Abstract

Ubiquitin specific protease 35 (USP35) is a member of deubiquitylases (DUBs). It remains largely unknown about the biological role and the regulation mechanism of USP35. Here, we first identified miR let-7a as a positive regulator of USP35 expression and showed that USP35 expression positively correlates with miR let-7a expression in different cancer cell lines and tissues. Then, we showed that USP35 expression was decreased dramatically in the tumor tissues compared with the adjacent non-cancerous tissues. USP35 overexpression inhibited cell proliferation in vitro and inhibited xenograft tumor growth in vivo. Furthermore, we revealed that USP35 acts as a functional DUB and stabilizes TNFAIP3 interacting protein 2 (ABIN-2) by promoting its deubiquitination. Functionally, both ABIN-2 and USP35 could inhibit TNFα-induced NF-κB activation and overexpression of ABIN-2 alleviated USP35-loss induced activation of NF-κB. Collectively, our data indicated that miR let-7a-regulated USP35 can inhibit NF-κB activation by deubiquitination and stabilization of ABIN-2 protein and eventually inhibit cell proliferation. Overall, our study provides a novel rationale of targeting miR let-7a-USP35-ABIN-2 pathway for the therapy of cancer patients.

Highlights

  • MicroRNA (MiR) let-7a has been widely studied and functions as a tumor suppressor [1]

  • To confirm the results obtained from analyzing the mRNA profiling, we transfected let-7a mimics or inhibitor into LNCaP cells and used Quantitative real time polymerase chain reaction (qRT-PCR) and Western blot to detect the expression of Ubiquitin specific protease 35 (USP35)

  • Consistent with the results of microarray analysis, the results showed that let-7a mimics increased, whereas let-7a inhibitor decreased both mRNA and protein level of USP35 (Figure 1A–1C)

Read more

Summary

Introduction

MicroRNA (MiR) let-7a has been widely studied and functions as a tumor suppressor [1]. Previous work has shown miR let-7a inhibits cell proliferation by downregulating oncogenes RAS/c-MYC [5] and HMGA-2 [6] at the translational level in lung cancer cells. MiR let-7a targets E2F2 and CCND2 to inhibit cell proliferation [4]. We previously demonstrated that miR let-7a directly targets the 3′ UTR of the IGF1R mRNA and inhibits its expression in prostate cancer cells [7]. Accumulating evidences indicate that miR let-7a exerts its anti-tumor activity by regulating oncogenes or tumor suppressor genes [8], the precise mechanisms of miR let-7a in the pathogenesis of human cancers still remain unclear and the full set of its regulatory substrates need to be further elucidated

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