Abstract

DHX15, a member of the DEAD-box RNA helicase family, acts as splicing factor in pre-messenger RNA splicing, pre-ribosomal RNA, and RNA metabolism. Our previous studies showed that DHX15 overexpression is associated with poor prognosis in acute myeloid leukemia (AML), and DHX15 regulates cell apoptosis via the NF-kB signaling pathway(Pan et al., 2017). However, its molecular function remains obscure in the blood system. First, with the Cre-loxp knockout system, Dhx15fl/fl male mice were crossed with vav-icre female mice (specifically expressed in blood system), we found that Dhx15fl/fl; vav-icre homozygous knockout mice (vav-Dhx15-KO) are lethal by E17.5-E18.5. Then, to explore the inner difference in blood system, the hematopoietic stem cells (HSC) were analyzed by flow cytometry in fetal liver (FL) (E13.5, E15.5). Compared with Dhx15fl/fl (wild-type, WT) mice, the lineage-negative cells (Lin- cells; Lin+ Cocktail, CD5, CD45R (B220), CD11b, Anti-Gr-1 (Ly-6G/C), 7-4 and Ter-119) significantly increased in vav-Dhx15-KO FL cells (P<0.0001) and the short-term HSC(ST-HSC) proportion compensatively increased in LSK (Lin-c-Kit+Sca1+cells) population (P<0.01), while the proportion of LK(Lin-c-Kit+Sca1-cells) (P<0.0001 in E13.5, P<0.001 in E15.5), LSK(Lin-c-Kit+Sca1+cells)(P<0.1 in E15.5), and multipotent progenitor (MPP) (P<0.0001 in E15.5) all witnessed decrease in the Lin-/LSK population respectively. Meanwhile, vav-Dhx15-KO mice exhibited impaired multi-lineage hematopoiesis especially in erythroid, where the precursors and immature erythrocytes increased, mature erythrocytes decreased (P<0.001 in E13.5). Next, in vivo competitive repopulation assay were performed to further examine any potential cell-intrinsic defect of HSC function caused by Dhx15-inactivation. Thus, 1,000,000 E13.5/E15.5 total FL donor cells (CD45.2, vav-Dhx15-KO or WT) together with 1,000,000 CD45.1 unfractionated BM competitor cells were intra-venous transplanted into lethally irradiated (9Gy) B6.SJL (CD45.1) recipient mice. Recipient mice were sacrificed 8-weeks after transplantation, flow cytometry was conducted to analysis the ratio of CD45.2 in mice, the results showed that vav-Dhx15-KO FL was severely deficient compared to WT FL(0.1±0.04%vs 62.2±2.8% in BM, 0.2±0.05% vs 64.5±6.1% in SP, 0.1±0.05% vs 45.1±5.5% in PB). These results indicate that Dhx15 is crucial in mice blood system, loss of its function may block cell differentiation, leading to the reduction of mature cells. At the same time, to further explore the molecular mechanism of DHX15 in blood system, MOLM-13 and NB4 leukemia cell lines were selected for shRNA-KD (knockdown) experiments due to the lethality of vav-Dhx15-KO mice. We found that Dhx15-KD significantly impaired the proliferation ability of these cell lines in vitro and in vivo, by experiments of survival competition, clone formation, subcutaneous and intra-venous tumorigenesis. Besides, our data demonstrate DHX15-KD-mediated G0/G1 cell cycle arrest and increase in early-late apoptotic cells by Hoechest 33342-EdU cell cycle and DAPI-Annexin V cell apoptosis assays. Mechanistically, we found that UV_RESPONSE signaling pathway which is mainly related to DNA damage was up-regulated by RNAseq and bioinformatics analysis (DHX15-KD cell line vs WT cell line). Correspondingly, the DNA damage indicators γH2AX and 53BP1 were also increased in DHX15-KD cell lines detected by WB and immunofluorescence experiments, suggesting that the deletion of DHX15 could lead to DNA damage-mediated cell cycle arrest and apoptosis in leukemia cell lines. In summary, these results show that DHX15 deficiency leads to DNA damage-induced cell cycle arrest and apoptosis which further impairs the proliferative capacity of leukemia cell lines and lost function of Dhx15 impairs multi-lineage hematopoiesis in mouse embryos, which may lead to its perinatal death.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call