Google Scholar] [CrossRef]
- Atkinson, R.; Fernandez-Martos, C.M.; Atkin, J.D.; Vickers, J.C.; King, A.E. C9ORF72 expression and cellular localization over mouse development. Acta Neuropathol. Commun. 2015, 3, 59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mori, K.; Weng, S.-M.; Arzberger, T.; May, S.; Rentzsch, K.; Kremmer, E.; Schmid, B.; Kretzschmar, H.A.; Cruts, M.; Van Broeckhoven, C.; et al. The C9orf72 GGGGCC Repeat Is Translated into Aggregating Dipeptide-Repeat Proteins in FTLD/ALS. Science 2013, 339, 1335–1338. [Google Scholar] [CrossRef] [PubMed]
- Almeida, S.; Gascon, E.; Tran, H.; Chou, H.J.; Gendron, T.F.; DeGroot, S.; Tapper, A.R.; Sellier, C.; Charlet-Berguerand, N.; Karydas, A.; et al. Modeling key pathological features of frontotemporal dementia with C9ORF72 repeat expansion in iPSC-derived human neurons. Acta Neuropathol. 2013, 126, 385–399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shi, Y.; Lin, S.; Staats, K.A.; Li, Y.; Chang, W.-H.; Hung, S.-T.; Hendricks, E.; Linares, G.R.; Wang, Y.; Son, E.Y.; et al. Haploinsufficiency leads to neurodegeneration in C9ORF72 ALS/FTD human induced motor neurons. Nat. Med. 2018, 24, 313–325. [Google Scholar] [CrossRef] [PubMed]
- Xi, Z.; Zinman, L.; Moreno, D.; Schymick, J.; Liang, Y.; Sato, C.; Zheng, Y.; Ghani, M.; Dib, S.; Keith, J.; et al. Hypermethylation of the CpG Island Near the G4C2 Repeat in ALS with a C9orf72 Expansion. Am. J. Hum. Genet. 2013, 92, 981–989. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, D.; Iyer, L.M.; He, F.; Aravind, L. Discovery of Novel DENN Proteins: Implications for the Evolution of Eukaryotic Intracellular Membrane Structures and Human Disease. Front. Genet. 2012, 3, 283. [Google Scholar] [CrossRef] [Green Version]
- Webster, C.P.; Smith, E.F.; Bauer, C.S.; Moller, A.; Hautbergue, G.M.; Ferraiuolo, L.; Myszczynska, M.A.; Higginbottom, A.; Walsh, M.J.; Whitworth, A.J.; et al. The C9orf72 protein interacts with Rab1a and the ULK 1 complex to regulate initiation of autophagy. EMBO J. 2016, 35, 1656–1676. [Google Scholar] [CrossRef]
- Webster, C.P.; Smith, E.F.; Grierson, A.J.; De Vos, K.J. C9orf72 plays a central role in Rab GTPase-dependent regulation of autophagy. Small GTPases 2018, 9, 399–408. [Google Scholar] [CrossRef] [Green Version]
- Iyer, S.; Subramanian, V.; Acharya, K.R. C9orf72, a protein associated with amyotrophic lateral sclerosis (ALS) is a guanine nucleotide exchange factor. PeerJ 2018, 6, e5815. [Google Scholar] [CrossRef] [Green Version]
- Farg, M.A.; Sundaramoorthy, V.; Sultana, J.M.; Yang, S.; Atkinson, R.A.; Levina, V.; Halloran, M.A.; Gleeson, P.A.; Blair, I.P.; Soo, K.Y.; et al. C9ORF72, implicated in amytrophic lateral sclerosis and frontotemporal dementia, regulates endosomal trafficking. Hum. Mol. Genet. 2014, 23, 3579–3595. [Google Scholar] [CrossRef]
- Elden, A.C.; Kim, H.-J.; Hart, M.P.; Chen-Plotkin, A.S.; Johnson, B.S.; Fang, X.; Armakola, M.; Geser, F.; Greene, R.; Lu, M.M.; et al. Ataxin-2 intermediate-length polyglutamine expansions are associated with increased risk for ALS. Nature 2010, 466, 1069–1075. [Google Scholar] [CrossRef] [PubMed]
- Daoud, H.; Belzil, V.; Martins, S.; Sabbagh, M.; Provencher, P.; Lacomblez, L.; Meininger, V.; Camu, W.; Dupré, N.; Dion, P.A.; et al. Association of Long ATXN2 CAG Repeat Sizes with Increased Risk of Amyotrophic Lateral Sclerosis. Arch. Neurol. 2011, 68, 739–742. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ross, O.A.; Rutherford, N.J.; Baker, M.; Soto-Ortolaza, A.I.; Carrasquillo, M.M.; DeJesus-Hernandez, M.; Adamson, J.; Li, M.; Volkening, K.; Finger, E.; et al. Ataxin-2 repeat-length variation and neurodegeneration. Hum. Mol. Genet. 2011, 20, 3207–3212. [Google Scholar] [CrossRef] [PubMed]
- Lattante, S.; Millecamps, S.; Stevanin, G.; Rivaud-Péchoux, S.; Moigneu, C.; Camuzat, A.; Da Barroca, S.; Mundwiller, E.; Couarch, P.; Salachas, F.; et al. Contribution of ATXN2 intermediary polyQ expansions in a spectrum of neurodegenerative disorders. Neurology 2014, 83, 990–995. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sellier, C.; Campanari, M.-L.; Corbier, C.J.; Gaucherot, A.; Kolb-Cheynel, I.; Oulad-Abdelghani, M.; Ruffenach, F.; Page, A.; Ciura, S.; Kabashi, E.; et al. Loss of C9ORF72 impairs autophagy and synergizes with polyQ Ataxin-2 to induce motor neuron dysfunction and cell death. EMBO J. 2016, 35, 1276–1297. [Google Scholar] [CrossRef] [PubMed]
- Sudria-Lopez, E.; Koppers, M.; De Wit, M.; Van Der Meer, C.; Westeneng, H.-J.; Zundel, C.A.C.; Youssef, S.A.; Harkema, L.; De Bruin, A.; Veldink, J.H.; et al. Full ablation of C9orf72 in mice causes immune system-related pathology and neoplastic events but no motor neuron defects. Acta Neuropathol. 2016, 132, 145–147. [Google Scholar] [CrossRef] [Green Version]
- Aoki, Y.; Manzano, R.; Lee, Y.; Dafinca, R.; Aoki, M.; Douglas, A.G.L.; Varela, M.A.; Sathyaprakash, C.; Scaber, J.; Barbagallo, P.; et al. C9orf72 and RAB7L1 regulate vesicle trafficking in amyotrophic lateral sclerosis and frontotemporal dementia. Brain 2017, 140, 887–897. [Google Scholar] [CrossRef]
- Amick, J.; Roczniak-Ferguson, A.; Ferguson, S.M. C9orf72 binds SMCR8, localizes to lysosomes, and regulates mTORC1 signaling. Mol. Biol. Cell 2016, 27, 3040–3051. [Google Scholar] [CrossRef]
- Burk, K.; Pasterkamp, R.J. Disrupted neuronal trafficking in amyotrophic lateral sclerosis. Acta Neuropathol. 2019, 137, 859–877. [Google Scholar] [CrossRef] [Green Version]
- Huber, L.A.; Pimplikar, S.; Parton, R.G.; Virta, H.; Zerial, M.; Simons, K. Rab8, a small GTPase involved in vesicular traffic between the TGN and the basolateral plasma membrane. J. Cell Biol. 1993, 123, 35–45. [Google Scholar] [CrossRef]
- King, A.E.; Woodhouse, A.; Kirkcaldie, M.T.; Vickers, J.C. Excitotoxicity in ALS: Overstimulation, or overreaction? Exp. Neurol. 2016, 275 Pt 1, 162–171. [Google Scholar] [CrossRef]
- Bursch, F.; Kalmbach, N.; Naujock, M.; Staege, S.; Eggenschwiler, R.; Abo-Rady, M.; Japtok, J.; Guo, W.; Hensel, N.; Reinhardt, P.; et al. Altered calcium dynamics and glutamate receptor properties in iPSC derived motor neurons from ALS patients with C9orf72, FUS, SOD1 or TDP43 mutations. Hum. Mol. Genet. 2019, 28, 2835–2850. [Google Scholar] [CrossRef] [PubMed]
- Selvaraj, B.T.; Livesey, M.R.; Zhao, C.; Gregory, J.M.; James, O.T.; Cleary, E.M.; Chouhan, A.K.; Gane, A.B.; Perkins, E.M.; Dando, O.; et al. C9ORF72 repeat expansion causes vulnerability of motor neurons to Ca2+-permeable AMPA receptor-mediated excitotoxicity. Nat. Commun. 2018, 9, 347. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Moore, S.; Alsop, E.; Lorenzini, I.; Starr, A.; Rabichow, B.E.; Mendez, E.; Levy, J.L.; Burciu, C.; Reiman, R.; Chew, J.; et al. ADAR2 mislocalization and widespread RNA editing aberrations in C9orf72-mediated ALS/FTD. Acta Neuropathol. 2019, 138, 49–65. [Google Scholar] [CrossRef]
- Hideyama, T.; Yamashita, T.; Suzuki, T.; Tsuji, S.; Higuchi, M.; Seeburg, P.H.; Takahashi, R.; Misawa, H.; Kwak, S. Induced Loss of ADAR2 Engenders Slow Death of Motor Neurons from Q/R Site-Unedited GluR2. J. Neurosci. 2010, 30, 11917–11925. [Google Scholar] [CrossRef] [PubMed]
- Shi, Y.; Hung, S.-T.; Rocha, G.; Lin, S.; Linares, G.R.; Staats, K.A.; Seah, C.; Wang, Y.; Chickering, M.; Lai, J.; et al. Identification and therapeutic rescue of autophagosome and glutamate receptor defects in C9ORF72 and sporadic ALS neurons. JCI Insight 2019, 4. [Google Scholar] [CrossRef]
- Koppers, M.; Blokhuis, A.M.; Westeneng, H.; Terpstra, M.L.; Zundel, C.A.C.; De Sá, R.V.; Schellevis, R.D.; Waite, A.J.; Blake, D.J.; Veldink, J.H.; et al. C 9orf72 ablation in mice does not cause motor neuron degeneration or motor deficits. Ann. Neurol. 2015, 78, 426–438. [Google Scholar] [CrossRef] [Green Version]
- Jiang, J.; Zhu, Q.; Gendron, T.F.; Saberi, S.; McAlonis-Downes, M.; Seelman, A.; Stauffer, J.E.; Jafar-Nejad, P.; Drenner, K.; Schulte, D.; et al. Gain of Toxicity from ALS/FTD-Linked Repeat Expansions in C9ORF72 Is Alleviated by Antisense Oligonucleotides Targeting GGGGCC-Containing RNAs. Neuron 2016, 90, 535–550. [Google Scholar] [CrossRef]
- Atanasio, A.; Decman, V.; White, D.; Ramos, M.; Ikiz, B.; Lee, H.-C.; Siao, C.-J.; Brydges, S.; LaRosa, E.; Bai, Y.; et al. C9orf72 ablation causes immune dysregulation characterized by leukocyte expansion, autoantibody production and glomerulonephropathy in mice. Sci. Rep. 2016, 6, 23204. [Google Scholar] [CrossRef]
- O’Rourke, J.G.; Bogdanik, L.; Yáñez, A.; Lall, D.; Wolf, A.J.; Muhammad, A.K.M.G.; Ho, R.; Carmona, S.; Vit, J.P.; Zarrow, J.; et al. C9orf72 is required for proper macrophage and microglial function in mice. Science 2016, 351, 1324–1329. [Google Scholar] [CrossRef] [Green Version]
- Burberry, A.; Suzuki, N.; Wang, J.-Y.; Moccia, R.; Mordes, D.A.; Stewart, M.H.; Suzuki-Uematsu, S.; Ghosh, S.; Singh, A.; Merkle, F.T.; et al. Loss-of-function mutations in the C9ORF72 mouse ortholog cause fatal autoimmune disease. Sci. Transl. Med. 2016, 8, 347ra93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ugolino, J.; Ji, Y.J.; Conchina, K.; Chu, J.; Nirujogi, R.S.; Pandey, A.; Brady, N.R.; Hamacher-Brady, A.; Wang, J. Loss of C9orf72 Enhances Autophagic Activity via Deregulated mTOR and TFEB Signaling. PLoS Genet. 2016, 12, e1006443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sullivan, P.M.; Zhou, X.; Robins, A.M.; Paushter, D.H.; Kim, D.; Smolka, M.B.; Hu, F. The ALS/FTLD associated protein C9orf72 associates with SMCR8 and WDR41 to regulate the autophagy-lysosome pathway. Acta Neuropathol. Commun. 2016, 4, 51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Balendra, R.; Isaacs, A.M. C9orf72-mediated ALS and FTD: Multiple pathways to disease. Nat. Rev. Neurol. 2018, 14, 544–558. [Google Scholar] [CrossRef] [PubMed]
- Robberecht, W.; Philips, T. The changing scene of amyotrophic lateral sclerosis. Nat. Rev. Neurosci. 2013, 14, 248–264. [Google Scholar] [CrossRef]
- Chew, J.; Gendron, T.F.; Prudencio, M.; Sasaguri, H.; Zhang, Y.-J.; Castanedes-Casey, M.; Lee, C.W.; Jansen-West, K.; Kurti, A.; Murray, M.E.; et al. C9ORF72 repeat expansions in mice cause TDP-43 pathology, neuronal loss, and behavioral deficits. Science 2015, 348, 1151–1154. [Google Scholar] [CrossRef] [Green Version]
- Gendron, T.F.; Bieniek, K.F.; Zhang, Y.-J.; Jansen-West, K.; Ash, P.E.A.; Caulfield, T.; Daughrity, L.; Dunmore, J.H.; Castanedes-Casey, M.; Chew, J.; et al. Antisense transcripts of the expanded C9ORF72 hexanucleotide repeat form nuclear RNA foci and undergo repeat-associated non-ATG translation in c9FTD/ALS. Acta Neuropathol. 2013, 126, 829–844. [Google Scholar] [CrossRef] [Green Version]
- Mori, K.; Arzberger, T.; Grässer, F.A.; Gijselinck, I.; May, S.; Rentzsch, K.; Weng, S.-M.; Schludi, M.H.; Van Der Zee, J.; Cruts, M.; et al. Bidirectional transcripts of the expanded C9orf72 hexanucleotide repeat are translated into aggregating dipeptide repeat proteins. Acta Neuropathol. 2013, 126, 881–893. [Google Scholar] [CrossRef]
- Zu, T.; Liu, Y.; Bañez-Coronel, M.; Reid, T.; Pletnikova, O.; Lewis, J.; Miller, T.M.; Harms, M.B.; Falchook, A.E.; Subramony, S.H.; et al. RAN proteins and RNA foci from antisense transcripts in C9ORF72 ALS and frontotemporal dementia. Proc. Natl. Acad. Sci. USA 2013, 110, E4968–E4977. [Google Scholar] [CrossRef] [Green Version]
- Lagier-Tourenne, C.; Baughn, M.; Rigo, F.; Sun, S.; Liu, P.; Li, H.-R.; Jiang, J.; Watt, A.T.; Chun, S.; Katz, M.; et al. Targeted degradation of sense and antisense C9orf72 RNA foci as therapy for ALS and frontotemporal degeneration. Proc. Natl. Acad. Sci. USA 2013, 110, E4530–E4539. [Google Scholar] [CrossRef] [Green Version]
- Sareen, D.; O’Rourke, J.G.; Meera, P.; Muhammad, A.K.M.G.; Grant, S.; Simpkinson, M.; Bell, S.; Carmona, S.; Ornelas, L.; Sahabian, A.; et al. Targeting RNA Foci in iPSC-Derived Motor Neurons from ALS Patients with a C9ORF72 Repeat Expansion. Sci. Transl. Med. 2013, 5, 208ra149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- La Spada, A.R.; Taylor, J.P. Repeat expansion disease: Progress and puzzles in disease pathogenesis. Nat. Rev. Genet. 2010, 11, 247–258. [Google Scholar] [CrossRef] [PubMed]
- Fratta, P.; Mizielinska, S.; Nicoll, A.J.; Zloh, M.; Fisher, E.M.C.; Parkinson, G.N.; Isaacs, A.M. C9orf72 hexanucleotide repeat associated with amyotrophic lateral sclerosis and frontotemporal dementia forms RNA G-quadruplexes. Sci. Rep. 2012, 2, srep01016. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Goodrich, K.J.; Conlon, E.G.; Gao, J.; Erbse, A.H.; Manley, J.L.; Cech, T.R. C9orf72 and triplet repeat disorder RNAs: G-quadruplex formation, binding to PRC2 and implications for disease mechanisms. RNA 2019, 25, 935–947. [Google Scholar] [CrossRef] [Green Version]
- Taylor, J.P. Neurodegenerative diseases: G-quadruplex poses quadruple threat. Nature 2014, 507, 175–177. [Google Scholar] [CrossRef]
- Su, Z.; Zhang, Y.; Gendron, T.F.; Bauer, P.O.; Chew, J.; Yang, W.-Y.; Fostvedt, E.; Jansen-West, K.; Belzil, V.V.; Desaro, P.; et al. Discovery of a Biomarker and Lead Small Molecules to Target r(GGGGCC)-Associated Defects in c9FTD/ALS. Neuron 2014, 83, 1043–1050. [Google Scholar] [CrossRef] [Green Version]
- Haeusler, A.R.; Donnelly, C.J.; Periz, G.; Simko, E.A.J.; Shaw, P.G.; Kim, M.-S.; Maragakis, N.J.; Troncoso, J.C.; Pandey, A.; Sattler, R.; et al. C9orf72 nucleotide repeat structures initiate molecular cascades of disease. Nature 2014, 507, 195–200. [Google Scholar] [CrossRef]
- Reddy, K.; Zamiri, B.; Stanley, S.Y.R.; MacGregor, R.B.; Pearson, C.E. The Disease-associated r(GGGGCC) Repeat from the C9orf72 Gene Forms Tract Length-dependent Uni- and Multimolecular RNA G-quadruplex Structures. J. Biol. Chem. 2013, 288, 9860–9866. [Google Scholar] [CrossRef] [Green Version]
- Zhou, B.; Geng, Y.; Liu, C.; Miao, H.; Ren, Y.; Xu, N.; Shi, X.; You, Y.; Lee, T.; Zhu, G. Characterizations of distinct parallel and antiparallel G-quadruplexes formed by two-repeat ALS and FTD related GGGGCC sequence. Sci. Rep. 2018, 8, 2366. [Google Scholar] [CrossRef] [Green Version]
- Brčić, J.; Plavec, J. NMR structure of a G-quadruplex formed by four d(G4C2) repeats: Insights into structural polymorphism. Nucleic Acids Res. 2018, 46, 11605–11617. [Google Scholar] [CrossRef]
- Skourti-Stathaki, K.; Proudfoot, N.J.; Gromak, N. Human Senataxin Resolves RNA/DNA Hybrids Formed at Transcriptional Pause Sites to Promote Xrn2-Dependent Termination. Mol. Cell 2011, 42, 794–805. [Google Scholar] [CrossRef] [PubMed]
- Yüce, Ö.; West, S.C. Senataxin, Defective in the Neurodegenerative Disorder Ataxia with Oculomotor Apraxia 2, Lies at the Interface of Transcription and the DNA Damage Response. Mol. Cell. Biol. 2012, 33, 406–417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vatovec, S.; Kovanda, A.; Rogelj, B. Unconventional features of C9ORF72 expanded repeat in amyotrophic lateral sclerosis and frontotemporal lobar degeneration. Neurobiol. Aging 2014, 35, 2421.e1–2421.e12. [Google Scholar] [CrossRef] [PubMed]
- Kovanda, A.; Zalar, M.; Šket, P.; Plavec, J.; Rogelj, B. Anti-sense DNA d(GGCCCC)n expansions in C9ORF72 form i-motifs and protonated hairpins. Sci. Rep. 2015, 5, 17944. [Google Scholar] [CrossRef]
- Zhang, Y.; Roland, C.; Sagui, C. Structure and Dynamics of DNA and RNA Double Helices Obtained from the GGGGCC and CCCCGG Hexanucleotide Repeats That Are the Hallmark of C9FTD/ALS Diseases. ACS Chem. Neurosci. 2017, 8, 578–591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ash, P.E.; Bieniek, K.F.; Gendron, T.F.; Caulfield, T.; Lin, W.-L.; DeJesus-Hernandez, M.; Van Blitterswijk, M.M.; Jansen-West, K.; Paul, J.W., III; Rademakers, R.; et al. Unconventional Translation of C9ORF72 GGGGCC Expansion Generates Insoluble Polypeptides Specific to c9FTD/ALS. Neuron 2013, 77, 639–646. [Google Scholar] [CrossRef] [Green Version]
- Wen, X.; Tan, W.; Westergard, T.; Krishnamurthy, K.; Markandaiah, S.S.; Shi, Y.; Lin, S.; Shneider, N.A.; Monaghan, J.; Pandey, U.B.; et al. Antisense Proline-Arginine RAN Dipeptides Linked to C9ORF72-ALS/FTD Form Toxic Nuclear Aggregates that Initiate In Vitro and In Vivo Neuronal Death. Neuron 2014, 84, 1213–1225. [Google Scholar] [CrossRef] [Green Version]
- Tao, Z.; Wang, H.; Xia, Q.; Li, K.; Li, K.; Jiang, X.; Xu, G.; Wang, G.; Ying, Z. Nucleolar stress and impaired stress granule formation contribute to C9orf72 RAN translation-induced cytotoxicity. Hum. Mol. Genet. 2015, 24, 2426–2441. [Google Scholar] [CrossRef] [Green Version]
- Kanekura, K.; Yagi, T.; Cammack, A.J.; Mahadevan, J.; Kuroda, M.; Harms, M.B.; Miller, T.M.; Urano, F. Poly-dipeptides encoded by the C9ORF72 repeats block global protein translation. Hum. Mol. Genet. 2016, 25, 1803–1813. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Y.-J.; Jansen-West, K.; Xu, Y.-F.; Gendron, T.F.; Bieniek, K.F.; Lin, W.-L.; Sasaguri, H.; Caulfield, T.; Hubbard, J.; Daughrity, L.; et al. Aggregation-prone c9FTD/ALS poly(GA) RAN-translated proteins cause neurotoxicity by inducing ER stress. Acta Neuropathol. 2014, 128, 505–524. [Google Scholar] [CrossRef] [Green Version]
- May, S.; Hornburg, D.; Schludi, M.H.; Arzberger, T.; Rentzsch, K.; Schwenk, B.M.; Grässer, F.A.; Mori, K.; Kremmer, E.; Banzhaf-Strathmann, J.; et al. C9orf72 FTLD/ALS-associated Gly-Ala dipeptide repeat proteins cause neuronal toxicity and Unc119 sequestration. Acta Neuropathol. 2014, 128, 485–503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yamakawa, M.; Ito, D.; Honda, T.; Kubo, K.-I.; Noda, M.; Nakajima, K.; Suzuki, N. Characterization of the dipeptide repeat protein in the molecular pathogenesis of c9FTD/ALS. Hum. Mol. Genet. 2015, 24, 1630–1645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chang, Y.-J.; Jeng, U.-S.; Chiang, Y.-L.; Hwang, I.-S.; Chen, Y.-R. The Glycine-Alanine Dipeptide Repeat from C9orf72 Hexanucleotide Expansions Forms Toxic Amyloids Possessing Cell-to-Cell Transmission Properties. J. Biol. Chem. 2016, 291, 4903–4911. [Google Scholar] [CrossRef] [Green Version]
- Swaminathan, A.; Bouffard, M.; Liao, M.; Ryan, S.; Callister, J.B.; Pickering-Brown, S.M.; Armstrong, G.A.B.; Drapeau, P. Expression of C9orf72-related dipeptides impairs motor function in a vertebrate model. Hum. Mol. Genet. 2018, 27, 1754–1762. [Google Scholar] [CrossRef] [Green Version]
- Swinnen, B.; Bento-Abreu, A.; Gendron, T.F.; Boeynaems, S.; Bogaert, E.; Nuyts, R.; Timmers, M.; Scheveneels, W.; Hersmus, N.; Wang, J.; et al. A zebrafish model for C9orf72 ALS reveals RNA toxicity as a pathogenic mechanism. Acta Neuropathol. 2018, 135, 427–443. [Google Scholar] [CrossRef] [Green Version]
- Ohki, Y.; Wenninger-Weinzierl, A.; Hruscha, A.; Asakawa, K.; Kawakami, K.; Haass, C.; Edbauer, D.; Schmid, B. Glycine-alanine dipeptide repeat protein contributes to toxicity in a zebrafish model of C9orf72 associated neurodegeneration. Mol. Neurodegener. 2017, 12, 6. [Google Scholar] [CrossRef] [Green Version]
- Lee, K.-H.; Zhang, P.; Kim, H.J.; Mitrea, D.M.; Sarkar, M.; Freibaum, B.D.; Cika, J.; Coughlin, M.; Messing, J.; Molliex, A.; et al. C9orf72 Dipeptide Repeats Impair the Assembly, Dynamics, and Function of Membrane-Less Organelles. Cell 2016, 167, 774–788.e17. [Google Scholar] [CrossRef] [Green Version]
- Freibaum, B.D.; Lu, Y.; Lopez-Gonzalez, R.; Kim, N.C.; Almeida, S.; Lee, K.-H.; Badders, N.; Valentine, M.; Miller, B.L.; Wong, P.C.; et al. GGGGCC repeat expansion in C9orf72 compromises nucleocytoplasmic transport. Nature 2015, 525, 129–133. [Google Scholar] [CrossRef]
- Zhang, Y.-J.; Gendron, T.F.; Grima, J.C.; Sasaguri, H.; Jansen-West, K.; Xu, Y.-F.; Katzman, R.B.; Gass, J.; Murray, M.E.; Shinohara, M.; et al. C9ORF72 poly(GA) aggregates sequester and impair HR23 and nucleocytoplasmic transport proteins. Nat. Neurosci. 2016, 19, 668–677. [Google Scholar] [CrossRef]
- Schludi, M.H.; Becker, L.; Garrett, L.; Gendron, T.F.; Zhou, Q.; Schreiber, F.; Popper, B.; Dimou, L.; Strom, T.M.; Winkelmann, J.; et al. Spinal poly-GA inclusions in a C9orf72 mouse model trigger motor deficits and inflammation without neuron loss. Acta Neuropathol. 2017, 134, 241–254. [Google Scholar] [CrossRef]
- Yang, D.; Abdallah, A.; Li, Z.; Lu, Y.; Almeida, S.; Gao, F.-B. FTD/ALS-associated poly(GR) protein impairs the Notch pathway and is recruited by poly(GA) into cytoplasmic inclusions. Acta Neuropathol. 2015, 130, 525–535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mizielinska, S.; Grönke, S.; Niccoli, T.; Ridler, C.E.; Clayton, E.L.; Devoy, A.; Moens, T.; Norona, F.E.; Woollacott, I.O.C.; Pietrzyk, J.; et al. C9orf72 repeat expansions cause neurodegeneration in Drosophila through arginine-rich proteins. Science 2014, 345, 1192–1194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sakae, N.; Bieniek, K.F.; Zhang, Y.-J.; Ross, K.; Gendron, T.F.; Murray, M.E.; Rademakers, R.; Petrucelli, L.; Dickson, D. Poly-GR dipeptide repeat polymers correlate with neurodegeneration and Clinicopathological subtypes in C9ORF72-related brain disease. Acta Neuropathol. Commun. 2018, 6, 63. [Google Scholar] [CrossRef] [PubMed]
- Kwon, I.; Xiang, S.; Kato, M.; Wu, L.; Theodoropoulos, P.; Wang, T.; Kim, J.; Yun, J.; Xie, Y.; McKnight, S.L. Poly-dipeptides encoded by the C9orf72 repeats bind nucleoli, impede RNA biogenesis, and kill cells. Science 2014, 345, 1139–1145. [Google Scholar] [CrossRef] [Green Version]
- Flores, B.N.; Dulchavsky, M.; Krans, A.; Sawaya, M.R.; Paulson, H.L.; Todd, P.K.; Barmada, S.J.; Ivanova, M.I. Distinct C9orf72-Associated Dipeptide Repeat Structures Correlate with Neuronal Toxicity. PLoS ONE 2016, 11, e0165084. [Google Scholar] [CrossRef]
- Boeynaems, S.; Bogaert, E.; Kovacs, D.; Konijnenberg, A.; Timmerman, E.; Volkov, A.; Guharoy, M.; De Decker, M.; Jaspers, T.; Ryan, V.H.; et al. Phase Separation of C9orf72 Dipeptide Repeats Perturbs Stress Granule Dynamics. Mol. Cell 2017, 65, 1044–1055.e5. [Google Scholar] [CrossRef] [Green Version]
- Zhang, K.; Daigle, J.G.; Cunningham, K.M.; Coyne, A.N.; Ruan, K.; Grima, J.C.; Bowen, K.E.; Wadhwa, H.; Yang, P.; Rigo, F.; et al. Stress Granule Assembly Disrupts Nucleocytoplasmic Transport. Cell 2018, 173, 958–971.e17. [Google Scholar] [CrossRef] [Green Version]
- White, M.R.; Mitrea, D.M.; Zhang, P.; Stanley, C.B.; Cassidy, D.E.; Nourse, A.; Phillips, A.H.; Tolbert, M.; Taylor, J.P.; Kriwacki, R.W. C9orf72 Poly(PR) Dipeptide Repeats Disturb Biomolecular Phase Separation and Disrupt Nucleolar Function. Mol. Cell 2019, 74, 713–728.e6. [Google Scholar] [CrossRef]
- Shorter, J. Phase separation of RNA-binding proteins in physiology and disease: An introduction to the JBC Reviews thematic series. J. Biol. Chem. 2019, 294, 7113–7114. [Google Scholar] [CrossRef] [Green Version]
- Rossi, S.; Serrano, A.; Gerbino, V.; Giorgi, A.; Di Francesco, L.; Nencini, M.; Bozzo, F.; Schininà, M.E.; Bagni, C.; Cestra, G.; et al. Nuclear accumulation of mRNAs underlies G4C2-repeat-induced translational repression in a cellular model of C9orf72 ALS. J. Cell Sci. 2015, 128, 1787–1799. [Google Scholar] [CrossRef] [Green Version]
- Moens, T.G.; Niccoli, T.; Wilson, K.M.; Atilano, M.L.; Birsa, N.; Gittings, L.M.; Holbling, B.V.; Dyson, M.C.; Thoeng, A.; Neeves, J.; et al. C9orf72 arginine-rich dipeptide proteins interact with ribosomal proteins in vivo to induce a toxic translational arrest that is rescued by eIF1A. Acta Neuropathol. 2019, 137, 487–500. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cheng, W.; Wang, S.; Mestre, A.A.; Fu, C.; Makarem, A.; Xian, F.; Hayes, L.R.; Lopez-Gonzalez, R.; Drenner, K.; Jiang, J.; et al. C9ORF72 GGGGCC repeat-associated non-AUG translation is upregulated by stress through eIF2α phosphorylation. Nat. Commun. 2018, 9, 51. [Google Scholar] [CrossRef] [PubMed]
- Hartmann, H.; Hornburg, D.; Czuppa, M.; Bader, J.M.; Michaelsen, M.; Farny, D.; Arzberger, T.; Mann, M.; Meissner, F.; Edbauer, D. Proteomics and C9orf72 neuropathology identify ribosomes as poly-GR/PR interactors driving toxicity. Life Sci. Alliance 2018, 1, e201800070. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, Y.-J.; Gendron, T.F.; Ebbert, M.T.W.; O’Raw, A.D.; Yue, M.; Jansen-West, K.; Zhang, X.; Prudencio, M.; Chew, J.; Cook, C.N.; et al. Poly(GR) impairs protein translation and stress granule dynamics in C9orf72-associated frontotemporal dementia and amyotrophic lateral sclerosis. Nat. Med. 2018, 24, 1136–1142. [Google Scholar] [CrossRef] [PubMed]
- Lopez-Gonzalez, R.; Lu, Y.; Gendron, T.F.; Karydas, A.; Tran, H.; Yang, D.; Petrucelli, L.; Miller, B.L.; Almeida, S.; Gao, F.-B. Poly(GR) in C9ORF72-Related ALS/FTD Compromises Mitochondrial Function and Increases Oxidative Stress and DNA Damage in iPSC-Derived Motor Neurons. Neuron 2016, 92, 383–391. [Google Scholar] [CrossRef] [Green Version]
- Choi, S.Y.; Lopez-Gonzalez, R.; Krishnan, G.; Phillips, H.L.; Li, A.N.; Seeley, W.W.; Yao, W.-D.; Almeida, S.; Gao, F.-B. C9ORF72-ALS/FTD-associated poly(GR) binds Atp5a1 and compromises mitochondrial function in vivo. Nat. Neurosci. 2019, 22, 851–862. [Google Scholar] [CrossRef]
- Zhang, K.; Donnelly, C.J.; Haeusler, A.R.; Grima, J.C.; Machamer, J.B.; Steinwald, P.; Daley, E.L.; Miller, S.J.; Cunningham, K.M.; Vidensky, S.; et al. The C9orf72 repeat expansion disrupts nucleocytoplasmic transport. Nature 2015, 525, 56–61. [Google Scholar] [CrossRef] [Green Version]
- Boeynaems, S.; Bogaert, E.; Michiels, E.; Gijselinck, I.; Sieben, A.; Jovičić, A.; De Baets, G.; Scheveneels, W.; Steyaert, J.; Cuijt, I.; et al. Drosophila screen connects nuclear transport genes to DPR pathology in c9ALS/FTD. Sci. Rep. 2016, 6, 20877. [Google Scholar] [CrossRef]
- Khosravi, B.; Hartmann, H.; May, S.; Möhl, C.; Ederle, H.; Michaelsen, M.; Schludi, M.H.; Dormann, D.; Edbauer, D. Cytoplasmic poly-GA aggregates impair nuclear import of TDP-43 in C9orf72 ALS/FTLD. Hum. Mol. Genet. 2017, 26, 790–800. [Google Scholar] [CrossRef] [Green Version]
- Yang, Q.; Zhou, J.-W. Neuroinflammation in the central nervous system: Symphony of glial cells. Glia 2019, 67, 1017–1035. [Google Scholar] [CrossRef]
- Ghasemi, M.; Fatemi, A. Pathologic role of glial nitric oxide in adult and pediatric neuroinflammatory diseases. Neurosci. Biobehav. Rev. 2014, 45, 168–182. [Google Scholar] [CrossRef] [PubMed]
- Block, M.L.; Zecca, L.; Hong, J.-S. Microglia-mediated neurotoxicity: Uncovering the molecular mechanisms. Nat. Rev. Neurosci. 2007, 8, 57–69. [Google Scholar] [CrossRef] [PubMed]
- Ginhoux, F.; Greter, M.; Leboeuf, M.; Nandi, S.; See, P.; Gokhan, S.; Mehler, M.F.; Conway, S.J.; Ng, L.G.; Stanley, E.R.; et al. Fate Mapping Analysis Reveals That Adult Microglia Derive from Primitive Macrophages. Science 2010, 330, 841–845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aarum, J.; Sandberg, K.; Haeberlein, S.L.B.; Persson, M.A. Migration and differentiation of neural precursor cells can be directed by microglia. Proc. Natl. Acad. Sci. USA 2003, 100, 15983–15988. [Google Scholar] [CrossRef] [Green Version]
- Kettenmann, H.; Hanisch, U.-K.; Noda, M.; Verkhratsky, A. Physiology of Microglia. Physiol. Rev. 2011, 91, 461–553. [Google Scholar] [CrossRef]
- Morgan, S.C.; Taylor, D.L.; Pocock, J.M. Microglia release activators of neuronal proliferation mediated by activation of mitogen-activated protein kinase, phosphatidylinositol-3-kinase/Akt and delta-Notch signalling cascades. J. Neurochem. 2004, 90, 89–101. [Google Scholar] [CrossRef]
- Simard, M.; Nedergaard, M. The neurobiology of glia in the context of water and ion homeostasis. Neuroscience 2004, 129, 877–896. [Google Scholar] [CrossRef]
- Li, K.; Li, J.; Zheng, J.; Qin, S. Reactive Astrocytes in Neurodegenerative Diseases. Aging Dis. 2019, 10, 664–675. [Google Scholar] [CrossRef] [Green Version]
- Kacerovsky, J.B.; Murai, K.K. Stargazing: Monitoring subcellular dynamics of brain astrocytes. Neuroscience 2016, 323, 84–95. [Google Scholar] [CrossRef]
- Volterra, A.; Meldolesi, J. Astrocytes, from brain glue to communication elements: The revolution continues. Nat. Rev. Neurosci. 2005, 6, 626–640. [Google Scholar] [CrossRef]
- Liddelow, S.A.; Barres, B.A. Reactive Astrocytes: Production, Function, and Therapeutic Potential. Immunity 2017, 46, 957–967. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zamanian, J.L.; Xu, L.; Foo, L.C.; Nouri, N.; Zhou, L.; Giffard, R.G.; Barres, B.A. Genomic Analysis of Reactive Astrogliosis. J. Neurosci. 2012, 32, 6391–6410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lall, D.; Baloh, R.H. Microglia and C9orf72 in neuroinflammation and ALS and frontotemporal dementia. J. Clin. Investig. 2017, 127, 3250–3258. [Google Scholar] [CrossRef] [PubMed]
- Shao, Q.; Yang, M.; Liang, C.; Ma, L.; Zhang, W.; Jiang, Z.; Luo, J.; Lee, J.-K.; Liang, C.; Chen, J.-F. C9orf72 and smcr8 mutant mice reveal MTORC1 activation due to impaired lysosomal degradation and exocytosis. Autophagy 2020, 16, 1635–1650. [Google Scholar] [CrossRef] [PubMed]
- Ho, W.Y.; Navakkode, S.; Liu, F.; Soong, T.W.; Ling, S.-C. Deregulated expression of a longevity gene, Klotho, in the C9orf72 deletion mice with impaired synaptic plasticity and adult hippocampal neurogenesis. Acta Neuropathol. Commun. 2020, 8, 155. [Google Scholar] [CrossRef] [PubMed]
- Prudencio, M.; Belzil, V.V.; Batra, R.; Ross, C.A.; Gendron, T.F.; Pregent, L.J.; Murray, M.E.; Overstreet, K.K.; Piazza-Johnston, A.E.; Desaro, P.; et al. Distinct brain transcriptome profiles in C9orf72-associated and sporadic ALS. Nat. Neurosci. 2015, 18, 1175–1182. [Google Scholar] [CrossRef]
- Umoh, M.E.; Dammer, E.B.; Dai, J.; Duong, D.M.; Lah, J.J.; Levey, A.I.; Gearing, M.; Glass, J.D.; Seyfried, N.T. A proteomic network approach across the ALS-FTD disease spectrum resolves clinical phenotypes and genetic vulnerability in human brain. EMBO Mol. Med. 2018, 10, 48–62. [Google Scholar] [CrossRef]
- Oeckl, P.; Weydt, P.; Steinacker, P.; Anderl-Straub, S.; Nordin, F.; Volk, A.E.; Diehl-Schmid, J.; Andersen, P.M.; Kornhuber, J.; Danek, A.; et al. Different neuroinflammatory profile in amyotrophic lateral sclerosis and frontotemporal dementia is linked to the clinical phase. J. Neurol. Neurosurg. Psychiatry 2019, 90, 4–10. [Google Scholar] [CrossRef]
- Liu, Y.; Pattamatta, A.; Zu, T.; Reid, T.; Bardhi, O.; Borchelt, D.R.; Yachnis, A.T.; Ranum, L.P.W. C9orf72 BAC Mouse Model with Motor Deficits and Neurodegenerative Features of ALS/FTD. Neuron 2016, 90, 521–534. [Google Scholar] [CrossRef] [Green Version]
- Cistaro, A.; Pagani, M.; Montuschi, A.; Calvo, A.; Moglia, C.; Canosa, A.; Restagno, G.; Brunetti, M.; Traynor, B.J.; Nobili, F.; et al. The metabolic signature of C9ORF72-related ALS: FDG PET comparison with nonmutated patients. Eur. J. Nucl. Med. Mol. Imaging 2014, 41, 844–852. [Google Scholar] [CrossRef] [Green Version]
- Brettschneider, J.; Toledo, J.B.; Van Deerlin, V.M.; Elman, L.; McCluskey, L.; Lee, V.M.-Y.; Trojanowski, J.Q. Microglial Activation Correlates with Disease Progression and Upper Motor Neuron Clinical Symptoms in Amyotrophic Lateral Sclerosis. PLoS ONE 2012, 7, e39216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cardenas, A.M.; Sarlls, J.E.; Kwan, J.Y.; Bageac, D.; Gala, Z.S.; Danielian, L.E.; Ray-Chaudhury, A.; Wang, H.-W.; Miller, K.L.; Foxley, S.; et al. Pathology of callosal damage in ALS: An ex-vivo, 7 T diffusion tensor MRI study. NeuroImage Clin. 2017, 15, 200–208. [Google Scholar] [CrossRef] [PubMed]
- Heller, C.; Foiani, M.S.; Moore, K.M.; Convery, R.S.; Bocchetta, M.; Neason, M.; Cash, D.M.; Thomas, D.L.; Greaves, C.V.; Woollacott, I.O.C.; et al. Plasma glial fibrillary acidic protein is raised in progranulin-associated frontotemporal dementia. J. Neurol. Neurosurg. Psychiatry 2020, 91, 263–270. [Google Scholar] [CrossRef] [Green Version]
- Chistiakov, D.A.; Killingsworth, M.C.; Myasoedova, V.A.; Orekhov, A.N.; Bobryshev, Y.V. CD68/macrosialin: Not just a histochemical marker. Lab. Investig. 2017, 97, 4–13. [Google Scholar] [CrossRef] [Green Version]
- Zotova, E.; Bharambe, V.; Cheaveau, M.; Morgan, W.; Holmes, C.; Harris, S.; Neal, J.W.; Love, S.; Nicoll, J.A.R.; Boche, D. Inflammatory components in human Alzheimer’s disease and after active amyloid-β42 immunization. Brain 2013, 136, 2677–2696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Walker, D.G.; Lue, L.-F. Immune phenotypes of microglia in human neurodegenerative disease: Challenges to detecting microglial polarization in human brains. Alzheimer’s Res. Ther. 2015, 7, 56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Martinez, F.O.; Helming, L.; Milde, R.; Varin, A.; Melgert, B.N.; Draijer, C.; Thomas, B.; Fabbri, M.; Crawshaw, A.; Ho, L.P.; et al. Genetic programs expressed in resting and IL-4 alternatively activated mouse and human macrophages: Similarities and differences. Blood 2013, 121, e57–e69. [Google Scholar] [CrossRef] [PubMed]
- Ito, D.; Imai, Y.; Ohsawa, K.; Nakajima, K.; Fukuuchi, Y.; Kohsaka, S. Microglia-specific localisation of a novel calcium binding protein, Iba1. Mol. Brain Res. 1998, 57, 1–9. [Google Scholar] [CrossRef]
- Ising, C.; Venegas, C.; Zhang, S.; Scheiblich, H.; Schmidt, S.V.; Vieira-Saecker, A.; Schwartz, S.; Albasset, S.; McManus, R.M.; Tejera, D.; et al. NLRP3 inflammasome activation drives tau pathology. Nature 2019, 575, 669–673. [Google Scholar] [CrossRef]
- Dominy, S.S.; Lynch, C.; Ermini, F.; Benedyk, M.; Marczyk, A.; Konradi, A.; Nguyen, M.; Haditsch, U.; Raha, D.; Griffin, C.; et al. Porphyromonas gingivalis in Alzheimer’s disease brains: Evidence for disease causation and treatment with small-molecule inhibitors. Sci. Adv. 2019, 5, eaau3333. [Google Scholar] [CrossRef] [Green Version]
- Ohsawa, K.; Imai, Y.; Sasaki, Y.; Kohsaka, S. Microglia/macrophage-specific protein Iba1 binds to fimbrin and enhances its actin-bundling activity. J. Neurochem. 2004, 88, 844–856. [Google Scholar] [CrossRef] [PubMed]
- Mizielinska, S.; Lashley, T.; Norona, F.E.; Clayton, E.L.; Ridler, C.E.; Fratta, P.; Isaacs, A.M. C9orf72 frontotemporal lobar degeneration is characterised by frequent neuronal sense and antisense RNA foci. Acta Neuropathol. 2013, 126, 845–857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- DeJesus-Hernandez, M.; Finch, N.A.; Wang, X.; Gendron, T.F.; Bieniek, K.F.; Heckman, M.G.; Vasilevich, A.; Murray, M.E.; Rousseau, L.; Weesner, R.; et al. In-depth clinico-pathological examination of RNA foci in a large cohort of C9ORF72 expansion carriers. Acta Neuropathol. 2017, 134, 255–269. [Google Scholar] [CrossRef] [PubMed]
- MacKenzie, I.R.; Arzberger, T.; Kremmer, E.; Troost, D.; Lorenzl, S.; Mori, K.; Weng, S.-M.; Haass, C.; Kretzschmar, H.A.; Edbauer, D.; et al. Dipeptide repeat protein pathology in C9ORF72 mutation cases: Clinico-pathological correlations. Acta Neuropathol. 2013, 126, 859–879. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Saberi, S.; Stauffer, J.E.; Jiang, J.; Garcia, S.D.; Taylor, A.E.; Schulte, D.; Ohkubo, T.; Schloffman, C.L.; Maldonado, M.; Baughn, M.; et al. Sense-encoded poly-GR dipeptide repeat proteins correlate to neurodegeneration and uniquely co-localize with TDP-43 in dendrites of repeat-expanded C9orf72 amyotrophic lateral sclerosis. Acta Neuropathol. 2018, 135, 459–474. [Google Scholar] [CrossRef]
- Peters, O.M.; Cabrera, G.T.; Tran, H.; Gendron, T.F.; McKeon, J.E.; Metterville, J.; Weiss, A.; Wightman, N.; Salameh, J.; Kim, J.; et al. Human C9ORF72 Hexanucleotide Expansion Reproduces RNA Foci and Dipeptide Repeat Proteins but Not Neurodegeneration in BAC Transgenic Mice. Neuron 2015, 88, 902–909. [Google Scholar] [CrossRef] [Green Version]
- Westergard, T.; Jensen, B.K.; Wen, X.; Cai, J.; Kropf, E.; Iacovitti, L.; Pasinelli, P.; Trotti, D. Cell-to-Cell Transmission of Dipeptide Repeat Proteins Linked to C9orf72-ALS/FTD. Cell Rep. 2016, 17, 645–652. [Google Scholar] [CrossRef] [Green Version]
- Michell-Robinson, M.A.; Touil, H.; Healy, L.M.; Owen, D.R.; Durafourt, B.A.; Bar-Or, A.; Antel, J.P.; Moore, C.S. Roles of microglia in brain development, tissue maintenance and repair. Brain 2015, 138, 1138–1159. [Google Scholar] [CrossRef] [Green Version]
- Verkhratsky, A.; Nedergaard, M. Physiology of Astroglia. Physiol. Rev. 2018, 98, 239–389. [Google Scholar] [CrossRef]
- Murray, M.E.; DeJesus-Hernandez, M.; Rutherford, N.J.; Baker, M.; Duara, R.; Graff-Radford, N.R.; Wszolek, Z.K.; Ferman, T.J.; Josephs, K.A.; Boylan, K.B.; et al. Clinical and neuropathologic heterogeneity of c9FTD/ALS associated with hexanucleotide repeat expansion in C9ORF72. Acta Neuropathol. 2011, 122, 673–690. [Google Scholar] [CrossRef] [Green Version]
- Brettschneider, J.; Arai, K.; Del Tredici, K.; Toledo, J.B.; Robinson, J.L.; Lee, E.B.; Kuwabara, S.; Shibuya, K.; Irwin, D.J.; Fang, L.; et al. TDP-43 pathology and neuronal loss in amyotrophic lateral sclerosis spinal cord. Acta Neuropathol. 2014, 128, 423–437. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fatima, M.; Tan, R.; Halliday, G.M.; Kril, J.J. Spread of pathology in amyotrophic lateral sclerosis: Assessment of phosphorylated TDP-43 along axonal pathways. Acta Neuropathol. Commun. 2015, 3, 47. [Google Scholar] [CrossRef] [PubMed]
- Meyer, K.; Ferraiuolo, L.; Miranda, C.J.; Likhite, S.; McElroy, S.; Renusch, S.; Ditsworth, D.; Lagier-Tourenne, C.; Smith, R.A.; Ravits, J.; et al. Direct conversion of patient fibroblasts demonstrates non-cell autonomous toxicity of astrocytes to motor neurons in familial and sporadic ALS. Proc. Natl. Acad. Sci. USA 2014, 111, 829–832. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Madill, M.; McDonagh, K.; Ma, J.; Vajda, A.; McLoughlin, P.; O’Brien, T.; Hardiman, O.; Shen, S. Amyotrophic lateral sclerosis patient iPSC-derived astrocytes impair autophagy via non-cell autonomous mechanisms. Mol. Brain 2017, 10, 22. [Google Scholar] [CrossRef] [PubMed]
- Varcianna, A.; Myszczynska, M.A.; Castelli, L.M.; O’Neill, B.; Kim, Y.; Talbot, J.; Nyberg, S.; Nyamali, I.; Heath, P.R.; Stopford, M.J.; et al. Micro-RNAs secreted through astrocyte-derived extracellular vesicles cause neuronal network degeneration in C9orf72 ALS. EBioMedicine 2019, 40, 626–635. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Allen, S.P.; Hall, B.; Castelli, L.M.; Francis, L.; Woof, R.; Siskos, A.P.; Kouloura, E.; Gray, E.; Thompson, A.G.; Talbot, K.; et al. Astrocyte adenosine deaminase loss increases motor neuron toxicity in amyotrophic lateral sclerosis. Brain 2019, 142, 586–605. [Google Scholar] [CrossRef] [Green Version]
- Allen, S.P.; Hall, B.; Woof, R.; Francis, L.; Gatto, N.; Shaw, A.C.; Myszczynska, M.; Hemingway, J.; Coldicott, I.; Willcock, A.; et al. C9orf72 expansion within astrocytes reduces metabolic flexibility in amyotrophic lateral sclerosis. Brain 2019, 142, 3771–3790. [Google Scholar] [CrossRef]
- Onesto, E.; Colombrita, C.; Gumina, V.; Borghi, M.O.; Dusi, S.; Doretti, A.; Fagiolari, G.; Invernizzi, F.; Moggio, M.; Tiranti, V.; et al. Gene-specific mitochondria dysfunctions in human TARDBP and C9ORF72 fibroblasts. Acta Neuropathol. Commun. 2016, 4, 47. [Google Scholar] [CrossRef] [Green Version]
- Fomin, V.; Richard, P.; Hoque, M.; Li, C.; Gu, Z.; Fissore-O’Leary, M.; Tian, B.; Prives, C.; Manley, J.L. The C9ORF72 Gene, Implicated in Amyotrophic Lateral Sclerosis and Frontotemporal Dementia, Encodes a Protein That Functions in Control of Endothelin and Glutamate Signaling. Mol. Cell. Biol. 2018, 38, e00155-18. [Google Scholar] [CrossRef] [Green Version]
- D’Antoni, S.; Ranno, E.; Spatuzza, M.; Cavallaro, S.; Catania, M.V. Endothelin-1 Induces Degeneration of Cultured Motor Neurons through a Mechanism Mediated by Nitric Oxide and PI3K/Akt Pathway. Neurotox. Res. 2017, 32, 58–70. [Google Scholar] [CrossRef]
- Rostalski, H.; Leskelä, S.; Huber, N.; Katisko, K.; Cajanus, A.; Solje, E.; Marttinen, M.; Natunen, T.; Remes, A.M.; Hiltunen, M.; et al. Astrocytes and Microglia as Potential Contributors to the Pathogenesis of C9orf72 Repeat Expansion-Associated FTLD and ALS. Front. Neurosci. 2019, 13, 486. [Google Scholar] [CrossRef] [PubMed]
- Siddique, N.; Siddique, T. Amyotrophic Lateral Sclerosis Overview. In GeneReviews®; Adam, M.P., Ardinger, H.H., Pagon, R.A., Wallace, S.E., Bean, L.J.H., Stephens, K., Amemiya, A., Eds.; University of Washington: Seattle, WA, USA, 1993. [Google Scholar]
- Goutman, S.A.; Chen, K.S.; Paez-Colasante, X.; Feldman, E.L. Emerging understanding of the genotype–phenotype relationship in amyotrophic lateral sclerosis. Handb. Clin.Neurol. 2018, 148, 603–623. [Google Scholar] [CrossRef] [PubMed]
- Gendron, T.F.; Van Blitterswijk, M.; Bieniek, K.F.; Daughrity, L.M.; Jiang, J.; Rush, B.K.; Pedraza, O.; Lucas, J.A.; Murray, M.E.; Desaro, P.; et al. Cerebellar c9RAN proteins associate with clinical and neuropathological characteristics of C9ORF72 repeat expansion carriers. Acta Neuropathol. 2015, 130, 559–573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schludi, M.H.; May, S.; Grässer, F.A.; Rentzsch, K.; Kremmer, E.; Küpper, C.; Klopstock, T.; German Consortium for Frontotemporal Lobar Degeneration; Bavarian Brain Banking Alliance; Arzberger, T.; et al. Distribution of dipeptide repeat proteins in cellular models and C9orf72 mutation cases suggests link to transcriptional silencing. Acta Neuropathol. 2015, 130, 537–555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gendron, T.F.; Chew, J.; Stankowski, J.N.; Hayes, L.R.; Zhang, Y.-J.; Prudencio, M.; Carlomagno, Y.; Daughrity, L.M.; Jansen-West, K.; Perkerson, E.A.; et al. Poly(GP) proteins are a useful pharmacodynamic marker forC9ORF72-associated amyotrophic lateral sclerosis. Sci. Transl. Med. 2017, 9, eaai7866. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cardani, R.; Mancinelli, E.; Giagnacovo, M.; Sansone, V.; Meola, G. Ribonuclear inclusions as biomarker of myotonic dystrophy type 2, even in improperly frozen or defrozen skeletal muscle biopsies. Eur. J. Histochem. 2009, 53, 107–111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ransohoff, R.M. How neuroinflammation contributes to neurodegeneration. Science 2016, 353, 777–783. [Google Scholar] [CrossRef] [PubMed]
- Collins, M.A.; An, J.; Hood, B.L.; Conrads, T.P.; Bowser, R.P. Label-Free LC–MS/MS Proteomic Analysis of Cerebrospinal Fluid Identifies Protein/Pathway Alterations and Candidate Biomarkers for Amyotrophic Lateral Sclerosis. J. Proteome Res. 2015, 14, 4486–4501. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.; Liu, X.-H.; Wu, J.-J.; Ren, H.-M.; Wang, J.; Ding, Z.-T.; Jiang, Y.-P. Proteomic analysis of cerebrospinal fluid in amyotrophic lateral sclerosis. Exp. Ther. Med. 2016, 11, 2095–2106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, X.; Chen, Y.; Wei, Q.-Q.; Ou, R.; Cao, B.; Zhao, B.; Shang, H. Assessment of a multiple biomarker panel for diagnosis of amyotrophic lateral sclerosis. BMC Neurol. 2016, 16, 173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Moreau, C.; Gosset, P.; Brunaud-Danel, V.; Lassalle, P.; Degonne, B.; Destée, A.; Defebvre, L.; Devos, D. CSF profiles of angiogenic and inflammatory factors depend on the respiratory status of ALS patients. Amyotroph. Lateral Scler. 2009, 10, 175–181. [Google Scholar] [CrossRef] [PubMed]
- Vu, L.T.; Bowser, R.P. Fluid-Based Biomarkers for Amyotrophic Lateral Sclerosis. Neurotherapeutics 2017, 14, 119–134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ismail, A.; Cooper-Knock, J.; Highley, J.R.; Milano, A.; Kirby, J.; Goodall, E.; Lowe, J.; Scott, I.; Constantinescu, C.S.; Walters, S.J.; et al. Concurrence of multiple sclerosis and amyotrophic lateral sclerosis in patients with hexanucleotide repeat expansions of C9ORF72. J. Neurol. Neurosurg. Psychiatry 2013, 84, 79–87. [Google Scholar] [CrossRef] [PubMed]
- Katisko, K.; Solje, E.; Korhonen, P.; Jääskeläinen, O.; Loppi, S.; Hartikainen, P.; Koivisto, A.M.; Kontkanen, A.; Korhonen, V.E.; Helisalmi, S.; et al. Peripheral inflammatory markers and clinical correlations in patients with frontotemporal lobar degeneration with and without the C9orf72 repeat expansion. J. Neurol. 2020, 267, 76–86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bennett, S.A.; Tanaz, R.; Cobos, S.N.; Torrente, M.P. Epigenetics in amyotrophic lateral sclerosis: A role for histone post-translational modifications in neurodegenerative disease. Transl. Res. 2019, 204, 19–30. [Google Scholar] [CrossRef] [PubMed]
- Benigni, M.; Ricci, C.; Jones, A.R.; Giannini, F.; Al-Chalabi, A.; Battistini, S. Identification of miRNAs as Potential Biomarkers in Cerebrospinal Fluid from Amyotrophic Lateral Sclerosis Patients. NeuroMolecular Med. 2016, 18, 551–560. [Google Scholar] [CrossRef]
- De Felice, B.; Annunziata, A.; Fiorentino, G.; Borra, M.; Biffali, E.; Coppola, C.; Cotrufo, R.; Brettschneider, J.; Giordana, M.L.; Dalmay, T.; et al. miR-338-3p is over-expressed in blood, CFS, serum and spinal cord from sporadic amyotrophic lateral sclerosis patients. Neurogenetics 2014, 15, 243–253. [Google Scholar] [CrossRef]
- Takahashi, I.; Hama, Y.; Matsushima, M.; Hirotani, M.; Kano, T.; Hohzen, H.; Yabe, I.; Utsumi, J.; Sasaki, H. Identification of plasma microRNAs as a biomarker of sporadic Amyotrophic Lateral Sclerosis. Mol. Brain 2015, 8, 67. [Google Scholar] [CrossRef]
- Freischmidt, A.; Müller, K.; Ludolph, A.; Weishaupt, J. Systemic dysregulation of TDP-43 binding microRNAs in amyotrophic lateral sclerosis. Acta Neuropathol. Commun. 2013, 1, 42. [Google Scholar] [CrossRef] [Green Version]
- Williams, A.H.; Valdez, G.; Moresi, V.; Qi, X.; McAnally, J.; Elliott, J.L.; Bassel-Duby, R.; Sanes, J.R.; Olson, E.N. MicroRNA-206 Delays ALS Progression and Promotes Regeneration of Neuromuscular Synapses in Mice. Science 2009, 326, 1549–1554. [Google Scholar] [CrossRef] [Green Version]
- Bowser, R.; Turner, M.R.; Shefner, J.M. Biomarkers in amyotrophic lateral sclerosis: Opportunities and limitations. Nat. Rev. Neurol. 2011, 7, 631–638. [Google Scholar] [CrossRef]
- Mendez, E.F.; Sattler, R. Biomarker development for C9orf72 repeat expansion in ALS. Brain Res. 2015, 1607, 26–35. [Google Scholar] [CrossRef] [PubMed]
- De Godoy Rousseff Prado, L.; Bicalho, I.C.S.; Magalhães, D.; Caramelli, P.; Teixeira, A.L.; De Souza, L.C. C9ORF72 and the FTD-ALS spectrum: A systematic review of neuroimaging studies. Dement. Neuropsychol. 2015, 9, 413–421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Boeve, B.F.; Boylan, K.B.; Graff-Radford, N.R.; DeJesus-Hernandez, M.; Knopman, D.S.; Pedraza, O.; Vemuri, P.; Jones, D.; Lowe, V.; Murray, M.E.; et al. Characterization of frontotemporal dementia and/or amyotrophic lateral sclerosis associated with the GGGGCC repeat expansion in C9ORF72. Brain 2012, 135, 765–783. [Google Scholar] [CrossRef] [PubMed]
- Mahoney, C.J.; Beck, J.; Rohrer, J.D.; Lashley, T.; Mok, K.; Shakespeare, T.; Yeatman, T.; Warrington, E.K.; Schott, J.M.; Fox, N.C.; et al. Frontotemporal dementia with the C9ORF72 hexanucleotide repeat expansion: Clinical, neuroanatomical and neuropathological features. Brain 2012, 135, 736–750. [Google Scholar] [CrossRef] [PubMed]
- Bede, P.; Bokde, A.L.; Byrne, S.; Elamin, M.; McLaughlin, R.L.; Kenna, K.; Fagan, A.J.; Pender, N.; Bradley, D.G.; Hardiman, O. Multiparametric MRI study of ALS stratified for the C9orf72 genotype. Neurology 2013, 81, 361–369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chew, S.; Atassi, N. Positron Emission Tomography Molecular Imaging Biomarkers for Amyotrophic Lateral Sclerosis. Front. Neurol. 2019, 10, 135. [Google Scholar] [CrossRef] [Green Version]
- Zürcher, N.R.; Loggia, M.L.; Lawson, R.; Chonde, D.B.; Izquierdo-Garcia, D.; Yasek, J.E.; Akeju, O.; Catana, C.; Rosen, B.R.; Cudkowicz, M.E.; et al. Increased in vivo glial activation in patients with amyotrophic lateral sclerosis: Assessed with [11C]-PBR28. NeuroImage Clin. 2015, 7, 409–414. [Google Scholar] [CrossRef] [Green Version]
- Alshikho, M.J.; Zürcher, N.R.; Loggia, M.; Cernasov, P.; Reynolds, B.; Pijanowski, O.; Chonde, D.B.; Izquierdo-Garcia, D.; Mainero, C.; Catana, C.; et al. Integrated magnetic resonance imaging and [11C]-PBR28 positron emission tomographic imaging in amyotrophic lateral sclerosis. Ann. Neurol. 2018, 83, 1186–1197. [Google Scholar] [CrossRef]
- Alshikho, M.J.; Zürcher, N.R.; Loggia, M.L.; Cernasov, P.; Chonde, D.B.; Izquierdo-Garcia, D.; Yasek, J.E.; Akeju, O.; Catana, C.; Rosen, B.R.; et al. Glial activation colocalizes with structural abnormalities in amyotrophic lateral sclerosis. Neurology 2016, 87, 2554–2561. [Google Scholar] [CrossRef] [Green Version]
- Dolgin, E. The brain’s traffic problems. Science 2019, 363, 221–223. [Google Scholar] [CrossRef] [PubMed]
C9orf72 Knockout Method | Immune System | Motor | Cognition/Behavior | Ref |
---|---|---|---|---|
Non-conditional exons 2–6 | Splenomegaly and cervical lymphadenopathy | Mild motor deficits only on a rotarod assay at 12 months | Mild social interaction and social recognition deficits | [58] |
Non-conditional full gene | Splenomegaly, systemic lymphadenopathy, glumerulonephropathy, and ↑ serum IL-12, IL-17a, IL-10, TNF-α, plasma cells, and activated T cells. | Mild motor deficits, tremor, and rigidity at 40 weeks | NR | [59] |
Non-conditional exons 2–6 or zinc finger deletion | Splenomegaly and cervical lymphadenopathy | Normal function | NR | [60] |
NR, all tissues full knockout | Splenomegaly, cervical lymphadenopathy, and B-cell lymphomas | Normal function | NR | [46] |
Non-conditional exons 2–6 knockouts in a C57BL/6 background or CRISPR/Cas9 | Splenomegaly, cervical lymphadenopathy, hepatomegaly, and ↑ serum IL-22, IL-28, IL-23, IL-6, MCP-1, IL-31, IL-5, IL-10, IL-1β, IL-15/IL-15R, IFNγ, IL-3, GM-CSF, IL-17A, IFNα, MIP-1B, LIF, GROα | NR | NR | [61] |
Non-conditional CRISPR/Cas9 | Splenomegaly and systemic lymphadenopathy | NR | NR | [63] |
Non-conditional exons 2–6 | Splenomegaly | NR | Lethargy | [62] |
Non-conditional exons 2–6 | Splenomegaly, systemic lymphadenopathy, and ↑ serum IL-6 | NR | NR | [134] |
Non-conditional exons 2–6 | Splenomegaly | Normal function at 3 months | NR | [135] |
Species | Region | Results | Ref |
---|---|---|---|
C9orf72−/− mice | Brain | ↑ LysoTracker- and Lamp1-positive structures in microglia | [60] |
Isolated spinal cord microglia | ↑ IL-6 and IL-1b levels | ||
C9orf72−/− mice | Brain and spinal cord | No change in GFAP and Iba1 staining at 18 months | [57] |
(G4C2)500 BAC transgenic mice | Hippocampus | ↑ Iba1 staining in acute end-stage (20–40 weeks) mice | [139] |
Motor cortex (layers I–III & layer V) and hippocampus | ↑ GFAP staining in acute end-stage (20–40 weeks) mice | ||
Transgenic mice expressing poly-(GA)149 | Spinal cord | ↑ CD68 and Iba1 immunostaining and mRNA expression at six months, but little at one month; No change in GFAP immunostaining and mRNA expression | [100] |
Transgenic mice expressing poly-(GA)50 | Brain | ↑ GFAP mRNA expression, but No change in Iba1 mRNA expression at six months | [99] |
Cortex, motor cortex, and hippocampus | ↑ GFAP immunostaining and immunohistochemistry | ||
Transgenic mice expressing poly-(GR)100 | Brain | ↑ GFAP and Iba1 mRNA expression and immunostaining at 1.5 > 3 > 6 months | [114] |
C9orf72 ALS patients | Postmortem motor cortex and spinal cord | ↑ Iba1 and Lamp1 immunostaining | [60] |
C9orf72 ALS patients | Postmortem pyramidal tract at all levels (white matter underlying motor cortex, mid-crus cerebri, medullary pyramids, and lateral and anterior corticospinal tracts) | ↑ CD68 immunohistochemistry | [4] |
C9orf72 ALS patients | Postmortem white matter of the medulla and the motor cortex | ↑ CD68 and Iba1 immunostaining | [141] |
C9orf72 ALS patients | Postmortem corpus callosum | ↑ CD68 immunohistochemistry in the body more than genu or splenium of the callosum | [142] |
C9orf72 FTD patients | Plasma | No change in GFAP concentration between pre- and symptomatic cases and non-carriers | [143] |
C9orf72 ALS patients | [18F]FDG PET in C9orf72 ALS vs. sporadic ALS | ↓ Metabolism in the anterior and posterior cingulate cortex, insula, caudate and thalamus, the left frontal and superior temporal cortex, ↑ Metabolism in the midbrain, bilateral occipital cortex, globus pallidus, and left inferior temporal cortex | [140] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Ghasemi, M.; Keyhanian, K.; Douthwright, C. Glial Cell Dysfunction in C9orf72-Related Amyotrophic Lateral Sclerosis and Frontotemporal Dementia. Cells 2021, 10, 249. https://doi.org/10.3390/cells10020249
Ghasemi M, Keyhanian K, Douthwright C. Glial Cell Dysfunction in C9orf72-Related Amyotrophic Lateral Sclerosis and Frontotemporal Dementia. Cells. 2021; 10(2):249. https://doi.org/10.3390/cells10020249
Chicago/Turabian StyleGhasemi, Mehdi, Kiandokht Keyhanian, and Catherine Douthwright. 2021. "Glial Cell Dysfunction in C9orf72-Related Amyotrophic Lateral Sclerosis and Frontotemporal Dementia" Cells 10, no. 2: 249. https://doi.org/10.3390/cells10020249
APA StyleGhasemi, M., Keyhanian, K., & Douthwright, C. (2021). Glial Cell Dysfunction in C9orf72-Related Amyotrophic Lateral Sclerosis and Frontotemporal Dementia. Cells, 10(2), 249. https://doi.org/10.3390/cells10020249