Abstract

Identification of new genetic pathways or molecular targets that sensitize cancer cells to chemotherapeutic drugs may improve the efficacy of current chemotherapy. Here, we report that downmodulation of UHRF1 (ubiquitin‐like with PHD and RING finger domains 1) in retinoblastoma (RB) cells increases the sensitivity to histone deacetylase (HDAC) inhibitors, augmenting apoptotic cell death. We found that UHRF1 depletion downregulates two redox‐responsive genes GSTA4 (glutathione S‐transferase α4) and TXN2 (thioredoxin‐2) in RB cells, and increases the basal level of intracellular oxidative stress. Antioxidant treatment significantly reduced both basal and HDAC inhibitor‐induced DNA damage and apoptosis in UHRF1‐depleted cells. Knockdown of GSTA4 or TXN2 sensitized RB cells to HDAC inhibitors, demonstrating that GSTA4 and TXN2 play key roles in redox homeostasis in RB cells and the susceptibility to HDAC inhibitor treatment upon UHRF1 depletion. In human primary RB, GSTA4 and TXN2 proteins were found to be mostly elevated along with high UHRF1 expression. In addition to augmentation of apoptosis in UHRF1‐depleted RB cells, we also show that UHRF1 downmodulation derepresses the expression of photoreceptor‐specific genes in RB cells in cooperation with a HDAC inhibitor MS‐275 and promotes neuron‐like differentiation. However, further investigation revealed that the enhanced growth‐inhibitory effects of MS‐275 in UHRF1‐depleted cells were still mainly due to robust apoptosis induction rather than differentiation‐mediated growth arrest. Consistent with our findings, UHRF1 depletion in RB cells increased the therapeutic efficacy of MS‐275 in murine orthotopic xenografts. These results provide a novel basis for potential benefits of UHRF1 targeting for RB treatment.

Highlights

  • Retinoblastoma (RB) is a major intraocular cancer occurring in children and is initiated by inactivation of the RB1 tumor suppressor gene in the developing retina (Dimaras and Corson, 2019)

  • We examined the relative viability of control and UHRF1-knockdown Y79 RB cells in response to several histone deacetylase (HDAC) inhibitors (Fig. 1A–C)

  • As UHRF1 depletion derepresses the expression of photoreceptor genes in RB cells, we investigated whether UHRF1-depleted cells would have a higher differentiation potential and whether this feature can contribute to growth inhibition in combination with MS-275 treatment

Read more

Summary

Introduction

Retinoblastoma (RB) is a major intraocular cancer occurring in children and is initiated by inactivation of the RB1 tumor suppressor gene in the developing retina (Dimaras and Corson, 2019). As a standard treatment option, chemotherapy has been widely used in combination with various types of adjuvant focal therapy to save the eye and reduce the long-term risks of developing secondary tumors (Chan et al, 2005; Wyse et al, 2016). The current chemotherapy for RB has limitations such as drug resistance and Abbreviations GSTA4, glutathione S-transferase a4; HDAC, histone deacetylase; NaBu, sodium butyrate; NAC, N-acetylcysteine; PDL, poly-D-lysine; RA, alltrans retinoic acid; RB, retinoblastoma; RCVRN, recoverin; ROS, reactive oxygen species; RXRG, retinoid X receptor c; SAHA, suberoylanilide hydroxamic acid; TXN2, thioredoxin-2; UHRF1, ubiquitin-like with PHD and RING finger domains 1.

Methods
Discussion
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