scholarly journals Mutations in the Spliceosome Lead to Alternative Splicing of CSF3R in Promoting Myelodysplastic Syndromes

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1978-1978
Author(s):  
Frances Austin ◽  
Hrishikesh M Mehta ◽  
Chonghui Cheng ◽  
Seth Corey

Abstract Introduction: Despite advances into the genetic basis of myelodysplastic syndromes (MDS), the mechanisms of their pathophysiology remain poorly understood, and frequently terminates in acute myeloid leukemia. Mutually exclusive mutations have been identified in splicing factors U2AF1, SF3B1, SRSF2, SF3A1, ZRSR2 and LUcL7 in 85% of MDS cases. We and others have identified aberrant forms of Granulocyte Colony Stimulating Factor Receptor (CSF3R) in patients with myeloid malignancies and partially characterized their phenotypic and signal transduction effects. Two of the seven isoforms of CSF3R have clinical relevance: Class I (full-length form) and Class IV (alternatively spliced form). Compared to Class I, Class IV isoform is differentiation defective and displays enhanced proliferation. Increased expression of the Class IV isoform has been found in MDS/AML and results in enhanced cell proliferation and impaired differentiation of granulocytes (Mehta et al, Leukemia 2014). The Class IV CSF3R transcript is derived by intron excision, whereas Class I results from retention of that intron. We hypothesize aberrant splicing of the CSF3R transcript promoted by mutations in splicing factors drives abnormal granulopoiesis in MDS due to increased expression of the differentiation defective Class IV CSF3R. Methods: We identified U2AF1, SRSF2, and SAM68 sites within the nucleotide sequence of the spliced intron. To study the effect of splicing factor mutations on CSF3R splicing, we constructed a minigene consisting of 5' exon of the CAT gene with an ATG site and a partial intron fused to intron region of G-CSFR upstream of Exon 17. The retained intron is present in the exon 17 of CSF3R. The minigene consists of 2 introns and 3 exons. We transduced 293FT cells with the minigene ± wild-type U2AF1 or U2AF1 S34F mutant, as well as the minigene ± wild-type SF3B1 or SF3B1 K300E. The cells were then treated with either 1µM sodium orthovanadate (Na3VO4) or 10 nM phorbol myristic acetate (PMA). The former inhibits tyrosine phosphatases, promoting tyrosine protein phosphorylation, whereas the latter promotes activation of the protein kinase C pathway, which eventually activates ERK1/2 an upstream activator of SAM68. Protein phosphorylation of spliceosome proteins is fundamental to regulation of splicing. The effect of PMA and Na3VO4 were compared with each other and those treated with control diluent. Qualitative and semi-quantitative evaluations were performed. Results: We identified expression with SF3B1 K700E enhances CSF3R splicing resulting in increased Class IV, however the U2AF1 S34F inhibits splicing resulting in reduced Class IV. The increase and decrease were in relation to the wild type. Further exploration of how cellular protein phosphorylation influences splicing using Na3VO4 (identified that cells transduced with SF3B1, U2AF1 wt or mutant shown decreased splicing resulting in Class IV transcript. PMA however had no effect on either class expression. Conclusions: These data suggest that CSF3R transcript processing is sensitive to diverse tyrosine kinase signaling, resulting in increased intron retention, suggesting that phosphorylation events inhibit Class IV transcript formation. Additionally, the mutations in splicing factors which are recurrently found in MDS are associated with altered CSF3Rtranscripts and may affect CSF3R signaling and cell phenotype. This work is supported by DOD Bone Marrow Failure Idea Award and NIH R01HL128173. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3897-3897
Author(s):  
Frances Austin ◽  
Hrishikesh M Mehta ◽  
Seth J. Corey ◽  
Chonghui Cheng

Abstract Despite advances into the genetic basis of myelodysplastic syndromes (MDS), the mechanisms of their pathophysiology remain poorly understood, and frequently terminates in acute myeloid leukemia. Mutually exclusive mutations have been identified in splicing factors U2AF1, SF3B1, SRSF2, SF3A1, ZRSR2 and LUcL7 in 85% of MDS cases. We and others have identified aberrant forms of Granulocyte Colony Stimulating Factor Receptor (CSF3R) in patients with myeloid malignancies and partially characterized their phenotypic and signal transduction effects. Two of the seven isoforms of CSF3R have clinical relevance: Class I (full-length form) and Class IV (alternatively spliced form). Compared to Class I, Class IV isoform is differentiation defective and displays enhanced proliferation. Increased expression of the Class IV isoform has been found in MDS/AML and results in enhanced cell proliferation and impaired differentiation of granulocytes (Mehta et al, Leukemia 2014). The Class IV CSF3R transcript is derived by intron excision, whereas Class I results from retention of that intron. We hypothesize aberrant splicing of the CSF3R transcript promoted by mutations in splicing factors drives abnormal granulopoiesis in MDS due to increased expression of the differentiation defective Class IV CSF3R. We identified U2AF1, SRSF2, and SAM68 sites within the nucleotide sequence of the spliced intron. To study the effect of splicing factor mutations on CSF3R splicing, we constructed a minigene consisting of 5' exon of the CAT gene with an ATG site and a partial intron fused to intron region of G-CSFR upstream of Exon 17. The retained intron is present in the exon 17 of CSF3R. The minigene consists of 2 introns and 3 exons. We transduced 293FT cells with the minigene ± wild-type U2AF1 or U2AF1 S34F mutant, as well as the minigene ± wild-type SF3B1 or SF3B1 K300E. The cells were then treated with either 1µM sodium orthovanadate (Na3VO4) or 10 nM phorbol myristic acetate (PMA). The former inhibits tyrosine phosphatases, promoting tyrosine protein phosphorylation, whereas the latter promotes activation of the protein kinase C pathway, which eventually activates ERK1/2 an upstream activator of SAM68. Protein phosphorylation of spliceosome proteins is fundamental to regulation of splicing. The effect of PMA and Na3VO4 were compared with each other and those treated with control diluent. Qualitative and semi-quantitative evaluations were performed. We identified expression with SF3B1 K700E enhances CSF3R splicing resulting in increased Class IV, however the U2AF1 S34F inhibits splicing resulting in reduced Class IV. The increase and decrease were in relation to the wild type. Further exploration of how cellular protein phosphorylation influences splicing using Na3VO4 (identified that cells transduced with SF3B1, U2AF1 wt or mutant shown decreased splicing resulting in Class IV transcript. PMA however had no effect on either class expression. These data suggest that CSF3R transcript processing is sensitive to diverse tyrosine kinase signaling, resulting in increased intron retention, suggesting that phosphorylation events inhibit Class IV transcript formation. Additionally the mutations in splicing factors which are recurrently found in MDS are associated with altered CSF3R transcripts and may affect CSF3R signaling and cell phenotype. (This work is supported by DOD and NIH R01R01HL128173.) Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1224-1224
Author(s):  
Junke Zheng ◽  
Chengcheng Zhang

Abstract Abstract 1224 How stem cells interact with the microenvironment to regulate their cell fates and metabolism is largely unknown. Here we show that, in a hematopoietic stem cell (HSC) -specific inducible knockout model, the cytoskeleton-modulating protein profilin 1 (pfn1) is essential for the maintenance of multiple cell fates and metabolism of HSCs. The deletion of pfn1 in HSCs led to bone marrow failure, loss of quiescence, increased apoptosis, and mobilization of HSCs in vivo. In reconstitution analyses, pfn1-deficient cells were selectively lost from mixed bone marrow chimeras. By contrast, pfn1 deletion did not significantly affect differentiation or homing of HSCs. When compared to wild-type cells, levels of expression of Hif-1a, EGR1, and MLL were lower and an earlier switch from glycolysis to mitochondrial respiration with increased ROS level was observed in pfn1-deficient HSCs. This switch preceded the detectable alteration of other cell fates. Importantly, treatment of pfn1-deficient mice with the antioxidant N-acetyl-l-cysteine reversed the ROS level and loss of quiescence of HSCs, suggesting that pfn1 maintained metabolism is required for the quiescence of HSCs. Furthermore, we demonstrated that expression of wild-type pfn1 but not the actin-binding deficient or poly-proline binding-deficient mutants of pfn1 rescued the defective phenotype of pfn1-deficient HSCs. This result indicates that actin-binding and proline-binding activities of pfn1 are required for its function in HSCs. Thus, pfn1 plays an essential role in regulating the retention and metabolism of HSCs in the bone marrow microenvironment. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3153-3153
Author(s):  
Virginie Chesnais ◽  
Marie-laure Arcangeli ◽  
Caroline Delette ◽  
Alice Rousseau ◽  
M'boyba Khadija Diop ◽  
...  

Abstract Introduction Myelodysplastic syndromes (MDS) are clinically diverse malignant disorders of aging with a propensity to evolve to acute myeloid leukemia (AML) or bone marrow failure. In early MDS, whole genome sequencing identified mutations distributed in few clones. Evidence have been provided for the existence of a MDS-initiating cell in cases harboring a 5q deletion. However, the clonal heterogeneity and its consequences on the phenotypic diversity of non-del(5q) MDS is little documented. Here we focused on studying the hierarchical organization and the functionality of clones defined by molecular profiling. The clonal architecture of the CD34+CD38- hematopoietic stem/progenitor cell (HSPC) compartment was investigated and dominant clones were examined as MDS-initiating cells. Material and methods Bone marrow (BM) samples were obtained from 20 patients with non-del(5q) MDS enrolled in the national Programme Hospitalier de Recherche Clinique MDS-04 after informed consent in accordance with ethics committee guidelines. BM samples, cytaphereses from age-matched healthy individuals and cord bloods were used as controls. Targeted NGS of a selected panel of 39 genes was used to define the mutational landscape on BM mononuclear cells (MNC). To study the clonal architecture at the HSPC level, single CD34+CD38- cells were seeded in 96-well plates coated with MS-5 stromal cells and cultured in H5100 MyeloCult medium (StemCell Technologies, Vancouver, Canada) with cytokines for six weeks. For long-term culture-initiating cell (LTC-IC) assays, CD34+ progenitors were cultured for six weeks on MS-5-coated plates without cytokines and then tested for colony-forming cells. For clonogenic assays, CD34+CD38- cells were seeded in methylcellulose for two weeks. All animal experimentations were performed in NSG mice. Results In the 20 cases of non-del(5q) MDS, genomic lesions were traced down to single CD34+CD38- HSPC-derived colonies. Clonal organization was mostly linear in 13/17 patients and branched in 4 cases with retention of a dominant subclone. The clone detected in LTC-IC compartment and that reconstituted short-term human hematopoiesis in xenotransplantation models was usually the dominant clone, which gave rise to the myeloid and to a lesser extent to the lymphoid lineage. Other mutations not detected in LTC-IC can appear in CD34+CD38- compartment or at the level of lineage-committed progenitors. The pattern of mutations may differ between common myeloid (CMP), granulo-monocytic (GMP) and megakaryocytic-erythroid (MEP) progenitors. For instance, a major truncating BCOR gene mutation affecting HSPC and CMP was beneath the threshold of detection in GMP or MEP. Consistently, BCOR knockdown by shRNA in normal CD34+ progenitors impaired their granulocytic and erythroid differentiation. By contrast, a STAG2 gene mutation, not detected in CMP or MEP, amplified in a GMP, which drove the transformation to AML. Conclusion In the present study, we characterized the first genetic hits that initiate disease in a dominant clone of the CD34+CD38- HSPC compartment, which exhibits LTC-IC activity and reconstitutes human short-term hematopoiesis in NSG mice. The genetic heterogeneity in non-del(5q) MDS arises within the HSPC compartment and in lineage-committed progenitors which ultimately support the transformation into AML. The clonal architecture of HSPC compartment and mutations selection along differentiation contribute to the phenotype of MDS. Defining the hierarchy of driver mutations provides insights into the process of transformation, and may guide the search for novel therapeutic strategies. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2367-2367
Author(s):  
Yan Xiu ◽  
Chen Zhao

Abstract We previously demonstrated that the NF-B non-canonical signaling way positively and intrinsically regulates hematopoietic stem/progenitor cell (HSPC) self-renewal and maintains stromal/osteoblastic niches (Stem Cells 2012 30:709-18). These results lead us to think that persistent activation of NF-B non-canonical signaling would have favorable effects on the HSPC pool size and self-renewal capacity. NF-B-inducing kinase (NIK) plays a critical role in non-canonical NF-B signaling by directly phosphorylating IKK. It is constitutively degraded by TRAF3 in unstimulated cells to prevent unwanted NF-B activation. To investigate the enhanced NF-kB non-canonical signaling specifically in hematopoietic cells, we crossed Vav-Cre mice with a mouse strain in which a mutated form of NIK lacking the TRAF3-binding domain is expressed under the control of the ROSA26 promoter after Cre-mediated deletion of the LoxP-flanked STOP cassette (NIKΔT3Cre mice). In contrast to what we expected in these preliminary studies, the NIKΔT3Cre mice rapidly developed anemia, pancytopenia, with a reduced HSPC pool and marrow cellularity and postnatal lethality, mimicking many of the findings in humans with bone marrow failure syndrome, and different from recently published mice with deficiency in A20, which also activates NF-B signaling. Furthermore, the NIK activated HSPCs have profoundly impaired engraftment and self-renewal activity after transplantation into wild-type recipients. Further analysis showed that the mutant cells are proliferate faster and predispose to apoptosis than wild type cells. These observations suggest that finely controlled NF-B activity is crucial for HSC maintenance. Currently, we are focusing on the analysis of the underlying molecular mechanisms. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 14-14
Author(s):  
Larissa Doll ◽  
Narges Aghaallaei ◽  
Julia Skokowa ◽  
Karl Welte ◽  
Baubak Bajoghli

Severe congenital neutropenia (CN) is a bone marrow failure characterized by an impaired neutrophil development. This disease can be caused by different germline mutations, which one of them is Jagunal homolog 1 (JAGN1). CN patients are prone to develop life-threatening infections from birth on and the treatment of choice is recombinant human granulocyte colony-stimulating factor (rhG-CSF). However, patients with JAGN1 deficiency poorly respond to this treatment. Therefore, the development of new therapeutic strategies to treat this disease is essential. Thus far, lack of an animal model was the main limitation to study the function of JAGN1 in neutropenia and perform high-throughput compound screening. In this work, we studied the role of jagn1b in zebrafish granulopoiesis and hematopoiesis. Two different approaches were used. First, we injected antisense morpholino that efficiently blocked the translation of jagn1b mRNA. As a second approach, we used the CRISPR-Cas9 technique to introduce mutations in the zebrafish jagn1b gene and analyzed the crispants. Both of these approaches were tested in wild-type embryos as well as the transgenic mpo:GFP reporter line, in which neutrophils are labeled with GFP. Our results showed that interference with Jagn1b reduced the number of neutrophils, without affecting the early myelopoiesis and monocytes/macrophages. Further analysis revealed that apoptosis was enhanced in embryos injected with the jagn1b morpholino, however, this effect was not restricted to the hematopoietic tissues. Our results also suggested that unfolded protein response (UPR) was activated because the expression levels of genes involved in this pathway such as chop, atf4a and atf4b were significantly upregulated in the jagn1b morphants, when compared with the wild-type counterparts. Overall, we have successfully established a novel in vivo model for studying the role of JAGN1 in granulopoiesis, and the possible role of Jagn1b in UPR response might be useful to find new targets for the development of therapeutic strategies for CN. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 19-19
Author(s):  
Anne Olazabal-Herrero ◽  
Allison M. Green ◽  
Xiaoyong Chen ◽  
Patrick Sung ◽  
Manoj M Pillai ◽  
...  

BACKGROUND. Fanconi Anemia (FA) is a rare disease characterized by congenital abnormalities, bone marrow failure and cancer susceptibility. The pivotal role of the FA pathway is DNA interstrand crosslink (ICL) repair, where FANCD2-FANCI complex monoubiquitination is the key regulatory step. Recent studies have linked defects in the FA pathway to R-loop metabolism and implicated R-loops as an endogenous source of genomic instability that contributes to the FA phenotype, although the mechanism remains elusive. Splicing factors, such as SRSF1 (ASF/SF2), have also been linked to R loops, cancer, and myelodysplastic syndrome (MDS), so we hypothesized a link between SRSF1 and FA. METHODS. Cells: FA-D2 mutant (PD20) and corrected (D2), HeLa. siRNA transfections were used to reduce SRSF1 and FANCD2 protein levels. Survival assays were performed to determine cell sensitivity to DNA damaging agents. In vitro immunoprecipitation was performed using SRSF1 purified from bacteria and FANCD2-FANCI purified from insect cells. R-loops were studied using the Damage At RNA Transcription (DART) assay and S9.6 immunofluorescence. RESULTS. In order to study the involvement of splicing factors in the FA pathway, we knocked down SRSF1 by using siRNA. SRSF1 knockdown lead to impaired FANCD2 monoubiquitination after DNA damage as well as diminished cell survival, thus creating a FA-like phenotype (A). In addition, immunoprecipitation in cells (B) and in vitro showed direct interaction between FANCD2 and SRSF1. We found that double FANCD2+SRSF1 knockdown led to increased R-loops compared to FANCD2/SRSF1 single knockdown visualized by S9.6 immunofluorescence. In addition, DART assay revealed colocalization of SRSF1 and FANCD2 with R-loops in highly transcribed genomic sites, known to be R-loop hotspots (C). Finally, we found that SRSF1 interacts with the mRNA export factor NFX1 in a FANCD2-dependent fashion (D) through RNA, suggesting that they could cooperate in the prevention of R-loop formation by exporting accumulated RNA in the nucleus. CONCLUSION. Our findings suggest a novel non-canonical mechanism of FANCD2 via regulation of SRSF1 in R-loop prevention via facilitation of mRNA export, thus preventing genomic instability (E). CLINICAL APPLICABILITY. These results will reveal a novel mechanism for a non-canonical role of the FA pathway in prevention of genomic instability and will shed light upon the most basic cellular processes of the FA pathway, which is germane for delineating more generalized mechanisms of oncogenesis and genome maintenance. As accumulation of R-loops is associated with FA, we propose that R-loops and R-loop processing proteins (such as SRSF1) offer new avenues as relevant therapeutic targets not only for FA but also for multiple cancers. Disclosures No relevant conflicts of interest to declare.


Microbiology ◽  
2003 ◽  
Vol 149 (10) ◽  
pp. 2901-2908 ◽  
Author(s):  
Youko Sakayori ◽  
Mizuho Muramatsu ◽  
Satoshi Hanada ◽  
Yoichi Kamagata ◽  
Shinichi Kawamoto ◽  
...  

The emergence and spread of mutants resistant to bacteriocins would threaten the safety of using bacteriocins as food preservatives. To determine the physiological characteristics of resistant mutants, mutants of Enterococcus faecium resistant to mundticin KS, a class IIa bacteriocin, were isolated. Two types of mutant were found that had different sensitivities to other antimicrobial agents such as nisin (class I) and kanamycin. Both mutants were resistant to mundticin KS even in the absence of Mg2+ ions. The composition of unsaturated fatty acids in the resistant mutants was significantly increased in the presence of mundticin KS. The composition of the phospholipids in the two resistant mutants also differed from those in the wild-type strain. The putative zwitterionic amino-containing phospholipid in both mutants significantly increased, whereas amounts of phosphatidylglycerol and cardiolipin decreased. These changes in membrane structure may influence resistance of enterococci to class IIa and class I bacteriocins.


1991 ◽  
Vol 174 (6) ◽  
pp. 1629-1637 ◽  
Author(s):  
J H Cox ◽  
J R Bennink ◽  
J W Yewdell

The E3/19K glycoprotein of adenovirus functions to diminish recognition of adenovirus-infected cells by major histocompatibility complex class I-restricted cytotoxic T lymphocytes (CTLs) by binding intracellular class I molecules and preventing them from reaching the plasma membrane. In the present study we have characterized the nature of the interaction between E3/19K and the H-2Kd (Kd) molecule. An E3/19K molecule genetically engineered to terminate six residues from its normal COOH terminus (delta E19), was found to associate with Kd in a manner indistinguishable from wild-type E3/19K. Unlike E3/19K, however, delta E19 was transported through the Golgi complex to the plasma membrane, where it could be detected biochemically and immunocytochemically using a monoclonal antibody specific for the lumenal domain of E3/19K. Importantly, delta E19 also differed from E3/19K in being unable to prevent the presentation of Kd-restricted viral proteins to CTLs. This is unlikely to be due to delta E19 having a lower avidity for Kd than E3/19K, since delta E19 was able to compete with E3/19K for Kd binding, both physically, and functionally in nullifying the E3/19K blockade of antigen presentation. These findings indicate that the ability of E3/19K to block antigen presentation is due solely to its ability to retain newly synthesized class I molecules in the endoplasmic reticulum.


Neurology ◽  
2003 ◽  
Vol 60 (4) ◽  
pp. 538-547 ◽  
Author(s):  
J. Engel ◽  
S. Wiebe ◽  
J. French ◽  
M. Sperling ◽  
P. Williamson ◽  
...  

Objectives/Methods: To examine evidence for effectiveness of anteromesial temporal lobe and localized neocortical resections for disabling complex partial seizures by systematic review and analysis of the literature since 1990.Results: One intention-to-treat Class I randomized, controlled trial of surgery for mesial temporal lobe epilepsy found that 58% of patients randomized to be evaluated for surgical therapy (64% of those who received surgery) were free of disabling seizures and 10 to 15% were unimproved at the end of 1 year, compared with 8% free of disabling seizures in the group randomized to continued medical therapy. There was a significant improvement in quantitative quality-of-life scores and a trend toward better social function at the end of 1 year for patients in the surgical group, no surgical mortality, and infrequent morbidity. Twenty-four Class IV series of temporal lobe resections yielded essentially identical results. There are similar Class IV results for localized neocortical resections; no Class I or II studies are available.Conclusions: A single Class I study and 24 Class IV studies indicate that the benefits of anteromesial temporal lobe resection for disabling complex partial seizures is greater than continued treatment with antiepileptic drugs, and the risks are at least comparable. For patients who are compromised by such seizures, referral to an epilepsy surgery center should be strongly considered. Further studies are needed to determine if neocortical seizures benefit from surgery, and whether early surgical intervention should be the treatment of choice for certain surgically remediable epileptic syndromes.


Sign in / Sign up

Export Citation Format

Share Document