Isogenic iPSC Models of SRSF2-Mutant Myelodysplastic Syndrome Capture Disease Phenotypes, Splicing Defects and Drug Responses

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 962-962
Author(s):  
Chan-Jung Chang ◽  
Andriana Kotini ◽  
Julie Teruya-Feldstein ◽  
Omar Abdel-Wahab ◽  
Robert Bradley ◽  
...  

Abstract A major recent advance for the biology of Myelodysplastic Syndrome (MDS) was the discovery of recurrent mutations in genes encoding splicing factors (SFs). SRSF2 is a SF that binds to RNA sequences called exonic splicing enhancers (ESEs) to promote inclusion of exons containing these motifs. Mutations of SRSF2 are found in 20-30% of MDS patients, are associated with adverse prognosis and are almost always heterozygous missense mutations (P95 L/R/H). This strongly suggests a gain or alteration of function mechanism, corroborated by recent findings by us and others in a knockin mouse model and in K562 cells expressing mutant SRSF2, showing that the SRSF2 P95H mutation changes the normal RNA binding specificity of SRSF2 (Kim et al. Cancer Cell, 2015; Zhang et al. PNAS, 2015). While mis-splicing of hematopoietic regulators, such as EZH2, was proposed as a potential downstream mechanism of SF mutations, systematically identifying the key downstream mediators remains a challenge. No human hematopoietic cell lines harboring SF mutations in the context of a normal diploid genome exist, primary MDS cells afford limited experimental opportunities and mouse modeling is likely to miss disease relevant targets, as only a quarter of regulated alternative splicing events are conserved between human and mouse. Our lab has pioneered the modeling of MDS with human induced pluripotent stem cells (iPSCs). We previously derived iPSCs with the SRSF2 P95L mutation and a chr7q deletion (del7q), as well as normal iPSCs from residual normal hematopoietic cells from the same MDS patient (Kotini et al. Nat. Biotech, 2015). To develop a model of mutant SRSF2, we used CRISPR/Cas9 technology to generate a panel of isogenic iPSCs with or without the SRSF2 P95 mutation, isolated or with the cooperating del(7q), all in the same genetic background, by both introducing the SRSF2 P95L mutation in normal iPSCs and correcting it in MDS-iPSCs from the same patient. Both patient-derived and genetically engineered SRSF2 P95L-iPSCs showed decreased growth and increased cell death of hematopoietic progenitor cells (HPCs), decreased clonogenic capacity and features of morphologic dysplasia. This phenotype is consistent with the SRSF2 mutation being an early, potentially initiating, event, supported by its frequent presence in the dominant clone and in individuals with clonal hematopoiesis of indeterminate potential (CHIP) without overt MDS. Using a competitive cell growth assay, we found that the splicing inhibitor E7107, as well as small molecule inhibitors of kinases modulating splicing, preferentially inhibit the growth of SRSF2 mutant, but not of isogenic normal, iPSC-derived HPCs. To investigate the effects of mutant SRSF2 in mRNA splicing, we performed RNA sequencing of purified CD34+, CD34+/CD45+ HPCs and undifferentiated iPSCs from mutant and isogenic wild-type (WT) iPSCs. SRSF2 mutant cells exhibited genome-wide alterations in ESE preferences, recapitulating the altered RNA binding found in patient cells, with mutant SRSF2 preferentially recognizing a CCNG motif versus a GGNG motif, while WT SRSF2 binds to both with similar affinity. Genes found mis-spliced in the SRSF2 mutant cells included previously reported genes of potential disease relevance, like EZH2 and FYN. While the majority of differentially spliced genes overlapped among the 3 cell states, cell type-specific differences were also noted, highlighting the importance of performing these analyses in the appropriate cell type. Importantly, iPSC-derived HPCs recapitulated a higher percentage of the mis-spliced events observed in patient cells than either the knockin mouse or the K562 models, thus capturing disease-relevant splicing alterations more faithfully than other models. To identify critical direct targets of mutant SRSF2, we used a second round of CRISPR/Cas9 gene editing to introduce a 3xFLAG epitope tag at the carboxyl terminus of the endogenous SRSF2 locus in isogenic SRSF2mutant and WT iPSCs. CLIP-seq experiments in undifferentiated cells and HPCs derived from them are underway to identify genes differentially bound by the mutant vs the WT SRSF2. The model we describe here will be valuable for dissecting the pathogenesis of MDS with SF mutations, testing drugs and identifying new therapeutic targets for drug development. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 457-457 ◽  
Author(s):  
Valeria Visconte ◽  
Hideki Makishima ◽  
Anna M Jankowska ◽  
Fabiola Traina ◽  
Hadrian Szpurka ◽  
...  

Abstract Abstract 457 Ring sideroblasts (RS) are abnormal nucleated erythroblasts characterized by iron granules in mitochondrial cristae. RS are seen in acquired and congenital sideroblastic anemia. Dysfunction in mitochondrial metabolism has been implicated in the pathogenesis of RS. However, the true mechanism leading to RS formation remains elusive. Clonal sideroblastic anemias are usually acquired in the context of MDS: 15% or more RS in the bone marrow (BM) with appropriate morphologic and cytogenetic criteria in MDS is best classified as RARS, but varying quantities of RS (<15%) can also occur in other myeloid malignancies. RARS presenting with thrombocytosis (RARS-T) is a form of MDS/MPN often associated with JAK2, TET2, and MPL mutations. To date, no recurrent mutations or chromosomal defects have been found. We hypothesized that they likely exist in RARS and therefore systematic unbiased application of whole exome next generation sequencing may lead to identification of cryptic clonal mutations of pathogenetic significance. Initially, we applied whole exome sequencing to 15 patients with MDS and found a new somatic mutation in SF3B1 (splicing factor 3b, subunit 1) on chromosome 2q in a case of RARS with high platelets. SF3B1 is a component of the U2-small nuclear ribonucleoprotein complex (U2 snRNP), a part of the U2-dependent spliceosome in eukaryotes. The index mutation resulted in a lysine to glutamic acid substitution (K700E) and its somatic nature was confirmed in non-clonal CD3 cells. Further screening of patients with a similar phenotype (N=81) revealed somatic missense mutations in 9/14 (64%) and 13/18 (72%) patients with RARS and RARS-T, respectively. No mutations were detected in 49 MDS and MDS/MPN patients with <15% RS. All SF3B1 mutations were heterozygous affecting mostly exons 15 [15%] and 14 [7%]; no corresponding hemizygous deletions were found (N=430). Similarly, no mutations were detected in congenital sideroblastic anemias. SF3B1 mutations were associated with JAK2 V617F (4/13; 30%), MPL (3/13; 23%), DNMT3A (1/18; 5%), TET2 (1/18; 5%), ASXL1 (1/18; 5%), and LNK (1/18; 5%) in RARS-T and TET2 (1/14; 7%) and DNMT3A (4/14; 29%) in RARS. There was no difference, adjusted for IPSS in terms of survival and AML progression between wild type (WT) and carriers of SF3B1 but the latter showed a higher frequency of thrombotic events in (p=.04). To confirm the pathogenetic role of SF3B1 mutations and their impact on phenotype, we investigated an engineered SF3B1+/− B6 knockout mouse model. While no overt anemia and only mild leukopenia were observed, BM from SF3B1+/− mice showed numerous RS compared to WT B6 controls, further confirming that SF3B1 alterations may lead to RS. Since around 95% of multiexonic genes are differentially spliced, errors in splicing mechanism are important in maintaining genomic diversity and may lead to cancer; in fact SF3B1 mutations have been previously described in ovarian cancer and melanoma. To assess if other members of the spliceosome machinery are also affected, SF314 and SF3B4 were tested, but to date no mutations were found. Since DYRKA1 encodes a protein that phosphorylates SF3B1 and its mutation could also affect SF3B1 function, we performed Sanger sequencing and found no mutations. Altogether these observations led us to speculate that disruption or mutations in SF3B1 may have a key role in the manifestation of RS phenotype. To examine the functional consequences of defects in SF3B1, we utilized a spliceosome inhibitor that targets SF3B1, Meayamycin (MM). Healthy donors' BM cells were cultured with EPO and MM for 7 days. Vehicle treated cells served as controls. Erythroblast cultures treated with MM showed numerous RS, absent in controls. Furthermore, cultures with MM displayed marked dyserythropoiesis. We concluded that MM induces RS by blocking SF3B1 and produces a phenotype similar to patients with SF3B1 mutations. Quantification of the levels of unspliced and spliced U2-dependent introns using real-time PCR-based spliceosome assays are ongoing but preliminary results suggest differences in the rate of splicing between mutant and WT patients. In conclusion, our findings suggest that defects in the spliceosome machinery contribute to pathogenesis of MDS. In particular, SF3B1, a member of the spliceosome complex, is a novel tumor suppressor gene frequently mutated in patients with RS. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2911-2911
Author(s):  
Kamaleldin E Elagib ◽  
Chih-Huan Lu ◽  
Ewelina Zasadzińska ◽  
Daniel R Foltz ◽  
Peter Balogh ◽  
...  

Abstract Infantile (fetal/neonatal) megakaryocytes (Mk) differ from "adult" Mk in several important aspects. They are smaller, more proliferative, less polyploid and show leaky expression of erythroid genes. Their distinctive properties contribute to multiple disease states including neonatal thrombocytopenia, poor platelet recovery post umbilical cord blood (CB) transplantation, and acute Mk leukemias (AMkL). Their leukemic propensity is highlighted by the capacity of the AMkL oncoprotein GATA1s to transform infantile but not adult Mk. Despite their altered morphogenesis, infantile megakaryocytes retain most features of the adult differentiation and signaling program. Their principal signaling perturbation has been characterized as excessive responsiveness to thrombopoietin (Tpo) particularly with regard to the mTOR pathway. Thus Tpo agonists used to treat adult thrombocytopenia may not offer appropriate therapy for neonatal thrombocytopenia. We previously identified a signaling pathway that drives megakaryocyte morphogenesis and is disrupted by the GATA1s oncoprotein in AMkL (Elagib et al., Dev. Cell, 2013). A central feature of this pathway is the irreversible activation of the P-TEFb kinase (Cdk9/Cyclin T). This cascade is initiated by downregulation of core components of the repressive 7SK snRNP complex (MePCE, LARP7, 7SK snRNA). The resulting constitutive P-TEFb activation drives multiple features of Mk differentiation: induction of cytoskeletal morpho-genetic factors, silencing of erythroid genes, and promotion of histone H2B K120 monoubiquitiniation (H2BUb1). A critical, rate-limiting step triggering this pathway comprises MePCE proteolysis by calpain 2. GATA1s disrupts this pathway by preventing induction of calpain 2 by wild type GATA1. We now report that infantile Mk display intrinsic defects in the Mk P-TEFb activation pathway. In repeated experiments, human CB Mk failed to upregulate P-TEFb-dependent cytoskeletal factors, exhibited global deficiency in H2BUb1, and incompletely silenced erythroid antigen expression. Their defective P-TEFb activation resulted from an inability to downregulate 7SK snRNA, despite downregulation of the key 7SK stabilizers MePCE and LARP7. The inexplicable stabilization of 7SK in CB Mk argues for the existence of an alternative, infantile 7SK snRNP complex refractory to activation by calpain. Accordingly, screening studies identified candidate 7SK binding factors preferentially expressed in CB as opposed to adult progenitors. Among these factors, the RNA binding protein IGF2BP3 showed high abundance in CB Mk but complete absence from adult peripheral blood-derived (PB) Mk. Furthermore, immunoprecipitation studies consistently showed interaction of endogenous IGF2BP3 and HEXIM1 in K562 cells. HEXIM1 is the 7SK snRNP component that mediates repression of P-TEFb. Immunoprecipitation of epitope-tagged IGF2BP3 from HEK293 cells consistently identified an association with endogenous 7SK snRNA. In addition, enforced expression of IGF2BP3 in HEK293 cells, to levels seen in CB Mk, shifted the fractionation pattern of HEXIM1 on glycerol gradient sedimentation. Notably, a similar difference in HEXIM1 fractionation was seen when comparing CB and adult Mk by glycerol gradient sedimentation. Thus, IGF2BP3 represents a fetal/neonatal factor that reconfigures the composition and/or conformation of the 7SK snRNP, potentially altering regulation of P-TEFb. Contribution of IGF2BP3 to the infantile Mk phenotype was supported by experiments in which shRNA-mediated knockdown in CB Mk consistently enhanced enlargement, polyploidization, growth arrest, and erythroid silencing. Conversely, enforced expression of IGF2BP3 in adult Mk inhibited their enlargement, polyploidization, and growth arrest. Our results thus implicate IGF2BP3 as a key contributor to the infantile Mk phenotype, interfering with morphogenesis by stabilizing 7SK and thwarting irreversible P-TEFb activation. In light of our prior published results on the inhibitory effects of GATA1s in Mk morphogenesis (Elagib et al., Dev. Cell, 2013), our current findings highlight P-TEFb regulation as a convergence point for oncogenic stimuli during megakaryopoiesis. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 130-130
Author(s):  
Eddy HJ van Roon ◽  
Ronald Stam ◽  
Merel Willekes ◽  
Roland P. Kuiper ◽  
Rutger W.W. Brouwer ◽  
...  

Abstract Background and purpose: Acute lymphoblastic leukemia (ALL) in infants (<1 year of age) carrying MLL translocations represent a highly aggressive subtype of leukemia with an exceedingly poor prognosis. Although MLL rearrangements have been considered to be causative for this type of leukemia, recent studies using mouse models suggest that the expression of chimeric MLL fusion genes alone may not be sufficient to induce leukemia. In an attempt to identify additional mutations that synergize with MLLfusion genes in driving leukemogenesis, we performed exome-sequencing in primary infant ALL patient samples (n=13) carrying the t(4;11)-translocation, giving rise to the MLL-AF4 fusion protein. Materials and Methods: Bone marrow or peripheral blood samples from untreated infants diagnosed with t(4;11)+ ALL were collected at our laboratory as part of the international collaborative INTERFANT treatment protocols. Genomic DNA was extracted from ~5×106leukemic cells. Exome capture has been performed using the Sure Select Human All Exon v2 kit (Agilent Technologies). The samples were prepped for sequencing according to the TruSeq v3 protocol (Illumina) prior to sequencing on the Hiseq2000 with a v3 flowcell for 100bp = 7bp index (Illumina). The reads have been aligned against the human reference genome build 19 (hg19) using a BWA and NARWHAL based pipeline. Sequential filtering selected for single nucleotide variations (SNVs) in canonical splice sites, insertions/deletions (indels) providing frameshifts, indels involving regions conserved between species (phyloP >3), nonsense mutations and nonsynonymous missense mutations involving regions conserved between species. All known annotated single nucleotide polymorphisms according to dbSNP build 138 and an in-house database of 1354 sequenced exomes were excluded. Recurrent mutations were validated by Sanger sequencing on an extended infant ALL patient cohort (n=122). Results: The sequenced exomes had an average coverage depth of 102.5 reads per region or 109 per base. Exome sequencing revealed that, after filtering, t(4;11)+ infant ALL patients on average carried 71 variants (range 58-98, including rare germline variants and somatic mutations). To identify recurrent mutations we selected 23 genes in which two or more patients carried a mutation for further validation by Sanger sequencing. Although all 23 genes were successfully validated, no additional patients carrying these mutations were found. However, recurrent secondary mutations within the same functional region as the initial mutations were found in two genes: PXDN and DSP. The average incidences of the PXDN and DSP mutations amongst MLL-rearranged infant ALL patients were 32% and 20%, respectively. Interestingly, MLL-rearranged infant ALL patients carrying a PXDN mutation had a significantly (p=0.013) better prognosis compared with patients not carrying a PXDNmutation, with mean EFS rates of 6.4 vs. 3.2 years, respectively. Both gene products are involved in intracellular interactions, desmosome formation, extracellular matrix consolidation and phagocytosis and no prior associations with ALL have been made before. Conclusion: We identified 23 genes with two or more mutations in our initial exome sequencing cohort. None of these mutations were found in additional patients in our validation cohort. Despite an average of 71 variations per patient, no recurrent mutations were identified and our study provides evidence supporting previous observations that infant ALL has one of the lowest mutation rates observed in human cancer. However, we identified additional mutations within the same functional region as the initial mutation in DSP and PXDN. Interestingly, PXDN mutations show an association with a favorable prognosis in MLL-R patients. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 645-645
Author(s):  
Zhuobin Liang ◽  
Yaqun Teng ◽  
Jingchun Liu ◽  
Simonne Longerich ◽  
Xiaoyong Chen ◽  
...  

Abstract Fanconi anemia (FA) is characterized by developmental abnormalities, bone marrow failure, and a strong cancer predisposition. FA cells are hypersensitive to DNA replicative stress, and accumulate co-transcriptional R-loops. Previous work has demonstrated that BRCA2 binds to R loops, and increased R loops are noted in FA-D2 mutant cells. Additionally, it is understood that at least one FA protein, FANCA, binds RNA. The goal of this study was to understand the relationship between FANCD2 and RNA, especially with regard to manifestation of R loops as a part of the pathophysiology of FA. First, we confirmed the increased presence of R loops in FA mutant cells using the S9.6 monoclonal antibody immunofluorescence microscopy. RNAseH overexpression removes R loop signal and increases cell survival upon mitomycin C treatment. We also showed the presence of increased R loops in an actively transcribed region of the actin gene by bisulfite DNA sequencing. We used the Damage At RNA Transcription (DART) assay, which is designed to combine oxidative DNA damage and the genomic insertion of a hyper transcription site (Fig A). Coactivation of transcription and DNA damage results in colocalization of FANCD2 and S9.6/R loop signal at the transcriptional site (Fig B and C). Consistent with the S9.6 IF, wild type RNAseH overexpression resulted in the abrogation of FANCD2 colocalization. We then asked if FANCD2 binds RNA. FANCD2 in cell lysate bound to biotinylated RNA species, preferring GC rich RNAs. Using recombinant FANCI-FANCD2 (ID2) protein (Fig D), we found that ID2 binds preferably to single stranded RNA in a more robust manner than DNA (Fig E and F). Interestingly, an ID2 complex with a known DNA binding mutation in FANCI also was defective for RNA binding. Furthermore, ID2 bound to R loops but was mediated via the single stranded DNA component of the structure. Importantly, an in vitro monoubiquitination reconstitution system using FANCL as the E3 ligase demonstrated that monoubiquitination of ID2 was stimulated to an equal or greater degree by RNA versus DNA, with greater signal in presence of GC-rich, single-stranded RNA as well as R loops (Fig G and H and data not shown). Collectively, our results support a novel mechanism the ID2 complex suppresses the formation of pathogenic R-loops by binding RNA species, thereby activating the FA pathway (Fig I). Figure. Figure. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 917-917
Author(s):  
Daichi Inoue ◽  
Stanley C Lee ◽  
Akihide Yoshimi ◽  
Justin Taylor ◽  
Lillian E Bitner ◽  
...  

Inversions between chromosome 3q21 and 3q26 ("inv(3)/t(3;3)") mark an aggressive, poor prognosis form of AML with limited treatment options and also occasionally occur in MDS and CML. Inv(3)/t(3;3) repositions a distal GATA2enhancer from 3q21 to the EVI1locus at 3q26 inducing ectopic EVI1 expression and reducing GATA2 expression. At the same time, additional genomic alterations exist in inv(3)/t(3;3) leukemias but the contributions of these events to inv(3)/t(3;3) leukemia are not well understood. Here we evaluated genomic alterations in 63 patients with inv(3)/t(3;3) leukemia. Mutations in NRAS and the core RNA splicing factor SF3B1 were the most common individual alterations (each occurring in 27% of patients; Fig.A). We next quantified gene expression and splicing in AML samples with and without inv(3)/t(3;3) abnormalities and SF3B1 mutations. Amongst inv(3)/t(3;3) AML, the majority of gene expression changes were driven by the inv(3)/t(3;3) rearrangement while the majority of splicing changes were driven by mutant SF3B1. The most abundant category of splicing change in inv(3)/SF3B1 co-mutant cells was alteration in 3' splice site usage (Fig.B). Intriguingly, one of the most robust changes in splicing in inv3/SF3B1 co-mutant cells was aberrant splicing of EVI1itself such that SF3B1 mutant cells promoted expression of a novel isoform of EVI1using an intron proximal 3' splice site (Fig.C; the official gene name "MECOM" corresponds to the genes MDS1and EVI1at this locus). While several mRNA isoforms of EVI1have been previously described, these all result in loss of EVI1 functional domains. In contrast, the unique EVI1 isoform in SF3B1 mutant/inv(3) AMLs contains in an in-frame insertion of six amino acids in the second zinc finger domain (ZF2) of EVI1 (a region of EVI1 known to be affected by germline mutations in leukemia predisposition syndromes). These data identify that nearly one-third of inv(3) AML patients express a heretofore undescribed isoform of EVI1. Of note, this unannotated EVI1isoform is also present in EVI1expressing/SF3B1K700Emutant leukemias lacking inv(3)). The above findings highlight a novel model where inv(3)/t(3;3) AML is driven by ectopic expression of distinct oncogenic isoforms of EVI1. To test this model and understand the contribution of SF3B1K700Eto inv(3)/t(3;3) AML, we crossed transgenic mice bearing the entire human inv(3)(q21;q26) locus whereby the GATA2enhancer misdirects human EVI1expression ("inv3 mice"; Yamazaki et al. Cancer Cell2014) to Sf3b1K700Econditional knockin mice (Mx1-cre Sf3b1K700E/WT).Given that the human MECOMlocus (coding and noncoding regions) was recapitulated in this mouse model, the concordant novel EVI1isoform was expressed in inv3/Sf3b1K700Emice as in patients. While Mx1-cre Sf3b1K700Emice develop an MDS-like disorder, inv3 mice develop myeloid and lymphoid leukemias with lethality ~300 days after birth. However, expression of the Sf3b1K700E/WTmutation in inv3 hematopoietic cells resulted in a highly penetrant MDS, which transformed to a lethal AML by a median of 241 days (Fig.D;p=0.0021). In the first 6 months following transplant, Mx1-cre inv3 Sf3b1K700Emice had leukopenia, macrocytic anemia, and morphologic dysplasia (Fig.E-F) that eventually transformed to a disease with a high WBC count and large numbers of immature cells around time of death. In competitive reconstitution assays, Mx1-cre inv3 Sf3b1K700Ehematopoietic stem cells (HSCs) failed to differentiate into mature peripheral blood cells despite having a competitive advantage at the level of HSCs (Fig.G-H). RNA-seq of hematopoietic precursors from the above models identified (i) a substantial change in splicing in inv3/Sf3b1K700Emutant leukemias versus those driven by inv3 alone, and (ii) alterations in a host of RNA binding proteins in inv3/Sf3b1K700Emutant leukemias (Fig.I). These data highlight a high occurrence of SF3B1 mutations in inv(3)/t(3;3) leukemias, present a new genetically accurate model for inv(3) AML, and uncover a novel oncogenic isoform of EVI1 expressed in a large proportion of inv(3)/t(3;3) patients. Ongoing work focused on identifying the mechanistic effect of the SF3B1-mutant induced aberrant EVI1isoform may provide novel insight into the role of EVI1 in promoting leukemogenesis and engender development of therapeutic opportunities targeting EVI1splicing. Figure Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. 931-931
Author(s):  
Marilyn Parra ◽  
Benjamin W Booth ◽  
Gene W Yeo ◽  
Richard Weiszmann ◽  
Susan E Celniker ◽  
...  

Abstract Proper expression of the MDS-disease gene, SF3B1, ensures appropriate pre-mRNA splicing in erythroid progenitors and during terminal erythropoiesis. We previously showed that the SF3B1 gene is post-transcriptionally regulated in a differentiation stage-specific manner by intron retention (IR), such that ~50% of its transcripts in mature erythroblasts retain intron 4. Based on new mechanistic studies, we propose a model in which mostly unannotated and noncoding exons within intron 4 function as splicing decoys; i.e., they promote retention of intron 4 by interacting with, and blocking splice sites of, the adjacent exons 4 and 5. A total of six putative decoy exons were revealed via RT-PCR and RNA-seq analysis of RNA from erythroblasts treated with inhibitors of nonsense-mediated decay. That decoy exons have IR-promoting activity is suggested by several criteria. First, the frequency of interaction between constitutive exons 4 and 5 and putative decoy exons within intron 4, measured by the abundance of splice junctions in RNA-seq read data, is temporally correlated with levels of intron 4 retention during terminal erythropoiesis. Both IR and decoy splice junctions were low in early stage erythroblasts and much higher in mature erythroblasts. Second, selected decoy exons exhibited IR-promoting activity in the context of minigene splicing reporters expressing the exon 3-6 region of SF3B1 in transfected K562 cells. The wild type minigene reproduced the intron-specific retention phenotype, since it was fully spliced at introns 3 and 5 but exhibited substantial retention of intron 4, whereas deletion of decoy exon 4e, or mutation of its splice sites, substantially decreased IR. Third, RBP (RNA binding protein) cross-linking data from K562 cells show that 3' splice site factors including U2AF1 and U2AF2 can bind specifically to 3' splice sites of intron 4's decoy exons. Finally, several experiments showed that IR-promoting activity of decoy exons is a more general phenomenon that likely governs IR in other erythroid genes. We observed not only that SF3B1 intron 4 decoy exons could promote IR in heterologous contexts, but also that predicted decoy exons from other erythroblast transcripts could promote IR in the SF3B1 minigene. Apart from this experimental data, comparative genomics revealed that the SF3B1 decoy exons are extremely conserved among vertebrate genomes, with two of the exons being essentially identical from fish to humans. Together this data supports the hypothesis that a subset of up-regulated IR events in late erythroblasts are controlled by decoy exons that block productive splicing at the flanking exons. We propose that regulated IR is an important post-transcriptional mechanism for adjusting cellular splicing capacity during terminal erythropoiesis by regulating expression of key splicing factors such as SF3B1. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Author(s):  
Laramie D. Lemon ◽  
Sneha Kanna ◽  
Kim Wai Mo ◽  
Miranda Adams ◽  
Haley Choi ◽  
...  

Somatic missense mutations in histone genes turn these essential proteins into oncohistones, which can drive oncogenesis. Understanding how missense mutations alter histone function is challenging in mammals as the changes occur in a single histone gene. For example, described oncohistone mutations predominantly occur in the histone H3.3 gene, despite the human genome encoding 15 H3 genes. To understand how oncogenic histone missense mutations alter histone function, we leverage the budding yeast model, which encodes only two H3 genes, to explore the functional consequences of oncohistones H3K36M, H3G34W, H3G34L, H3G34R, and H3G34V. An analysis of cells that express each of these variants as the sole copy of H3 reveals that H3K36-mutants show different drug sensitivities compared to H3G34 mutants. This finding suggests that changes to proximal amino acids in the H3 N-terminal tail alter distinct biological pathways. We exploited the caffeine sensitive growth of H3K36 mutant cells to perform a high copy suppressor screen. This screen identified genes linked to histone function and transcriptional regulation, the histone H4/H2A acetyltransferase, Esa1, a forkhead-associated domain-containing gene expression regulator, Tos4, an m6A RNA binding protein, Pho92, and a cyclin-dependent kinase, Sgv1/Bur1. We show that the Esa1 lysine acetyltransferase activity is critical for suppression of the caffeine sensitive growth of H3K36R mutant cells while neither of the characterized binding interactions of Tos4 nor Pho92 are required for suppression. Finally, Sgv1/Bur1-mediated suppression may occur through a dominant negative mechanism. This screen identifies pathways that could be altered by oncohistone mutations and highlights the value of yeast genetics to identify pathways altered by such mutations.


Genetics ◽  
2001 ◽  
Vol 159 (4) ◽  
pp. 1617-1630
Author(s):  
Suk-Won Jin ◽  
Nancy Arno ◽  
Adam Cohen ◽  
Amy Shah ◽  
Qijin Xu ◽  
...  

Abstract FOG-1 controls germ cell fates in the nematode Caenorhabditis elegans. Sequence analyses revealed that FOG-1 is a cytoplasmic polyadenylation element binding (CPEB) protein; similar proteins from other species have been shown to bind messenger RNAs and regulate their translation. Our analyses of fog-1 mutations indicate that each of the three RNA-binding domains of FOG-1 is essential for activity. In addition, biochemical tests show that FOG-1 is capable of binding RNA sequences in the 3′-untranslated region of its own message. Finally, genetic assays reveal that fog-1 functions zygotically, that the small fog-1 transcript has no detectable function, and that missense mutations in fog-1 cause a dominant negative phenotype. This last observation suggests that FOG-1 acts in a complex, or as a multimer, to regulate translation. On the basis of these data, we propose that FOG-1 binds RNA to regulate germ cell fates and that it does so by controlling the translation of its targets. One of these targets might be the fog-1 transcript itself.


2021 ◽  
Author(s):  
Saumya Agrawal ◽  
Tanvir Alam ◽  
Masaru Koido ◽  
Ivan V. Kulakovskiy ◽  
Jessica Severin ◽  
...  

AbstractTranscription of the human genome yields mostly long non-coding RNAs (lncRNAs). Systematic functional annotation of lncRNAs is challenging due to their low expression level, cell type-specific occurrence, poor sequence conservation between orthologs, and lack of information about RNA domains. Currently, 95% of human lncRNAs have no functional characterization. Using chromatin conformation and Cap Analysis of Gene Expression (CAGE) data in 18 human cell types, we systematically located genomic regions in spatial proximity to lncRNA genes and identified functional clusters of interacting protein-coding genes, lncRNAs and enhancers. Using these clusters we provide a cell type-specific functional annotation for 7,651 out of 14,198 (53.88%) lncRNAs. LncRNAs tend to have specialized roles in the cell type in which it is first expressed, and to incorporate more general functions as its expression is acquired by multiple cell types during evolution. By analyzing RNA-binding protein and RNA-chromatin interaction data in the context of the spatial genomic interaction map, we explored mechanisms by which these lncRNAs can act.


Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. 79-79
Author(s):  
Andrew Crispin ◽  
Paul Schmidt ◽  
Dean Campagna ◽  
Chang Cao ◽  
Daniel Lichtenstein ◽  
...  

Abstract Congenital sideroblastic anemias (CSAs) are uncommon inherited diseases resulting from defects in heme biosynthesis, mitochondrial translation or mitochondrial iron-sulfur cluster (ISC) assembly. CSAs are characterized by pathological mitochondrial iron deposits in bone marrow erythroblasts. Recently, mutations in mitochondrialheat shock protein 70 (HSPA9), a critical chaperone involved in mitochondrial ISC assembly, have been reported as a cause of non-syndromic CSA. Human heat shock cognate protein 20 (HSCB), a highly conserved mitochondrial co-chaperone, is the primary binding partner of HSPA9. HSCB allows the transfer of nascent ISC to HSPA9 and stimulates its ATPase activity, promoting ISC transfer to target proteins. To identify novel genes responsible for CSA, we performed whole exome sequencing on more than 75 CSA probands and their family members. In one patient, a young woman, with pancytopenia characterized by a normocytic anemia with numerous bone marrow ringed sideroblasts, we identified two variants in HSCB : a paternally-inherited promoter variant (c.-134C&gt;A) predicted to disrupt a conserved ETS transcription factor binding site, and a maternally-inherited frameshift (c.259dup, p.T87fs*27). A fibroblast cell-line derived from the proband showed a decrease in HSCB expression, but normal HSPA9 expression compared to healthy, unrelated controls. Impairment of ETS1-dependent transcriptional activation of the promoter variant was demonstrated in K562 cells transfected with an HSCB-luciferase reporter construct. K562 cells were also employed to determine if reduced expression of HSCB could result in impaired erythroid metabolism, maturation, or proliferation. K562 cells infected with shRNA directed against HSCB were deficient in multiple mitochondrial respiratory complexes, had abnormal iron metabolism and a defect of protein lipoylation, all consistent with defective ISC metabolism. In addition, both IRP1 and IRP2 expression were decreased and cell surface transferrin receptor 1 (TFR1) expression was enhanced, suggesting disturbed cellular iron metabolism. Nevertheless, cells lacking HSCB partially retained an ability to respond to iron chelation and iron overload. Cells lacking HSCB lose their ability to hemoglobinize in response to sodium butyrate treatment (Figure 1A). This defect was confirmed in vivo using a morpholino strategy in zebrafish, as fish lacking HSCB are also unable to hemoglobize (Fig 1B). We generated an Hscb conditional mouse to better elucidate the underlying pathophysiology of the disease. Heterozygous (Hscb+/-) animals have no discernable phenotype; however, null animals die prior to embryonic day E7.5. Thus, to avoid this lethality, we employed Vav-cre animals (Tg(Vav1-cre)1Graf) to evaluate the loss of HSCB specifically in the hematopoietic compartment. Hscbc/- Vav-cre+ pups are pale and growth retarded compared to control littermates and die at approximately p10 with severe pancytopenia. To assess the loss of HSCB specifically in the erythroid lineage, we bred conditional animals to EpoR-cre (Eportm1(EGFP/cre)Uk) mice. Hscbc/- EpoR-cre+ mice die at approximately E12.5 due to a complete failure of erythropoiesis (Figure 1C). Finally, temporally inducible, hematopoietic-specific deletion animals were generated by transplantation of fetal livers from Mx-Cre (Tg(Mx1-cre)1Cgn) positive Hscbc/- animals. After polyinosinic:polycytidylic acid (pIpC) induction, global defects of hematopoiesis were observed in Mx-Cre+ animals, leading to their death 3-weeks post-induction from profound pancytopenia. A transient siderocytosis was seen in the peripheral blood between days 6-8 post-pIpC. Flow cytometry using FSC-TER119-CD44 gating strategy confirmed the defect in erythropoiesis. Taken together, these data demonstrate that HSCB is essential for hematopoiesis; both whole animal and in vitro cell culture models recapitulate the patient's phenotype, suggesting that the two patient mutations are likely disease-causing. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document