21 research outputs found

    ATXN2-CAG42 sequesters PABPC1 into insolubility and induces FBXW8 in cerebellum of old ataxic knock-in mice

    Get PDF
    Spinocerebellar Ataxia Type 2 (SCA2) is caused by expansion of a polyglutamine encoding triplet repeat in the human ATXN2 gene beyond (CAG)31. This is thought to mediate toxic gain-of-function by protein aggregation and to affect RNA processing, resulting in degenerative processes affecting preferentially cerebellar neurons. As a faithful animal model, we generated a knock-in mouse replacing the single CAG of murine Atxn2 with CAG42, a frequent patient genotype. This expansion size was inherited stably. The mice showed phenotypes with reduced weight and later motor incoordination. Although brain Atxn2 mRNA became elevated, soluble ATXN2 protein levels diminished over time, which might explain partial loss-of-function effects. Deficits in soluble ATXN2 protein correlated with the appearance of insoluble ATXN2, a progressive feature in cerebellum possibly reflecting toxic gains-of-function. Since in vitro ATXN2 overexpression was known to reduce levels of its protein interactor PABPC1, we studied expansion effects on PABPC1. In cortex, PABPC1 transcript and soluble and insoluble protein levels were increased. In the more vulnerable cerebellum, the progressive insolubility of PABPC1 was accompanied by decreased soluble protein levels, with PABPC1 mRNA showing no compensatory increase. The sequestration of PABPC1 into insolubility by ATXN2 function gains was validated in human cell culture. To understand consequences on mRNA processing, transcriptome profiles at medium and old age in three different tissues were studied and demonstrated a selective induction of Fbxw8 in the old cerebellum. Fbxw8 is encoded next to the Atxn2 locus and was shown in vitro to decrease the level of expanded insoluble ATXN2 protein. In conclusion, our data support the concept that expanded ATXN2 undergoes progressive insolubility and affects PABPC1 by a toxic gain-of-function mechanism with tissuespecific effects, which may be partially alleviated by the induction of FBXW8

    The characterization of the Atxn2-CAG42-knock-in mouse as a model for Spinocerebellar Ataxia Type 2

    No full text
    Die Ursache der neurodegenerativen Erkrankung Spinozerebelläre Ataxie Typ 2 (SCA2) ist eine expandierte Polyglutamin-Domäne im humanen ATXN2-Gen von normalerweise 22 auf über 31 CAGs. Von der Degeneration sind vorwiegend die zerebellären Purkinje Neuronen betroffen, in denen zunehmend zytoplasmatische Aggregate sichtbar werden. Auch wenn die genaue Funktion von ATXN2 und die zugrunde liegenden molekularen Mechanismen noch immer ungeklärt sind, werden ein toxischer Funktionsgewinn sowie der Verlust der normalen Proteinfunktion als mögliche Ursachen diskutiert.rnUm ein wirklichkeitsgetreues Tiermodell für die SCA2 zu haben, wurde eine knock-in Maus generiert, deren einzelnes CAG im Atxn2-Gen durch 42 CAGs ersetzt wurde. Dieses Mausmodell ist durch eine stabile Vererbung der Expansion charakterisiert. Weiterhin zeigt sie ein verringertes Körpergewicht sowie eine spät beginnende motorische Inkoordination, was dem Krankheitsbild von SCA2 entspricht. rnIm Weiteren konnte gezeigt werden, dass, obwohl die Atxn2 mRNA-Spiegel in Großhirn und Kleinhirn erhöht waren, die Menge an löslichem ATXN2 im Laufe der Zeit abnahm und dies mit einem Auftreten an unlöslichem ATXN2 korrelierte. Dieser im Kleinhirn progressive Prozess resultierte schließlich in zytoplasmatischen Aggregaten innerhalb der Purkinje Neuronen alter Mäuse. Der Verlust an löslichem ATXN2 könnte Effekte erklären, die auf einen partiellen Funktionsverlust von ATXN2 zurückzuführen sind, wobei die Aggregatbildung einen toxischen Funktionsgewinn wiederspiegeln könnte. Neben ATXN2 wurde auch sein Interaktor PABPC1 zunehmend unlöslich. Während dies im Großhirn eine Erhöhung der PABPC1 mRNA- und löslichen Proteinspiegel zur Folge hatte, konnte keine kompensatorische Veränderung seiner mRNA und zudem eine Verminderung an löslichem PABPC1 im Kleinhirn beobachtet werden. Auch PABPC1 wurde in Aggregate sequestriert. Diese Unterschiede zwischen Großhirn und Kleinhirn könnten zu der spezifischen Vulnerabilität des Kleinhirns beitragen.rnUm die Folgen auf mRNA-Prozessierung zu untersuchen, wurde ein Transkriptomprofil im mittleren sowie fortgeschrittenen Alter der Mäuse erstellt. Hierbei war eine erhöhte Expression von Fbxw8 im Kleinhirn alter Mäuse auffällig. Als Komponente eines Ubiquitin-E3-Ligase-Komplexes, hilft FBXW8 in der Degradierung von Zielproteinen und könnte somit die Toxizität des expandieren ATXN2 verringern. rnZur näheren Beschreibung der physiologischen Funktion von ATXN2, konnte in ATXN2-knock-out Mäusen gezeigt werden, dass das Fehlen von ATXN2 zu einer reduzierten globalen Proteinsyntheserate führte und somit eine Rolle als Translationsaktivator möglich erscheint. Kompensatorisch wurde eine erhöhte S6-Phosphorylierung gemessen.The neurodegenerative disease Spinocerebellar Ataxia Type 2 (SCA2) is caused by an expanded polyglutamine domain in the human ATXN2 gene from usually 22 to more than 31 CAGs. The primary site of degeneration is the cerebellar Purkinje neurons in which cytoplasmic aggregates become visible during late stages of the disease. The exact function of ATXN2 and the underlying molecular mechanisms are still not fully understood, while both toxic gain-of-function as well as partial loss-of-function mechanisms are under discussion.rnAs a faithful animal model, a knock-in mouse replacing the single CAG of murine Atxn2 with CAG42 was generated. This expansion size was inherited stably. The mice showed phenotypes with reduced weight and later motor incoordination, which resembles the clinical picture of SCA2. rnAlthough Atxn2 mRNA became elevated in cortex and cerebellum, soluble ATXN2 protein levels diminished over time and this further correlated with an increase of insoluble ATXN2. This feature was progressive in the cerebellum and resulted in cytoplasmic aggregates within the Purkinje neurons of old mice. The reduced soluble ATXN2 levels might explain partial loss-of-function effects, while the appearance of aggregates possibly reflects toxic gains-of-function. Besides ATXN2, also its interactor PABPC1 became progressively insoluble. This was accompanied by increased transcript and soluble protein levels in the cortex, while no compensatory regulation of PABPC1 mRNA and a reduction of soluble PABPC1 were observed in the cerebellum. PABPC1 was also sequestered into aggregates. These different observations in cortex and cerebellum might explain the specific vulnerability of the cerebellum.rnTo understand consequences on mRNA processing, transcriptome profiles at medium and old age were studied and demonstrated a selective induction of Fbxw8 in the old cerebellum. As a component of an ubiquitin-E3-ligase complex, FBXW8 aids in the degradation of target proteins and might thus reduce the toxicity of the expanded ATXN2.rnTo further describe the physiological function of ATXN2, an Ataxin-2 knock-out mouse model was exploited. The loss of ATXN2 resulted in diminished global protein synthesis rates, which make a role as translational activator possible. As compensatory effort, an increased S6-phosphorylation was observed. r

    Atxn2 knockout and CAG42-knock-in cerebellum shows similarly dysregulated expression in calcium homeostasis pathway

    No full text
    Spinocerebellar ataxia type 2 (SCA2) is an autosomal dominantly inherited neurodegenerative disorder with preferential affection of Purkinje neurons, which are known as integrators of calcium currents. The expansion of a polyglutamine (polyQ) domain in the RNA-binding protein ataxin-2 (ATXN2) is responsible for this disease, but the causal roles of deficient ATXN2 functions versus aggregation toxicity are still under debate. Here, we studied mouse mutants with Atxn2 knockout (KO) regarding their cerebellar global transcriptome by microarray and RT-qPCR, in comparison with data from Atxn2-CAG42-knock-in (KIN) mouse cerebellum. Global expression downregulations involved lipid and growth signaling pathways in good agreement with previous data. As a novel effect, downregulations of key factors in calcium homeostasis pathways (the transcription factor Rora, transporters Itpr1 and Atp2a2, as well as regulator Inpp5a) were observed in the KO cerebellum, and some of them also occurred subtly early in KIN cerebellum. The ITPR1 protein levels were depleted from soluble fractions of cerebellum in both mutants, but accumulated in its membrane-associated form only in the SCA2 model. Coimmunoprecipitation demonstrated no association of ITPR1 with Q42-expanded or with wild-type ATXN2. These findings provide evidence that the physiological functions and protein interactions of ATXN2 are relevant for calcium-mediated excitation of Purkinje cells as well as for ATXN2-triggered neurotoxicity. These insights may help to understand pathogenesis and tissue specificity in SCA2 and other polyQ ataxias like SCA1, where inositol regulation of calcium flux and RORalpha play a role

    Both ubiquitin ligases FBXW8 and PARK2 are sequestrated into insolubility by ATXN2 polyQ expansions, but only FBXW8 expression is dysregulated

    No full text
    The involvement of the ubiquitin-proteasome system (UPS) in the course of various age-associated neurodegenerative diseases is well established. The single RING finger type E3 ubiquitin-protein ligase PARK2 is mutated in a Parkinson’s disease (PD) variant and was found to interact with ATXN2, a protein where polyglutamine expansions cause Spinocerebellar ataxia type 2 (SCA2) or increase the risk for Levodopa-responsive PD and for the motor neuron disease Amyotrophic lateral sclerosis (ALS). We previously reported evidence for a transcriptional induction of the multi-subunit RING finger Skp1/Cul/F-box (SCF) type E3 ubiquitin-protein ligase complex component FBXW8 in global microarray profiling of ATXN2-expansion mouse cerebellum and demonstrated its role for ATXN2 degradation in vitro. Now, we documented co-localization in vitro and co-immunoprecipitations both in vitro and in vivo, which indicate associations of FBXW8 with ATXN2 and PARK2. Both FBXW8 and PARK2 proteins are driven into insolubility by expanded ATXN2. Whereas the FBXW8 transcript upregulation by ATXN2- expansion was confirmed also in qPCR of skin fibroblasts and blood samples of SCA2 patients, a FBXW8 expression dysregulation was not observed in ATXN2-deficient mice, nor was a PARK2 transcript dysregulation observed in any samples. Jointly, all available data suggest that the degradation of wildtype and mutant ATXN2 is dependent on FBXW8, and that ATXN2 accumulation selectively modulates FBXW8 levels, while PARK2 might act indirectly through FBXW8. The effects of ATXN2-expansions on FBXW8 expression in peripheral tissues like blood may become useful for clinical diagnostic

    Genetic ablation of ataxin-2 increases several global translation factors in their transcript abundance but decreases translation rate

    Get PDF
    Spinocerebellar ataxia type 2 (SCA2) and amyotrophic lateral sclerosis (ALS) are neurodegenerative disorders, caused or modified by an unstable CAG-repeat expansion in the SCA2 gene, which encodes a polyglutamine (polyQ) domain expansion in ataxin-2 (ATXN2). ATXN2 is an RNA-binding protein and interacts with the poly(A)-binding protein PABPC1, localizing to ribosomes at the rough endoplasmic reticulum. Under cell stress, ATXN2, PABPC1 and small ribosomal subunits are relocated to stress granules, where mRNAs are protected from translation and from degradation. It is unknown whether ATXN2 associates preferentially with specific mRNAs or how it modulates RNA processing. Here, we investigated the RNA profile of the liver and cerebellum from Atxn2 knockout (Atxn2−/−) mice at two adult ages, employing oligonucleotide microarrays. Prominent increases were observed for Lsm12/Paip1 (>2-fold), translation modulators known as protein interactor/competitor of ATXN2 and for Plin3/Mttp (>1.3-fold), known as apolipoprotein modulators in agreement with the hepatosteatosis phenotype of the Atxn2−/− mice. Consistent modest upregulations were also observed for many factors in the ribosome and the translation/secretion apparatus. Quantitative reverse transcriptase PCR in liver tissue validated >1.2-fold upregulations for the ribosomal biogenesis modulator Nop10, the ribosomal components Rps10, Rps18, Rpl14, Rpl18, Gnb2l1, the translation initiation factors Eif2s2, Eif3s6, Eif4b, Pabpc1 and the rER translocase factors Srp14, Ssr1, Sec61b. Quantitative immunoblots substantiated the increased abundance of NOP10, RPS3, RPS6, RPS10, RPS18, GNB2L1 in SDS protein fractions, and of PABPC1. In mouse embryonal fibroblasts, ATXN2 absence also enhanced phosphorylation of the ribosomal protein S6 during growth stimulation, while impairing the rate of overall protein synthesis rates, suggesting a block between the enhanced translation drive and the impaired execution. Thus, the physiological role of ATXN2 subtly modifies the abundance of cellular translation factors as well as global translation

    APADB : a database for alternative polyadenylation and microRNA regulation events

    No full text
    Alternative polyadenylation (APA) is a widespread mechanism that contributes to the sophisticated dynamics of gene regulation. Approximately 50% of all protein-coding human genes harbor multiple polyadenylation (PA) sites; their selective and combinatorial use gives rise to transcript variants with differing length of their 3' untranslated region (3'UTR). Shortened variants escape UTR-mediated regulation by microRNAs (miRNAs), especially in cancer, where global 3'UTR shortening accelerates disease progression, dedifferentiation and proliferation. Here we present APADB, a database of vertebrate PA sites determined by 3' end sequencing, using massive analysis of complementary DNA ends. APADB provides (A)PA sites for coding and non-coding transcripts of human, mouse and chicken genes. For human and mouse, several tissue types, including different cancer specimens, are available. APADB records the loss of predicted miRNA binding sites and visualizes next-generation sequencing reads that support each PA site in a genome browser. The database tables can either be browsed according to organism and tissue or alternatively searched for a gene of interest. APADB is the largest database of APA in human, chicken and mouse. The stored information provides experimental evidence for thousands of PA sites and APA events. APADB combines 3' end sequencing data with prediction algorithms of miRNA binding sites, allowing to further improve prediction algorithms. Current databases lack correct information about 3'UTR lengths, especially for chicken, and APADB provides necessary information to close this gap. Database URL: http://tools.genxpro.net/apadb

    FBXW8 is shifted into insolubility in <i>Atxn2</i>-CAG42-KIN mice due to interaction with expanded ATXN2.

    No full text
    <p>(A) Pulling either with anti-ATXN2 or anti-FBXW8 antibody, ATNX2 and FBXW8 show an interaction in the cerebellum of 18-month-old <i>Atxn2</i>-CAG42-KIN mice independent of the polyQ length (experiment repeated three times for anti-ATXN2 and once with anti-FBXW8, representative images). (B) In cerebellar tissue of 18-month-old <i>Atxn2</i>-CAG42-KIN mice FBXW8 protein level is downregulated in the RIPA-soluble fraction while it is upregulated in the SDS-soluble fraction (two independent experiments each with 4 <i>Atxn2</i><sup>CAG1/CAG1</sup> vs. 4 <i>Atxn2</i><sup>CAG42/CAG42</sup> mice).</p

    FBXW8 protein levels are dysregulated in SCA2 patient material.

    No full text
    <p>FBXW8 expression is upregulated at the transcript level in SCA2 patient skin fibroblasts (A; 4 CTL individuals vs. 4 SCA2 patients) as well as in SCA2 patient blood samples (B; 5 CTL individuals vs. 3 SCA2 patients). (C) At the protein level FBXW8 is decreased in the RIPA-soluble fraction while it is increased in the SDS-soluble fraction in SCA2 patient fibroblasts (4 CTL individuals vs. 4 SCA2 patients).</p

    Search for SCA2 blood RNA biomarkers highlights Ataxin-2 as strong modifier of the mitochondrial factor PINK1 levels

    No full text
    Ataxin-2 (ATXN2) polyglutamine domain expansions of large size result in an autosomal dominantly inherited multi-system-atrophy of the nervous system named spinocerebellar ataxia type 2 (SCA2), while expansions of intermediate size act as polygenic risk factors for motor neuron disease (ALS and FTLD) and perhaps also for Levodopa-responsive Parkinson's disease (PD)
    corecore