Abstract

Post-transcriptional gene regulation, including that by RNA binding proteins (RBPs), has recently been described as an important mechanism in cancer. We had previously identified a set of RBPs that were highly dysregulated in B-cell acute lymphoblastic leukemia (B-ALL) with MLL translocations, which carry a poor prognosis. Here, we sought to functionally characterize these dysregulated RBP genes by performing a focused CRISPR dropout screen in B-ALL cell lines, finding dependencies on several genes including EIF3E, EPRS and USO1. Validating our findings, CRISPR/Cas9-mediated disruption of USO1 in MLL-translocated B-ALL cells reduced cell growth, promoted cell death, and altered the cell cycle. Transcriptomic analysis of USO1-deficient cells revealed alterations in pathways related to mTOR signaling, RNA metabolism, and targets of MYC. In addition, USO1-regulated genes from these experimental samples were significantly and concordantly correlated with USO1 expression in primary samples collected from B-ALL patients. Lastly, we found that loss of Uso1 inhibited colony formation of MLL-transformed in primary bone marrow cells from Cas9-EGFP mice. Together, our findings demonstrate an approach to performing focused sub-genomic CRISPR screens and highlight a putative RBP vulnerability in MLL-translocated B-ALL, thus identifying potential therapeutic targets in this disease.

Highlights

  • Post-transcriptional gene regulation, including that by RNA binding proteins (RBPs), has recently been described as an important mechanism in cancer

  • Comparing the results across cell lines, we found that single guide RNAs (sgRNAs) targeting three genes, USO1, EIF3E and EPRS were significantly depleted in SEM cells (p < 0.001), when compared to NALM6 cells (Fig. 1E)

  • We sought to understand whether overexpression of putative RBPs, which we identified previously, contributes to the pathogenesis of MLL-AF4+ B-ALL15

Read more

Summary

Introduction

Post-transcriptional gene regulation, including that by RNA binding proteins (RBPs), has recently been described as an important mechanism in cancer. Patients with MLL-rearranged B-ALL have a dismal prognosis, with 5-year event-free survival rates hovering at 33.6% for i­nfants[7] and 50% for older children and a­ dults[8] Most of these patients are resistant to conventional treatment with chemotherapy and ­steroids[9], with bone marrow transplantation being the only curative therapeutic a­ lternative[10]. We performed a sub-genomic CRISPR/Cas[9] dropout screen using 36 highly upregulated RBPs in primary human B-ALL and identified several novel vulnerabilities that included three putative RBPs. Of these, USO1, a putative RBP and a known regulator of vesicular transport, was identified as a MLL-AF4 target gene. CRISPR/Cas9-mediated disruption of USO1 significantly altered cell growth and the cell cycle in B-ALL cell lines; as well as inhibited the colony forming potential of MLL-transformed primary murine bone marrow cells. Our studies provide a comprehensive rubric to functionally evaluate putative targets identified from expression profiling, and the identification of a novel potential target in MLL-rearranged leukemia

Methods
Results
Conclusion
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