Abstract

The gradual emerging of resistance to imatinib urgently calls for the development of new therapy for chronic myeloid leukemia (CML). The fusion protein Bcr-Abl, which promotes the malignant transformation of CML cells, is mainly located in the cytoplasm, while the c-Abl protein which is expressed in the nucleus can induce apoptosis. Based on the hetero-dimerization of FKBP (the 12-kDa FK506- and rapamycin-binding protein) and FRB (the FKBP-rapamycin binding domain of the protein kinase, mTOR) mediated by AP21967, we constructed a nuclear transport system to induce cytoplasmic Bcr-Abl into nuclear. In this study, we reported the construction of the nuclear transport system, and we demonstrated that FN3R (three nuclear localization signals were fused to FRBT2098L with a FLAG tag), HF2S (two FKBP domains were in tandem and fused to the SH2 domain of Grb2 with an HA tag) and Bcr-Abl form a complexus upon AP21967. Bcr-Abl was imported into the nucleus successfully by the nuclear transport system. The nuclear transport system inhibited CML cell proliferation through mitogen-activated protein kinase (MAPK) and signal transducer and activator of transcription 5 (STAT5) pathways mainly by HF2S. It was proven that nuclear located Bcr-Abl induced CML cell (including imatinib-resistant K562G01 cells) apoptosis by activation of p73 and its downstream molecules. In summary, our study provides a new targeted therapy for the CML patients even with Tyrosine Kinase Inhibitor (TKI)-resistance.

Highlights

  • Chronic myeloid leukemia (CML) is a malignant tumor derived from myeloid stem cells and characterized by the Philadelphia chromosome [1,2]

  • It has been previously reported that nuclear accumulation of Bcr-Abl with active tyrosine kinase activity triggers apoptosis, which was achieved by treatment with imatinib when nuclear export is blocked by Leptomycin B (LMB) [10,11]

  • We examined whether the nuclear transport system directs Bcr-Abl into the nucleus and depletes it from the cytoplasm, whether it inhibits growth and promotes apoptosis of CML cells, and the underlying mechanisms

Read more

Summary

Introduction

Chronic myeloid leukemia (CML) is a malignant tumor derived from myeloid stem cells and characterized by the Philadelphia chromosome [1,2]. AP21967 enhanced the suppression of effect on p-STAT5 (Figure 4D) These results confirmed that the HF2S significantly inhibited CML cell proliferation by down-regulation of the kinase activities of MAPK-Akt and STAT5 pathways. Addition of AP21967 synergistically enhanced the apoptotic effect on CML cells: 49.5% in K562 cells, 47.4% in K562G01 cells, and 58.9% in 32D-p210 cells These results suggested that blockade of Y177 by HF2S can induce CML cell apoptosis, while a combination of Y177 blockade and Bcr-Abl nuclear translocation significantly promoted CML cell apoptosis. Most Bcr-Abl resides in the cytoplasm [30,31] and causes the malignant proliferation of CML cells by activating multiple signaling pathways, including JAK/STAT, MAPK/RAS/ERK and PI3K/Akt pathways [32,33]. Since the distinct effects of c-Abl are in different subcellular domains, we constructed a nuclear transport system to

Discussion
Materials and Methods
Cell Culture
Cell Proliferation and Colony Formation Assay
Apoptosis Analyses
Western Blot
Co-Immunoprecipitation Assay
Immunofluorescence Assay
Findings
Conclusions
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