Immunocomplexes were visualized with chemiluminescence kit (GE Healthcare UK Ltd) according to the manufacturers instr-uctions

Immunocomplexes were visualized with chemiluminescence kit (GE Healthcare UK Ltd) according to the manufacturers instr-uctions. MEK/ERK signaling was required for enz-ATRA treatment-induced differentiation via modulation of the protein levels of C/EBP and/or PU.1. Enz-ATRA treatment collapsed mitochondrial transmembrane potential without the activation of caspase-3, -6 and -7. Moreover, caspase-3/7- and caspase-6-specific inhibitors had no inhibitory effect on enz-ATRA treatment-triggered apoptosis. Therefore, enz-ATRA treatment-induced apoptosis was mitochondria-dependent but caspase-independent. Enz-ATRA treatment degraded PML-RAR, which may be involved in enz-ATRA treatment-induced dual effects and may also be beneficial for APL eradication. These findings may provide a potential therapy for ATRA-resistant APL patients. and via targeting of PML-RAR [2]. However, the clinical applicability of LG-362B remains to be determined. Other agents, such as cAMP, STI571, granulocyte colony-stimulating factor, tumor necrosis factor, oridonin, dasatinib, matrine and interferon- have been shown to synergize with ATRA to induce differentiation in ATRA-resistant APL cells [10-17]. Clinical trials are urgently needed to verify their efficacy. Protein kinase C (PKC) is a family of serine/threonine kinases, which consists of 13 isozymes that are involved in proliferation, differentiation, apoptosis, cell migration and gene expression. Intensive studies have explored the role of PKC in carcinogenesis and have rendered it as an attractive target for cancer therapy. PKC is specifically down-regulated during human neutrophil terminal differentiation, suggesting Rabbit polyclonal to RBBP6 its negative role in neutrophil differentiation [18]. Although PKC activity has been confirmed to be increased by ATRA treatment, both in the APL cell line-NB4 and in APL primary cells, its role in ATRA-induced granulocytic differentiation has been controversial [19-22]. A structural-biology study showed that ATRA competed with a PKC activator URAT1 inhibitor 1 to URAT1 inhibitor 1 bind to the C2-domian of PKC and may thereby modulate PKC activity [23]. Interestingly, PKC and PKC are able to phosphorylate retinoic acid receptor (RAR) at S157 and subsequently disrupt the formation of RAR/retinoid X receptor (RXR) heterodimer, resulting in decreased transcriptional activity [24]. Therefore, there is interference between retinoic acid (RA)-signaling and PKC-signaling pathways. Moreover, PKC contributes to ATRA resistance by overexpression of topoisomerase II [19]. However, activated PKC has also been demonstrated to be required for ATRA-induced differentiation in APL cells [22]. Therefore, the role of PKC in ATRA-induced differentiation in APL cells has been disputed. Enzastaurin is an isoenzyme-specific derivative of PKC pan-inhibitor staurosporine. It was designed to suppress the activation of PKC by inhibiting the binding of ATP. Unlike the unacceptable toxicity of staurosporine, enzastaurin has been demonstrated to be safe and well tolerated in multiple clinical trials. Moreover, it has exhibited promising anti-cancer activity in a variety of preclinical studies [25]. For hematological malignances, enzastaurin either as a single agent or in combination with other medicines exerts anti-cancer activity in acute myeloid leukemia, lymphoma and multiple myeloma cells by inhibiting proliferation or promoting apoptosis [25]. However, to our knowledge, enzastaurin has not yet been reported to induce/enhance differentiation. As mentioned above, since PKC may be one of the mediators of ATRA resistance in APL-relapsed patients and may also be the negative regulator of neutrophil-terminal differentiation, these phenomena prompted us to investigate whether enzastaurin could restore ATRA sensitivity in ATRA-resistant APL cell lines. This study used clinically achievable concentrations of enzastaurin. Unexpectedly, the combination of enzastaurin and ATRA (enz-ATRA) induced both terminal granulocytic differentiation and apoptosis in ATRA-resistant APL cell lines, NB4-R1 and NB4-R2, URAT1 inhibitor 1 in a dose-dependent manner. Further study showed that the enz-ATRA combination-overcoming differentiation block required MEK/ERK-mediated modulation of the protein levels of CCAAT/enhancer-binding protein (C/EBP) and/or PU.1. Additionally, the enz-ATRA combination-induced apoptosis was mitochondria-dependent but caspase-independent. Enzastaurin also synergized with ATRA to degrade PML-RAR, the pathogenic protein of APL. Material and methods Reagents ATRA was purchased from Sigma-Aldrich (St Louis, MO, USA). Enzastaurin and sorafenib tosylate were purchased from Selleckchem Chemicals (Houston, TX, USA). U0126 and Z-DEVD-FMK were obtained from EMD Chemicals (San Diego, CA, USA). Z-VEID-FMK was purchased from R&D systems (Minneapolis, MN, USA). A PKC inhibitor was obtained from Merck (Darmstadt, Germany). All reagents were dissolved in dimethyl sulfoxide (DMSO). Cell culture, cell viability and cell proliferation The ATRA-resistant cell lines, NB4-R1 and NB4-R2 (kindly gifted from Dr Michel Lanotte, Hopital Saint Louis, Paris, France), were cultured in RPMI-1640, supplemented with 10% fetal calf serum (Thermo Fisher Scientific Inc, Waltham, MA, USA) in a humidified atmosphere of 95% air and 5% CO2 at 37C. Trypan-blue exclusion was used to evaluate cell viability. Cell differentiation assays Cell maturation was evaluated.