scholarly journals RNA splicing alterations induce a cellular stress response associated with poor prognosis in AML

Author(s):  
Govardhan Anande ◽  
Nandan P. Deshpande ◽  
Sylvain Mareschal ◽  
Aarif M. N. Batcha ◽  
Henry R. Hampton ◽  
...  

RNA splicing is a fundamental biological process that generates protein diversity from a finite set of genes. Recurrent somatic mutations of splicing factor genes are relatively uncommon in Acute Myeloid Leukemia (AML, < 20%). We examined whether RNA splicing differences exist in AML even in the absence of splicing factor mutations. Analyzing RNA-seq data from two independent cohorts of AML patients, we identified recurrent differential alternative splicing between patients with poor and good prognosis. These alternative splicing events occurred even in patients without any discernible splicing factor mutations. The alternative splicing events recurrently occurred in genes involved in specific molecular functions, primarily related to protein translation. Developing informatics tools to predict the functional impact of alternative splicing on the translated protein, we discovered that ~45% of the splicing events directly affected highly conserved protein domains. Several splicing factors were themselves misspliced in patients, and the splicing of their target transcripts were also altered. By studying differential gene expression in the same patients, we identified that alternative splicing of protein translation genes in ELNAdv patients resulted in the induction of an integrated stress response and up- regulation of inflammation-related genes. Lastly, using machine learning techniques, we identified a set of four genes whose alternative splicing can refine the accuracy of existing risk prognosis schemes and validated it in a completely independent cohort. Our discoveries therefore identify aberrant alternative splicing as a molecular feature of adverse AML with clinical relevance.

RNA ◽  
2007 ◽  
Vol 13 (11) ◽  
pp. 1988-1999 ◽  
Author(s):  
M. Alberstein ◽  
M. Amit ◽  
K. Vaknin ◽  
A. O'Donnell ◽  
C. Farhy ◽  
...  

2020 ◽  
Vol 26 (14) ◽  
pp. 3597-3607 ◽  
Author(s):  
Govardhan Anande ◽  
Nandan P. Deshpande ◽  
Sylvain Mareschal ◽  
Aarif M.N. Batcha ◽  
Henry R. Hampton ◽  
...  

2021 ◽  
Author(s):  
Samantha C Chomyshen ◽  
Cheng-Wei Wu

Splicing of pre-mRNA is an essential process for dividing cells and splicing defects have been linked to aging and various chronic diseases. Environmental stress has recently been shown to alter splicing fidelity and molecular mechanisms that protect against splicing disruption remains unclear. Using an in vivo RNA splicing reporter, we performed a genome-wide RNAi screen in Caenorhabditis elegans and found that protein translation suppression via silencing of the conserved initiation factor 4G (IFG-1/eIF4G) protects against cadmium-induced splicing disruption. Transcriptome analysis of an ifg-1 deficient mutant revealed an overall increase in splicing fidelity and resistance towards cadmium-induced alternative splicing compared to the wild-type. We found that the ifg-1 mutant up-regulates >80 RNA splicing regulatory genes that are controlled by the TGF-β transcription factor SMA-2. The extended lifespan of the ifg-1 mutant is partially reduced upon sma-2 depletion and completely nullified when core spliceosome genes including snr-1, snr-2, and uaf-2 are knocked down. Together, these data describe a molecular mechanism that provides resistance towards stress-induced alternative splicing and demonstrate an essential role for RNA homeostasis in promoting longevity in a translation-compromised mutant.


2013 ◽  
Vol 2013 ◽  
pp. 1-14 ◽  
Author(s):  
A. S. Dubrovina ◽  
K. V. Kiselev ◽  
Yu. N. Zhuravlev

Plants are sessile organisms capable of adapting to various environmental constraints, such as high or low temperatures, drought, soil salinity, or pathogen attack. To survive the unfavorable conditions, plants actively employ pre-mRNA splicing as a mechanism to regulate expression of stress-responsive genes and reprogram intracellular regulatory networks. There is a growing evidence that various stresses strongly affect the frequency and diversity of alternative splicing events in the stress-responsive genes and lead to an increased accumulation of mRNAs containing premature stop codons, which in turn have an impact on plant stress response. A number of studies revealed that some mRNAs involved in plant stress response are spliced counter to the traditional conception of alternative splicing. Such noncanonical mRNA splicing events includetrans-splicing, intraexonic deletions, or variations affecting multiple exons and often require short direct repeats to occur. The noncanonical alternative splicing, along with common splicing events, targets the spliced transcripts to degradation through nonsense-mediated mRNA decay or leads to translation of truncated proteins. Investigation of the diversity, biological consequences, and mechanisms of the canonical and noncanonical alternative splicing events will help one to identify those transcripts which are promising for using in genetic engineering and selection of stress-tolerant plants.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 457-457
Author(s):  
Govardhan Anande ◽  
Ashwin Unnikrishnan ◽  
Nandan Deshpande ◽  
Sylvain Mareschal ◽  
Aarif M. N. Batcha ◽  
...  

RNA splicing is a fundamental biological process that generates protein diversity from a finite set of genes. Recurrent somatic mutations of genes involved in RNA splicing are present at high frequency in Myelodysplasia (up to 70%) but less so in Acute Myeloid Leukemia (AML; less than 20%). To investigate whether there were aberrant and recurrent RNA splicing events in the AML transcriptome that were associated with poor prognosis in the absence of splicing factor mutations, we developed a bioinformatics pipeline to systematically annotate and quantify alternative splicing events from RNA-sequencing data (Fig A). We first analysed publicly available RNA-seq data from The Cancer Genome Atlas (TCGA, n=170). We focussed on non-M3 AML patients with no splicing factor mutations (based on reported genomic sequencing and verified by re-analysis of RNA-seq data from all patients) who had received intensive chemotherapy. We segregated these patients based on their European Leukaemia Net (ELN) risk classification and identified 1290 alternatively spliced events impacting 910 genes that were significantly different (FDR&lt;0.05) between all ELNAdv (n=41) versus all ELNFav patients (n=21, Fig B). The majority were exon skipping events (716 events, 62%, Fig B-C), followed by intron retention (201 events, 15.6%, Fig B). We next used RNA-seq data from a second non-M3 AML patient cohort (ClinSeq- Sweden; ELNAdv, n=75 and ELNFav, n=47), detecting 2507 events mapping to 1566 genes. Comparing across the two cohorts, 222 shared genes were detected to be affected by alternative splicing (Fig D). Ingenuity pathway analysis associated these genes with pathways related to protein translation. In order to prioritise those alternatively spliced events most likely to have a deleterious function, we developed an analytical framework to predict their impact on protein structure (Fig E). 87 alternatively spliced events, 25.81% of the commonly shared splicing events, relating to 78 genes (35.13% of all genes) were predicted to directly alter highly conserved protein domains within the affected genes, leading to either a complete (~25%, Fig E) or a partial loss of a domain (20%, Fig E). These in silico predictions are likely to be an underestimate of the true impact, as splicing alterations mapping to poorly annotated domains or affecting the tertiary structure of proteins would be missed. A number of splicing factors themselves were differentially spliced, with the alternative splicing predicted to have functional consequences. This was exemplified by hnRNPA1, a factor with well-established roles in splicing, is itself alternatively spliced in patients and predicted to be deleterious. Consistent with this, motif scanning analyses indicated that a number of mis-spliced transcripts had hnRNPA1 binding motifs (Fig F). To assess the impact of these alternatively spliced events (that were predicted to also disrupt highly conserved protein domains) on the transcriptome, we simultaneously quantified differential gene expression. IPA analysis of the 602 genes that were differentially expressed between ELNAdv and ELNFav patients and shared between both TCGA and ClinSeq cohorts indicated that they were associated with pathways (Fig G) that were distinct from those associated with aberrantly spliced genes (Fig D). A number of pathways related to inflammation were enriched amongst the genes observed to be upregulated in ELNAdv patients (Fig G). Network analyses integrating the alternatively spliced genes with differentially expressed genes revealed strong interactions (Fig H), indicating functional associations between these biological events. Given these strong network interactions, we investigated the potential prognostic significance of these alternatively spliced events. To this end, we utilised machine-learning methods to derive a "splicing signature" of four mis-spliced genes with a predictive capacity equivalent to the ELN (Fig I). The splicing signature further refined existing risk prediction algorithms to improve the classification of patients (Fig J). Taken together, we report the presence of extensive deregulation of RNA splicing in AML patients even in the absence of splicing factor mutations. Many of these events were shared in patients with adverse outcomes and their impact on the AML transcriptome points towards vulnerabilities that could be targeted. Figure Disclosures Unnikrishnan: Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Lehmann:TEVA: Consultancy, Membership on an entity's Board of Directors or advisory committees; Pfizer: Membership on an entity's Board of Directors or advisory committees; Abbive: Membership on an entity's Board of Directors or advisory committees. Pimanda:Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding.


2011 ◽  
Vol 92 (12) ◽  
pp. 2889-2899 ◽  
Author(s):  
Florencia N. Linero ◽  
María G. Thomas ◽  
Graciela L. Boccaccio ◽  
Luis A. Scolaro

Stress granules (SGs) are ephemeral cytoplasmic aggregates containing stalled translation preinitiation complexes involved in mRNA storage and triage during the cellular stress response. SG formation is triggered by the phosphorylation of the alpha subunit of eIF2 (eIF2α), which provokes a dramatic blockage of protein translation. Our results demonstrate that acute infection of Vero cells with the arenavirus Junín (JUNV), aetiological agent of Argentine haemorrhagic fever, does not induce the formation of SGs. Moreover, JUNV negatively modulates SG formation in infected cells stressed with arsenite, and this inhibition correlates with low levels of eIF2α phosphorylation. Transient expression of JUNV nucleoprotein (N) or the glycoprotein precursor (GPC), but not of the matrix protein (Z), inhibits SG formation in a similar manner, comparable to infectious virus. Expression of N and GPC also impaired eIF2α phosphorylation triggered by arsenite. A moderate inhibition of SG formation was also observed when DTT and thapsigargin were employed as stress inducers. In contrast, no inhibition was observed when infected cells were treated with hippuristanol, a translational inhibitor and inducer of SGs that bypasses the requirement for eIF2α phosphorylation. Finally, we analysed SG formation in persistently JUNV-infected cells, where N and GPC are virtually absent and truncated N products are expressed abundantly. We found that persistently infected cells show a quite normal response to arsenite, with SG formation comparable to that of uninfected cells. This suggests that the presence of GPC and/or N is crucial to control the stress response upon JUNV infection of Vero cells.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4110-4110
Author(s):  
Tran Ngoc-Tung ◽  
Hairui Su ◽  
Yuheng Lu ◽  
Christina S Leslie ◽  
Xinyang Zhao

Abstract RBM15, an RNA binding protein belongs to the SPEN family, which is evolutionally conserved from plant to mammals. The relevance of RBM15 to blood diseases came to spot light when RBM15 was discovered to be involved in chromosome translocation t(1;22) in acute megakaryoblastic leukemia. RBM15 is indispensible for the self-renewal of hematopoietic stem cells in response to stress and for megakaryocyte terminal differentiation. RBM15 knockdown results in the unchecked generation of megakaryocyte progenitors in mouse models. At the molecular level, RBM15 have been shown to be a transcriptional repressor in NOCTH signaling pathway by recruiting HDAC complexes like another family member SHARP. RBM15 interacts with transcriptional coactivator SET1/ASH2. RBM15 controls the alternative splicing of thrombopoietin receptor (MPL). However, the molecular mechanisms on how RBM15 regulates alternative RNA splicing is less known. We identified the RBM15-associated proteins with mass spectrometry analysis. Pathway analysis found that RBM15 is mainly associated with proteins involved in RNA splicing, which is also validated by immune-fluorescent microscopy with anti-RBM15 antibody. Among the RBM15-associated proteins are SF3B1 and U2AF, which are heavily mutated in myeloid dysplasia syndrome and chronic lymphocytic leukemia. The interaction between RBM15 and SF3B1 is further validated by co-immunoprecipitation assays. Furthermore, we found that RBM15-SF3B1 interaction is regulated by protein arginine methylation of RBM15. RBM15 is responsible for recruiting SF3B1 complex to the pre-mRNA intronic regions with weak U2snRNP binding sites. We did pair-end, 100bp read RNA-seq of RNA isolated from RBM15 knockdown leukemia cells, and used RBM15 antibody to immunoprecipiate sonicated RNAs to identify intronic regions bound by RBM15. Combining these next generation sequencing data, we found about 50% of the RBM15 binding sites are in the intronic regions and 45% of the RBM15 binding sites are in the 3'UTR regions. RBM15 regulates alternative splicing of a few known transcription factors involved in hematopoiesis such as GATA1, Fli1, GFIb, and RUNX1 as well as chromatin binding proteins such as BRD4, HDAC7 etc.. Therefore, RBM15-SF3B1 interaction fine-tunes the optimal dose of transcription factors needed for the generation of blood lineages in response to environmental stress. How the mutant SF3B1 proteins disrupt its interaction with RBM15 will be discussed. In addition to RBM15-mediated RNA splicing, we also found that RBM15 per se is regulated by its antisense counterpart (AS-RBM15). AS-RBM15 is a long non-coding RNA whose transcription is activated by RUNX1. AS-RBM15 promotes megakaryocyte differentiation by activating RBM15 protein translation. Therefore, RBM15 and AS-RBM15 are at the center of a RNA regulatory network, which is linked to epigenetic programs regulated by RUNX1 and GATA1. Given that both RUNX1 and SF3B1 are often mutated in myeloid dysplasia syndrome, understanding RBM15-mediated gene regulation offers much needed knowledge for understanding MDS. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document