scholarly journals A Drosophila Model of Pontocerebellar Hypoplasia Reveals a Critical Role for the RNA Exosome in Neurons

2019 ◽  
Author(s):  
Derrick J. Morton ◽  
Binta Jalloh ◽  
Lily Kim ◽  
Isaac Kremsky ◽  
Rishi J. Nair ◽  
...  

AbstractThe RNA exosome is an evolutionarily-conserved ribonuclease complex critically important for precise processing and/or complete degradation of a variety of cellular RNAs. The recent discovery that mutations in genes encoding structural RNA exosome subunits cause tissue-specific diseases makes defining the role of this complex within specific tissues critically important. Mutations in the RNA exosome component 3 (EXOSC3) gene cause Pontocerebellar Hypoplasia Type 1b (PCH1b), an autosomal recessive neurologic disorder. The majority of disease-linked mutations are missense mutations that alter evolutionarily-conserved regions of EXOSC3. The tissue-specific defects caused by these amino acid changes in EXOSC3 are challenging to understand based on current models of RNA exosome function with only limited analysis of the complex in any multicellular model in vivo. The goal of this study is to provide insight into how mutations in EXOSC3 impact the function of the RNA exosome. To assess the tissue-specific roles and requirements for the Drosophila ortholog of EXOSC3 termed Rrp40, we utilized tissue-specific RNAi drivers. Depletion of Rrp40 in different tissues reveals a general requirement for Rrp40 in the development of many tissues including the brain, but also highlight an agedependent requirement for Rrp40 in neurons. To assess the functional consequences of the specific amino acid substitutions in EXOSC3 that cause PCH1b, we used CRISPR/Cas9 gene editing technology to generate flies that model this RNA exosome-linked disease. These flies show reduced viability; however, the surviving animals exhibit a spectrum of behavioral and morphological phenotypes. RNA-seq analysis of these Drosophila Rrp40 mutants reveals increases in the steady-state levels of specific mRNAs and ncRNAs, some of which are central to neuronal function. In particular, Arc1 mRNA, which encodes a key regulator of synaptic plasticity, is increased in the Drosophila Rrp40 mutants. Taken together, this study defines a requirement for the RNA exosome in specific tissues/cell types and provides insight into how defects in RNA exosome function caused by specific amino acid substitutions that occur in PCH1b can contribute to neuronal dysfunction.Author SummaryPontocerebellar Hypoplasia Type 1b (PCH1b) is a devastating genetic neurological disorder that preferentially affects specific regions of the brain. Typically, children born with PCH1b have structural defects in regions of the brain including those associated with key autonomic functions. Currently, there is no cure or treatment for the disease. PCH1b is caused by mutations in the RNA exosome component 3 (EXOSC3) gene, which encodes a structural component of the ubiquitous and essential multi-subunit RNA exosome complex. The RNA exosome is critical for both precise processing and turnover of multiple classes of RNAs. To elucidate the functional consequences of amino acid changes in EXOSC3 that cause PCH1b, we exploited well-established genetic approaches in Drosophila melanogaster that model EXOSC3 mutations found in individuals with PCH1b. Using this system, we find that the Drosophila EXOSC3 homolog (termed Rrp40) is essential for normal development and has an important function in neurons. Furthermore, PCH1b missense mutations modeled in Rrp40 cause reduced viability and produce neuronal-specific phenotypes that correlate with altered levels of target RNAs that encode factors with key roles in neurons. These results provide a basis for understanding how amino acid changes that occur in the RNA exosome contribute to neuronal dysfunction and disease.

Genetics ◽  
2016 ◽  
Vol 205 (1) ◽  
pp. 221-237 ◽  
Author(s):  
Milo B. Fasken ◽  
Jillian S. Losh ◽  
Sara W. Leung ◽  
Sergine Brutus ◽  
Brittany Avin ◽  
...  

1995 ◽  
Vol 15 (6) ◽  
pp. 2993-3002 ◽  
Author(s):  
S Seidman ◽  
M Sternfeld ◽  
R Ben Aziz-Aloya ◽  
R Timberg ◽  
D Kaufer-Nachum ◽  
...  

Tissue-specific heterogeneity among mammalian acetylcholinesterases (AChE) has been associated with 3' alternative splicing of the primary AChE gene transcript. We have previously demonstrated that human AChE DNA encoding the brain and muscle AChE form and bearing the 3' exon E6 (ACHE-E6) induces accumulation of catalytically active AChE in myotomes and neuromuscular junctions (NMJs) of 2- and 3-day-old Xenopus embryos. Here, we explore the possibility that the 3'-terminal exons of two alternative human AChE cDNA constructs include evolutionarily conserved tissue-recognizable elements. To this end, DNAs encoding alternative human AChE mRNAs were microinjected into cleaving embryos of Xenopus laevis. In contrast to the myotomal expression demonstrated by ACHE-E6, DNA carrying intron 14 and alternative exon E5 (ACHE-I4/E5) promoted punctuated staining of epidermal cells and secretion of AChE into the external medium. Moreover, ACHE-E6-injected embryos displayed enhanced NMJ development, whereas ACHE-I4/E5-derived enzyme was conspicuously absent from muscles and NMJs and its expression in embryos had no apparent effect on NMJ development. In addition, cell-associated AChE from embryos injected with ACHE-I4/E5 DNA was biochemically distinct from that encoded by the muscle-expressible ACHE-E6, displaying higher electrophoretic mobility and greater solubility in low-salt buffer. These findings suggest that alternative 3'-terminal exons dictate tissue-specific accumulation and a particular biological role(s) of AChE, associate the 3' exon E6 with NMJ development, and indicate the existence of a putative secretory AChE form derived from the alternative I4/E5 AChE mRNA.


2009 ◽  
Vol 136 (3) ◽  
pp. 872-882.e3 ◽  
Author(s):  
Simone M.R. Camargo ◽  
Dustin Singer ◽  
Victoria Makrides ◽  
Katja Huggel ◽  
Klaas M. Pos ◽  
...  

2020 ◽  
Author(s):  
Maria C. Sterrett ◽  
Liz Enyenihi ◽  
Sara W. Leung ◽  
Laurie Hess ◽  
Sarah E. Strassler ◽  
...  

AbstractRNA exosomopathies, a growing family of tissue-specific diseases, are linked to missense mutations in genes encoding the structural subunits of the conserved 10-subunit exoribonuclease complex, the RNA exosome. Such mutations in the cap subunit gene EXOSC2 cause the novel syndrome SHRF (Short stature, Hearing loss, Retinitis pigmentosa and distinctive Facies). In contrast, exosomopathy mutations in the cap subunit gene EXOSC3 cause pontocerebellar hypoplasia type 1b (PCH1b). Though having strikingly different disease pathologies, EXOSC2 and EXOSC3 exosomopathy mutations result in amino acid substitutions in similar, conserved domains of the cap subunits, suggesting that these exosomopathy mutations have distinct consequences for RNA exosome function. We generated the first in vivo model of the SHRF pathogenic amino acid substitutions using budding yeast by introducing the EXOSC2 mutations in the orthologous S. cerevisiae gene RRP4. The resulting rrp4 mutant cells have defects in cell growth and RNA exosome function. We detect significant transcriptomic changes in both coding and non-coding RNAs in the rrp4 variant, rrp4-G226D, which models EXOSC2 p.Gly198Asp. Comparing this rrp4-G226D mutant to the previously studied S. cerevisiae model of EXOSC3 PCH1b mutation, rrp40-W195R, reveals that these mutants have disparate effects on certain RNA targets, providing the first evidence for different mechanistic consequences of these exosomopathy mutations. Congruently, we detect specific negative genetic interactions between RNA exosome cofactor mutants and rrp4-G226D but not rrp40-W195R. These data provide insight into how SHRF mutations could alter the function of the RNA exosome and allow the first direct comparison of exosomopathy mutations that cause distinct pathologies.


2020 ◽  
Vol 105 ◽  
pp. 104389
Author(s):  
Liang Yan ◽  
Yongqi Ke ◽  
Yuhe Kan ◽  
Dao Lin ◽  
Jingkui Yang ◽  
...  

Author(s):  
Joshua O. Owolabi ◽  
Philip O. Ogunnaike

The implications of the use of psychoactive substances, that are usually abused on the retina are yet to be adequately explored. The retina is neural in nature. Most investigations on psychoactive agents have only studied their effects on the brain and behaviour. The mechanisms employed by these agents in producing their effects on the brain suggest that the retina, being neural, might also be significantly affected by the use of the substances. This research investigated the effects of the prolonged use of caffeine, nicotine and 4-methylenedioxymethamphetamine [MDMA] on the retina. Juvenile male experimental Wistar were grouped and administered the lower and higher dose of each agent while a reference group remained as the Group A. Groups B and C received the lower [30mg/kg body weight]and the higher [50mg/kg body weight]doses of caffeine respectively; Groups D and E received the lower [10mg/kg body weight] and higher [20mg/kg body weight] doses of nicotine respectively while Groups F and G received the lower [20 mg/kg body weight] and the higher [40 mg/kg body weight] doses of MDMA respectively. The substances had effects on the thickness of the retina with higher doses in each instance causing reductions in retina thickness; the patterns of GFAP expression were also aberrant with the MDMA treated groups being most aberrant. There was no sign of extensive loss of any type of retinal cells. Rhodopsin expression generally demonstrated active rods and provided insight into relatively heathy cones. There is evidence that these agents altered retina thickness and GFAP expression but without extensive disruptions to serve as pathological hallmarks of retina degeneration. The consequences of these might be further investigated.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3890-3890
Author(s):  
Sushree Sangita Sahoo ◽  
Caitlin Roake ◽  
Liliana Rindle ◽  
Claudia Rössig ◽  
Markus Schmugge ◽  
...  

Abstract Dyskeratosis congenita (DC) is a rare telomere disease with pleiotropic manifestations and bone marrow failure (BMF) as a major cause of mortality. The genes involved in DC pathogenesis play a role in telomere maintenance but their function in other in biological systems has not been well characterized. Compound heterozygous missense mutations in TCAB1(WRAP53) were reported so far in two pedigrees with classic DC (Zhong et al, 2011). TCAB1 functions as a scaffold protein recruiting TERC to the sub-organelle Cajal bodies, thus contributing to the assembly of telomerase enzyme; among its other functions it regulates DNA double-strand break repair. Here we functionally characterize novel TCAB1 mutations and describe transcriptome and proteome profiles in patient-derived lymphoblastoid cell lines (LCL). Among a cohort of 50 DC patients we identified three pedigrees with TCAB1 mutations, BMF and telomeres below first percentile. Index 1 carried compound heterozygous mutations R155X/Y345C also present in the affected brother. Both siblings suffered from transfusion-dependent pancytopenia manifesting in the childhood and typical dyskeratotic features. Index 2 harbored a single mutation Q7TfsX27 and presented with hypocellular BMF. Although non-hematologic symptoms were initially absent, the patient suffered from mild restrictive lung disease after stem cell transplantation. Index 3 carried biallelic mutations Y345C/G435R. He was diagnosed with microcephaly, cerebellar hypoplasia, early onset BMF with immunodeficiency, consistent with Hoyeraal-Hreidarsson syndrome (HHS). To gain insight into the functional consequences of novel mutations identified, we first assessed RNA, protein levels and localization in patient-derived LCLs and HeLa cells stably expressing wildtype (WT) and mutant TCAB1. The truncating mutations R155X and Q7TfsX27 were unable to translate to protein with abrogative localization in Cajal bodies, while the R155X mutation affected RNA stability through nonsense-mediated decay. The Y345C mutant was translated, but it was severely diminished in the nucleus and demonstrated defective localization in the Cajal bodies. The decline of TCAB1 signal in Cajal bodies was also observed in patients 1&3 with biallelic mutations. Next, to describe the effect of mutations on cellular processes we performed RNA-sequencing (RNAseq) and mass spectrometric-based quantitative proteomics using SILAC on LCLs from patients/carriers and WT controls. The principal component analysis (PCA) of RNAseq data clusters the transcriptomes of patients with missense mutations (Y345C/G435R, Y345C) and truncating mutations (R155X, Q7TfsX27) into separate groups, while index 1 (R155X/Y345C) demonstrated a very distinct transcriptomic profile (Fig 1A). The telomere maintenance processes were highly downregulated for index 1 and clinically healthy carrier parents (Fig 1B). This might suggest that heterozygous mutations also affect telomere biology; however the compensatory effect from the WT allele allows maintaining normal homeostasis. Based on disease severity and established pathogenic effect of mutations we then focused on index patients 1 and 3 carrying biallelic mutations. The differential gene expression analysis suggested RPSA, LILRB1 (downregulated) and SH3BP5, TSPYL5 (upregulated) as top candidates. Strikingly, the identification of downregulated RPSA points at dysregulated ribosomal function as a possible novel mechanism in TCAB1-mutated cells. Finally, positive correlation was observed for differential gene regulation on transcriptome and protein level. A gene set variation analysis indicated down-regulation of telomere maintenance, protein processing and ubiquitination. Surprisingly the members of the telomerase holoenzyme complex were enriched on the protein level however they were detected transcriptionally down-regulated in patients (Fig 1C). This might hint at defective protein transport or slower protein turnover due to decreased ubiquitination. In summary, we expand the clinical spectrum of TCAB1-associated DC ranging from BMF without mucocutaneous symptoms to severe HHS. We provide additional insights into the underlying cellular effects of TCAB1 mutations. The preliminary insight into the gene networks affected by TCAB1 mutations allows for first comprehension into the pleiotropic effects observed in patients. Disclosures No relevant conflicts of interest to declare.


2016 ◽  
Author(s):  
Adam J Dobson ◽  
Xiaoli He ◽  
Eric Blanc ◽  
Ekin Bolukbasi ◽  
Yodit Feseha ◽  
...  

AbstractAnimal lifespan can be extended by dietary restriction (DR), but at a cost to fitness. This phenomenon depends on essential amino acids (EAAs) and TOR signalling, but roles of specific tissues and downstream transcriptional regulators are poorly characterised. Manipulating relevant transcription factors (TFs) specifically in lifespan-limiting tissues may ameliorate ageing without costs of DR. Here we identify TFs which regulate the DR phenotype in Drosophila, analysing organs as an interacting system and reducing its transcriptional complexity by two orders of magnitude. Evolutionarily conserved GATA TFs are predicted to regulate the overlapping effects of DR and TOR on organs, and genetic analyses confirmed that these TFs interact with diet to determine lifespan. Importantly, Srp knockdown insulated fly lifespan from the pernicious effects of EAAs, but tissue-specific knockdown reduced the corrolary costs. These results provide the first indication that benefits of EAAs for early-life fitness can be decoupled from longevity by tissue-specific transcriptional reprogramming.


2019 ◽  
Author(s):  
Pouria Dasmeh

AbstractTo gain insight into the recent evolution of EBOLA virus (EBOV) proteins and identify evolutionary hotspots, I investigated patterns of amino acid substitutions in the proteomes of 49 members of EBOV genus including EBOV, Bundibugyo virus (BDBV), Reston virus (RESTV), Sudan virus (SUDV) and Taï Forest virus (TAFV) spanning outbreaks from 1974 to 2014 in Democratic Republic of Congo (DRC), Uganda, Gabon, Sudan, Ivory Coast, Philippines, and Guinea among humans, non-human primates, and pigs. Of the seven genes of the Ebola genome, I found significant variations in evolutionary rates of Glycoprotein (GP), RNA/dependent RNA polymerase (L), and Nucleoprotein (NP) across different lineages. GP and NP were found to evolve at 5- and 3-fold higher rate than other EBOV genes. Several residues in GP and NP show significant dN/dS >1 along the internal branches of the phylogenetic tree leading to Philippines and Sudan outbreaks. Most of these residues are located in solvent exposed areas shown previously to be antigenic. We further identified significant changes in specific amino acid properties of GP, NP and L during the recent evolutionary history of EBOV outbreaks and in particular during the 2014 outbreak. The positively selected residues and chemical properties are consistent with changes in epitope interaction and could thus be important in detecting adaptation of EBOV to the host immune defense system.


Sign in / Sign up

Export Citation Format

Share Document