The stress granule protein G3BP1 alleviates spinocerebellar ataxia-associated deficits

Abstract Polyglutamine diseases are a group of neurodegenerative disorders caused by an abnormal expansion of CAG repeat tracts in the codifying regions of nine, otherwise unrelated, genes. While the protein products of these genes are suggested to play diverse cellular roles, the pathogenic mutant proteins bearing an expanded polyglutamine sequence share a tendency to self-assemble, aggregate and engage in abnormal molecular interactions. Understanding the shared paths that link polyglutamine protein expansion to the nervous system dysfunction and the degeneration that takes place in these disorders is instrumental to the identification of targets for therapeutic intervention. Among polyglutamine diseases, spinocerebellar ataxias (SCAs) share many common aspects, including the fact that they involve dysfunction of the cerebellum, resulting in ataxia. Our work aimed at exploring a putative new therapeutic target for the two forms of SCA with higher worldwide prevalence, SCA type 2 (SCA2) and type 3 (SCA3), which are caused by expanded forms of ataxin-2 (ATXN2) and ataxin-3 (ATXN3), respectively. The pathophysiology of polyglutamine diseases has been described to involve an inability to properly respond to cell stress. We evaluated the ability of GTPase-activating protein-binding protein 1 (G3BP1), an RNA-binding protein involved in RNA metabolism regulation and stress responses, to counteract SCA2 and SCA3 pathology, using both in vitro and in vivo disease models. Our results indicate that G3BP1 overexpression in cell models leads to a reduction of ATXN2 and ATXN3 aggregation, associated with a decrease in protein expression. This protective effect of G3BP1 against polyglutamine protein aggregation was reinforced by the fact that silencing G3bp1 in the mouse brain increases human expanded ATXN2 and ATXN3 aggregation. Moreover, a decrease of G3BP1 levels was detected in cells derived from patients with SCA2 and SCA3, suggesting that G3BP1 function is compromised in the context of these diseases. In lentiviral mouse models of SCA2 and SCA3, G3BP1 overexpression not only decreased protein aggregation but also contributed to the preservation of neuronal cells. Finally, in an SCA3 transgenic mouse model with a severe ataxic phenotype, G3BP1 lentiviral delivery to the cerebellum led to amelioration of several motor behavioural deficits. Overall, our results indicate that a decrease in G3BP1 levels may be a contributing factor to SCA2 and SCA3 pathophysiology, and that administration of this protein through viral vector-mediated delivery may constitute a putative approach to therapy for these diseases, and possibly other polyglutamine disorders.

[1]  Carlos A. Matos,et al.  Autophagy in Spinocerebellar ataxia type 2, a dysregulated pathway, and a target for therapy , 2021, Cell Death & Disease.

[2]  Pablo Mier,et al.  Between Interactions and Aggregates: The PolyQ Balance , 2021, Genome biology and evolution.

[3]  Carlos A. Matos,et al.  Stress granules, RNA-binding proteins and polyglutamine diseases: too much aggregation? , 2021, Cell Death & Disease.

[4]  S. Hoshino,et al.  Direct evidence that Ataxin-2 is a translational activator mediating cytoplasmic polyadenylation , 2020, The Journal of Biological Chemistry.

[5]  Sung Bae Lee,et al.  The Mechanisms of Nuclear Proteotoxicity in Polyglutamine Spinocerebellar Ataxias , 2020, Frontiers in Neuroscience.

[6]  Carlos A. Matos,et al.  The cholesterol 24-hydroxylase activates autophagy and decreases mutant huntingtin build-up in a neuroblastoma culture model of Huntington’s disease , 2020, BMC Research Notes.

[7]  P. Ivanov,et al.  Phosphorylation of G3BP1-S149 does not influence stress granule assembly , 2019, The Journal of cell biology.

[8]  K. Lam,et al.  The stress granule protein G3BP1 binds viral dsRNA and RIG-I to enhance interferon-β response , 2019, The Journal of Biological Chemistry.

[9]  W. Xue,et al.  G3BP1 promotes DNA binding and activation of cGAS , 2018, Nature Immunology.

[10]  Carlos A. Matos,et al.  Machado–Joseph disease/spinocerebellar ataxia type 3: lessons from disease pathogenesis and clues into therapy , 2018, Journal of neurochemistry.

[11]  D. Kennedy,et al.  Rasputin a decade on and more promiscuous than ever? A review of G3BPs , 2018, Biochimica et Biophysica Acta (BBA) - Molecular Cell Research.

[12]  Edsel A. Peña,et al.  Axonal G3BP1 stress granule protein limits axonal mRNA translation and nerve regeneration , 2018, Nature Communications.

[13]  Lance T. Pflieger,et al.  Gene co-expression network analysis for identifying modules and functionally enriched pathways in SCA2 , 2017, Human molecular genetics.

[14]  U. Stochaj,et al.  Cytoplasmic stress granules: Dynamic modulators of cell signaling and disease. , 2017, Biochimica et biophysica acta. Molecular basis of disease.

[15]  S. Carmo-Silva,et al.  Unraveling the Role of Ataxin-2 in Metabolism , 2017, Trends in Endocrinology & Metabolism.

[16]  Carlos A. Matos,et al.  Proteolytic Cleavage of Polyglutamine Disease-Causing Proteins: Revisiting the Toxic Fragment Hypothesis. , 2017, Current pharmaceutical design.

[17]  B. Blencowe,et al.  Preferential binding of a stable G3BP ribonucleoprotein complex to intron‐retaining transcripts in mouse brain and modulation of their expression in the cerebellum , 2016, Journal of neurochemistry.

[18]  M. Gorospe,et al.  Mammalian ataxin-2 modulates translation control at the pre-initiation complex via PI3K/mTOR and is induced by starvation. , 2016, Biochimica et biophysica acta.

[19]  R. Parker,et al.  Principles and Properties of Stress Granules. , 2016, Trends in cell biology.

[20]  Joana Barbosa de Melo,et al.  Fibroblasts of Machado Joseph Disease patients reveal autophagy impairment , 2016, Scientific Reports.

[21]  H. Hirai,et al.  Re-establishing ataxin-2 downregulates translation of mutant ataxin-3 and alleviates Machado-Joseph disease. , 2015, Brain : a journal of neurology.

[22]  Martin L. Duennwald Cellular stress responses in protein misfolding diseases , 2015, Future science OA.

[23]  C. Larsson,et al.  Regulation of PMP22 mRNA by G3BP1 affects cell proliferation in breast cancer cells , 2013, Molecular Cancer.

[24]  O. Kristensen,et al.  Crystal Structures of the Human G3BP1 NTF2-Like Domain Visualize FxFG Nup Repeat Specificity , 2013, PloS one.

[25]  T. Ashizawa,et al.  Generation of Human-Induced Pluripotent Stem Cells to Model Spinocerebellar Ataxia Type 2 In vitro , 2013, Journal of Molecular Neuroscience.

[26]  N. Déglon,et al.  Overexpression of Mutant Ataxin-3 in Mouse Cerebellum Induces Ataxia and Cerebellar Neuropathology , 2013, The Cerebellum.

[27]  Janghoo Lim,et al.  Aggregation formation in the polyglutamine diseases: Protection at a cost? , 2013, Molecules and cells.

[28]  H. Hirai,et al.  Silencing Mutant Ataxin-3 Rescues Motor Deficits and Neuropathology in Machado-Joseph Disease Transgenic Mice , 2013, PloS one.

[29]  Wojciech J. Szlachcic,et al.  Mouse Models of Polyglutamine Diseases: Review and Data Table. Part I , 2012, Molecular Neurobiology.

[30]  S. Kügler,et al.  Calpastatin-mediated inhibition of calpains in the mouse brain prevents mutant ataxin 3 proteolysis, nuclear localization and aggregation, relieving Machado-Joseph disease. , 2012, Brain : a journal of neurology.

[31]  Carlos A. Matos,et al.  Polyglutamine diseases: The special case of ataxin-3 and Machado–Joseph disease , 2011, Progress in Neurobiology.

[32]  A. Takahashi,et al.  Molecular mechanisms involved in adaptive responses to radiation, UV light, and heat. , 2009, Journal of radiation research.

[33]  Philippe Pierre,et al.  SUnSET, a nonradioactive method to monitor protein synthesis , 2009, Nature Methods.

[34]  H. Hirai,et al.  Characterization of mutant mice that express polyglutamine in cerebellar Purkinje cells , 2009, Brain Research.

[35]  A. Koeppen,et al.  Striatal and nigral pathology in a lentiviral rat model of Machado-Joseph disease. , 2008, Human molecular genetics.

[36]  H. Hirai,et al.  Lentivector‐mediated rescue from cerebellar ataxia in a mouse model of spinocerebellar ataxia , 2008, EMBO reports.

[37]  Gabriele Varani,et al.  RNA is rarely at a loss for companions; as soon as RNA , 2008 .

[38]  H. Lehrach,et al.  Ataxin-2 interacts with the DEAD/H-box RNA helicase DDX6 and interferes with P-bodies and stress granules. , 2007, Molecular biology of the cell.

[39]  Stefan M Pulst,et al.  Expansion of the polyQ repeat in ataxin-2 alters its Golgi localization, disrupts the Golgi complex and causes cell death. , 2003, Human molecular genetics.

[40]  W. Welch,et al.  Polyglutamine protein aggregation and toxicity are linked to the cellular stress response. , 2003, Human molecular genetics.

[41]  K. Chébli,et al.  The RasGAP-associated endoribonuclease G3BP assembles stress granules , 2003, The Journal of cell biology.

[42]  P. Anderson,et al.  Stress granules: sites of mRNA triage that regulate mRNA stability and translatability. , 2002, Biochemical Society transactions.

[43]  H. Paulson,et al.  Live-cell imaging reveals divergent intracellular dynamics of polyglutamine disease proteins and supports a sequestration model of pathogenesis , 2002, Proceedings of the National Academy of Sciences of the United States of America.

[44]  Christopher A. Ross,et al.  Lentiviral-Mediated Delivery of Mutant Huntingtin in the Striatum of Rats Induces a Selective Neuropathology Modulated by Polyglutamine Repeat Size, Huntingtin Expression Levels, and Protein Length , 2002, The Journal of Neuroscience.

[45]  J. Tazi,et al.  RasGAP-Associated Endoribonuclease G3BP: Selective RNA Degradation and Phosphorylation-Dependent Localization , 2001, Molecular and Cellular Biology.

[46]  S. Pulst,et al.  Nuclear localization or inclusion body formation of ataxin-2 are not necessary for SCA2 pathogenesis in mouse or human , 2000, Nature Genetics.

[47]  Wei Li,et al.  RNA-Binding Proteins Tia-1 and Tiar Link the Phosphorylation of Eif-2α to the Assembly of Mammalian Stress Granules , 1999, The Journal of cell biology.

[48]  Georg Auburger,et al.  Spinocerebellar ataxia 2 (SCA2): morphometric analyses in 11 autopsies , 1999, Acta Neuropathologica.

[49]  K. Chébli,et al.  A Novel Phosphorylation-Dependent RNase Activity of GAP-SH3 Binding Protein: a Potential Link between Signal Transduction and RNA Stability , 1998, Molecular and Cellular Biology.

[50]  T Yuasa,et al.  Progressive atrophy of cerebellum and brainstem as a function of age and the size of the expanded CAG repeats in the MJD1 gene in Machado‐Joseph disease , 1998, Annals of neurology.

[51]  K. Fischbeck,et al.  Intranuclear Inclusions of Expanded Polyglutamine Protein in Spinocerebellar Ataxia Type 3 , 1997, Neuron.

[52]  Georg Auburger,et al.  Moderate expansion of a normally biallelic trinucleotide repeat in spinocerebellar ataxia type 2 , 1996, Nature Genetics.

[53]  P. Evans,et al.  The RNP domain: a sequence-specific RNA-binding domain involved in processing and transport of RNA. , 1995, Trends in biochemical sciences.

[54]  P. Anderson,et al.  Stressful initiations. , 2002, Journal of cell science.

[55]  J. Mattick,et al.  Characterization of G3BPs: Tissue specific expression, chromosomal localisation and rasGAP120 binding studies , 2001, Journal of cellular biochemistry.

[56]  Y. Arsenijévic,et al.  Self-inactivating lentiviral vectors with enhanced transgene expression as potential gene transfer system in Parkinson's disease. , 2000, Human gene therapy.

[57]  Shigenobu Nakamura,et al.  CAG expansions in a novel gene for Machado-Joseph disease at chromosome 14q32.1 , 1994, Nature Genetics.