Abstract

Myelodysplastic syndromes (MDS) are a diverse group of clonal marrow stem cell disorders characterized by ineffective hematopoiesis and dysplasia of the cellular elements. Although low-dose cytarabine (Ara-c) has been considered as one of the frontline therapies for high-risk MDS, the overall response rate of single Ara-c treatment was only 30%~40%. Nuclear factor erythroid 2-related factor 2 (NRF2), a basic leucine zipper transcription factor, has been reported to play a pivotal role in chemical detoxification. It is not yet known whether NRF2 has utility as a prognostic biomarker, or whether an elevated NRF2 level is associated with Ara-c resistance in MDS patients.To investigate the expression and effects of NRF2 in MDS patients, gene set enrichment analysis (GSEA) and immunohistochemistry (IHC) were performed. GSEA in a published MDS cohort (n=183) revealed that expressions of NRF2 target genes was significantly enriched in high risk MDS (REAB-1/2) compared to low-risks (RARS and RA). We further detected NRF2 expression levels using IHC staining in the bone marrow (BM) biopsies from a cohort of MDS patients (n=111). Consistent with GSEA results, our IHC data indicated that the NRF2 expression levels of BM from high-risks exceeded low-risk MDS patients by Revised International Prognosis Scoring System (IPSS-R). Importantly, MDS patients with higher NRF2 levels (IHC scores, 4~6) displayed worse overall survival (OS) than patients with lower NRF2 levels (IHC scores, 0~3) (median, 15.7 vs. 22.7 months, P=0.007). Subgroup analysis showed that higher NRF2 levels correlated with poor OS only in chemotherapy group (P=0.04) but not in decitabine (P=0.38) or supportive care group (P=0.19).To recapitulate our findings regarding the function of NRF2 in chemo-resistance, we performed in-vitro and in-vivo experiments. All experiments were performed both in human MDS-derived cell line SKM-1 and MDS mouse model cell line RUNX1 mutant-transduced MllPTD/WT BM cells (MllPTD/WT/RUNX1-S291fs). The cytotoxicity of Ara-c was determined by calculating the half maximal inhibitory concentration (IC50). NRF2 downregulation in MDS cell lines, by inhibitor luteolin, increases IC50 of Ara-c. On the contrary, upregulation of NRF2, mediated by agonist sulforaphane, induces resistance of MDS cells to Ara-C. To better define how suppression of NRF2 sensitized MDS cells to Ara-c, we used lentivirus-mediated shRNA for knockdown of NRF2. Knockdown of NRF2 markedly enhanced early and late apoptosis in MDS cell lines treated with Ara-c. We also established xenograft mouse models through intravenously injecting NRF2 shRNA or scramble shRNA SKM-1 cells into NOD/SCID-IL2Rγnull-SGM3 (NSGS) mice. Knockdown of NRF2 significantly sensitized tumors to Ara-c in our SKM-1 transplanted mouse model. To further investigate the mechanisms involved in NRF2-mediated Ara-c resistance, we analyzed published gene-expression profiles of a large cohort of MDS patients (n=183) and Ara-C sensitive (IC50 <6 μM AraC) and resistant (IC50 >80 μM) AML patient samples (n=10). The data indicated that a group of NRF2 target genes might be responsible for Ara-C resistence. Interestedly, DUSP1 was one of the genes upregulated in both high-risk MDS patients and Ara-c resistant AML patients. Our chromatin immunoprecipitation (ChIP) and qPCR results validated that DUSP1 was a NRF2 direct target gene. Importantly, downregulation of DUSP1 by inhibitor or lentivirus shRNA could abrogate Ara-c resistance in NRF2 elevated MDS cell lines, indicating that NRF2 confers Ara-c resistance partly through its downstream target gene DUSP1 in MDS cells.In conclusion, our clinical and experimental results reveal that 1) NRF2 expression levels are elevated in high-risk MDS patients and serve as a statistically significant prognostic variable for OS in patients with MDS, especially for MDS patients who have received chemotherapy. 2) Pharmacological inhibition of NRF2 re-sensitizes MDS cells to Ara-c treatment while activation of NRF2 by agonist resulted in the reduced sensitivity to Ara-c. 3) NRF2 mediates Ara-c resistance through its direct target gene DUSP1 . Taken together, our findings suggest that silencing NRF2 sensitizes MDS cells to Ara-c treatment; targeting NRF2 in combination with conventional chemotherapy could pave the way for high risk MDS therapy. DisclosuresNo relevant conflicts of interest to declare.

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