Background: Special AT-rich sequence-binding protein 1(SATB1) is a chromatin-remodeling protein that has been shown to regulate gene expressions and aberrant cell growth in different types of cancer. Up-regulation of SATB1 is linked with progression and metastasis of tumors. It is also reported that SATB1 is involved in the differentiation of hematopoietic lineages. However, less is known about the association between SATB1 and hematology malignancy. We previously reported that SATB1 expressions were decreased in T cell leukemia/lymphoma (T-ALL), and knockdown of SATB1 significantly enhanced invasiveness of Jurkat cell through the activation of Wnt/β-catenin signaling pathway in vitro. The contrary roles of SATB1 in solid organ tumors and hematology malignancy may provide hints to study the function of SATB1. Here, we investigated the role of SATB1 in acute myeloid leukemia (AML).

Methods: To characterize SATB1 mRNA and protein expression in AML, we performed qRT-PCR and Western blot on peripheral blood mononuclear cells (PBMCs) from 52 newly diagnosed AML patients, as well as AML cell lines NB4, HEL, THP-1 and HL-60. Stable HL-60 cell lines with knockdown of SATB1 by shRNAs sequences (HL-60 SATB1-shRNA1 and HL-60 SATB1-shRNA2) were established. CCK-8 cell proliferation assay and flow cytometric analysis were performed to evaluate the cell proliferation and cell cycle. The invasiveness of HL-60 SATB1-shRNAs and HL-60 CTR cells was analyzed and compared by transwell assay. In order to study the role of SATB1 in vivo, murine model was established using HL-60 SATB1-shRNAs treated nude mice and tumorigenicity was comopared by tumor size. After demonstrating the association between SATB1 knockdown and tumorigenicity in vitro and in vivo, we next investigated NF-κB, AKT and MAPK signaling pathways by western blot.

Results:l. SATB1 expression was significantly lower in AML patients than normal controls, which is similar to what we found in T-ALL patients. SATB1 expression was different before and after treatment, especially after complete remission (CR) of AML. Increased SATB1 expression was associated with better clinical outcome. We further investigated the roles of SATB1 on AML cells by shRNAs mediated knockdown of SATB1. Knockdown of SATB1 promoted the proliferation of HL-60 SATB1 shRNA1 cells and HL-60 SATB1 shRNA2 cells compared to the control. However, the transwell assay did not show increased invasiveness in SATB1 depleted HL-60 cells. The in vivo experiment showed that tumor size and weight in SATB1 knockdown mice were significantly increased compared to the control mice. In the mechanism study, we investigated NF-κB, AKT and MAPK signaling pathways by western blot. We found that the expression of NF-κB p65 in HL60 SATB1-shRNAs cells was increased, which suggested that SATB1 regulated AML cell growth via the activation of NF-κB signaling pathway.

Conclusions: Impairment of SATB1 is associated with AML. SATB1 expressions were decreased in AML. Knockdown of SATB1 promoted cell proliferation and tumorigenicity of AML via the activation of NF-κB signaling pathway.

Disclosures

No relevant conflicts of interest to declare.

Author notes

*

Asterisk with author names denotes non-ASH members.

Sign in via your Institution