Open Access

Cimigenol depresses acute myeloid leukemia cells protected by breaking bone marrow stromal cells via CXCR4/SDF‑1α

  • Authors:
    • Bangyun Ma
    • Huibo Dai
    • Xingbin Dai
    • Shushu Qian
    • Xiaocao Sha
    • Xuemei Sun
  • View Affiliations

  • Published online on: December 30, 2022     https://doi.org/10.3892/etm.2022.11779
  • Article Number: 80
  • Copyright: © Ma et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The purpose of the present study was to evaluate cimigenol (Cim) treatment effects to cell proliferation by breaking bone marrow stromal cells (BMSCs) through C‑X‑C chemokine receptor type 4 (CXCR4)/stromal cell‑derived factor‑1α (SDF‑1α) pathway. MV‑4‑11 and U937 cell lines were used. The present study was divided into two parts. First, the cell lines were divided into normal control (NC), BMSC (cells co‑cultured with BMSCs), BMSC + DMSO, BMSC + Low (treated with 5 mg/ml Cim), BMSC + Middle (treated with 10 mg/ml Cim), BMSC + High (treated with 20 mg/ml Cim). In the second step, the cell lines were divided into NC, BMSC, BMSC + BL8040 (treated with BL8040 which inhibits CXCR4), BMSC + Cim and BMSC + Cim + BL8040. EdU positive cell numbers were measured by EdU assay and apoptosis rate by flow cytometry and TUNEL assay. Relative gene and protein expression was measured by reverse transcription‑quantitative PCR and western blotting assay. BMSCs were able to protect proliferation of cancer cells and decreased cell apoptosis compared with the NC group (P<0.001, respectively). With Cim supplement, the cell proliferation was decreased with cell apoptosis increasing compared with NC group (P<0.001 respectively). However, the anti‑tumor effects of Cim were not significantly different from the BL8040 treated groups (P<0.001, respectively). In conclusion Cim decreased acute myeloid leukemia cells protected by BMSCs through the CXCR4/SDF‑1α pathway.

Introduction

Acute myeloid leukemia (AML), characterized by blocked differentiation and clonal proliferation of hematopoietic stem/progenitor cells, is the commonest hematological malignancy and seriously endangers human health. So far, multiple methods including combined chemotherapy, targeted therapy and hematopoietic stem cell transplantation have not been effective in the treatment of AML (1). Therefore, exploring new therapies for AML is a focus of research. Bone marrow microenvironment (BMME) is a vital research field. Abnormal BMME has long been considered as one of the important factors affecting the onset of AML. Relevant studies have also confirmed that some components of BMME can promote the survival of AML cells and induce chemotherapy resistance by secreting cytokines and interacting with AML cells (2,3).

Natural drug extracts play an important role in the prevention and treatment of tumors in various systems, and have their own distinctiveness in the clinical treatment of leukemia. The regulation of the tumor microenvironment through multi-target effect on tumor cells is one of the important characteristics of anti-tumor therapy of traditional Chinese medicine. Cimigenol (Cim) is one of the main effective components of natural Cimicifugae Rhizoma (4). Research results have shown that Cim has a clear inhibitory effect on the biological activity of tumor cells (5-7). However, the role of Cim in AML and relevant mechanisms remains to be elucidated. In clinical treatment, it was found that Cim did not directly affect AML cells promoted by BMSCs (in vitro research data not shown as it is a part of another study), and due to this result, the present study did not use Cim to treat MV-4-11 or U937 cells. Therefore, in present study, the role of Cim in inducing AML cell apoptosis and inhibiting AML-related angiogenesis was explored. Cim might affect the occurrence and progression of AML by regulating BMME, but the specific regulatory mechanism is unclear and requires further exploration.

Materials and methods

Reagents and instruments

Cim was purchased from MilliporeSigma, and RPMI 1640 culture medium and fetal bovine serum from Gibco; Thermo Fisher Scientific, Inc. EdU kit, TUNEL kit and Annexin V-APC/7-AAD apoptosis kit were purchased from Jiangsu KeyGEN Biotech Co., Ltd. RNA extraction kit, reverse transcription kit, One Step TB Green PrimeScript RT-PCR kit II (SYBR Green) were purchased from Takara Bio, Inc. Primers were synthesized by Sangon Biotech (Shanghai) Co., Ltd. Rabbit anti-human GAPDH was purchased from Jiangsu KeyGEN Biotech Co., Ltd., and rabbit anti-human C-X-C chemokine receptor type 4 (CXCR4), stromal cell-derived factor-1α (SDF-1α), vascular cell adhesion molecule 1 (VCAM1), leukocyte function-associated antigen-1 (LFA-1), Fms like tyrosine kinase receptor 3 (FLT3), nucleophosmin 1 (NPM-1), CCAAT/enhancer-binding protein alpha (C/EBPA), AKT, phosphorylated (p-)AKT, mTOR and p-mTOR were purchased from Abcam; very-late-antigen-4 (VLA-4) was purchased from ProteinTech Group, Inc. Flow cytometer FC500 was from Beckman Coulter, Inc., ND2000 ultra-microspectrophotometer was from Thermo Fisher Scientific, Inc., and gel image analysis system was produced by GeneGenius. BL8040 (CXCR4 antagonist) was from Roche Diagnostics.

Cell culture

Human AML cell lines MV-4-11 and U937 were purchased from ATCC and primary BMSCs cell were purchased from Procell Life Science & Technology Co., Ltd. (cat. no. CP-H166) and cultured in RPMI 1640 medium containing 10% fetal bovine serum in a 5% CO2 incubator at 37˚C. According to cell proliferation, the medium was changed from time to time for subculture. Cells in the logarithmic growth phase were collected for subsequent experiment. MV-4-11 and U937 cell lines were not contaminated and the STR profiles were positive. The present study was approved by ethics Committee of Affiliated Hospital of Nanjing University of Chinese Medicine (approval no. 2021010606).

Cell treatment

MV-4-11 or U937 cells were inoculated onto 96-well plate (2x105/well), and subsequent experiments were performed when confluence reached >80%. The cells in the NC group were routinely cultured. In the BMSC group, MV-4-11 or U937 cells were co-cultured with BMSCs. In the BMSC + DMSO group, MV-4-11 or U937 cells were co-cultured with BMSCs, to which was added 250 µl DMSO. In the BMSC + Low group, MV-4-11 or U937 cells were co-cultured with BMSCs and to 250 µl of the mixed solution was added 5 mg/l Cim (final concentration). In the BMSC + Middle group, MV-4-11 or U937 cells were co-cultured with BMSCs and to 250 µl of the mixed solution was added 10 mg/l Cim (final concentration). In the BMSC + High group, MV-4-11 or U937 cells were co-cultured with BMSCs and to 250 µl of the mixed solution was added 20 mg/l Cim (final concentration). In the BMSC + BL8040 group, MV-4-11 or U937 cells were co-cultured with BMSCs, to which was added 10 nM BL8040. In the BMSC + Cim group, MV-4-11 or U937 cells were co-cultured with BMSCs and to 250 µl of the mixed solution was added 20 mg/l Cim (final concentration). In the BMSC + Cim + BL8040 group, MV-4-11 or U937 cells were co-cultured with BMSCs and to 250 µl of the mixed solution was added 20 mg/l Cim (final concentration) and 10 nM BL8040. After the cells in each group were treated for 48 h at room temperature, the subsequent experiment was conducted.

The co-culture method for the BMSCs cells was to inoculate on a treated glass slide and when the cells adhered, place the slide into the dish of AML cells and co-culture them.

Detection of cell proliferation by EdU staining

After corresponding treatment in each group for 48 h, 50 µmol/l EdU staining solution was added to each well for incubation for 2 h at room temperature, followed by washing with PBS for 3 times. Then, 4% paraformaldehyde was added for fixation for 30 min, and 50 µl 2 mg/ml glycine was added for incubation on a shaking table for 5 min. Additionally, 100 µl 0.5% TritonX-100 was added for penetration enhancement at room temperature, followed by PBS washing for 3 times. Afterwards, each well had 100 µl Hoechst33342 staining solution added for reaction at room temperature in the dark for 30 min, followed by washing with PBS for 3 times. Finally, observation and capturing of images were conducted under a fluorescence microscope, with three duplicated wells in each group.

TUNEL assay

After corresponding treatment for 48 h, MV-4-11 or U937 cells were collected from each group, fixed with 4% paraformaldehyde for 30 min at room temperature, and then washed twice with PBS. After adding 100 µl TUNEL balanced buffer and incubation at room temperature for 5 min, 50 µl reaction buffer was finally added for incubation in the dark for 60 min at room temperature. Following centrifugation (8,000 x g, 4˚C, 2 min), the supernatant was discarded, followed by washing with 5x10-3 mg/l BSA. The morphological changes of cells were observation and capturing of images were conducted under a fluorescence microscope.

Flow cytometry

After corresponding treatment for 48 h, MV-4-11 or U937 cells were collected from each group. In 1 h after Annexin V/PI staining at room temperature, 10,000 cells were collected and fixed in each group, and the apoptotic rate of hepatoma cells was detected by flow cytometry. Samples were repeated three times in each group. The quantification was analyzed by FlowJo 7.6.5 software (FlowJo LLC). The apoptosis rate=early + late apoptotic cells/all cells x100%

Reverse transcription-quantitative (RT-q) PCR

After corresponding treatment for 48 h, MV-4-11 or U937 cells (10,000 cells) were collected from each group. Total RNA was extracted from cells using TRIzol® (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. Then, 0.5 µg RNA was converted into cDNA at 37˚C for 1 h using PrimeScript RT MasterMix (Takara Bio, Inc.). qPCR was performed using ChamQ SYBR® qPCR MasterMix (Vazyme Biotech, Co., Ltd.). Primer sequences are listed in Table I. The reaction system (20 µl) included: 10 µl SYBR FAST qPCR Mix (2X), 1 µl upstream primer (10 µmol/l), 1 µl downstream primer (10 µmol/l), 2 µl cDNA template and 6 µl ddH2O. The reaction conditions were as follows: 95˚C for 5 min, 95˚C for 15 sec, 60˚C for 1 min for 40 cycles. Finally, the relative expression of each gene was analyzed using the 2-ΔΔCq method (8). The experiment was repeated three times.

Table I

Primer sequences.

Table I

Primer sequences.

GeneForward (5'-3')Reverse (5'-3')Size
AKT CAGGATGTGGACCAACGTGA AAGGTGCGTTCGATGACAGT137 bp
c/EBPα GACAAGAACAGCAACGAGTACC GTCATTGTCACTGGTCAGCTC132 bp
CXCR4 TTCCAGTTTCAGCACATCATGG GTCGATGCTGATCCCAATGTAG192 bp
FLT3 GTGAATCCTTACCCTGGCATTC GTCAAATTAGGGAAGGATGGCC164 bp
LFA-1 GGTTGACGTGGTGTATGAGAAG GAAACCAACCTTGTACAGCACT109 bp
mTOR AACCTCCTCCCCTCCAATGA TCAGCGGTAAAAGTGTCCCC92 bp
NPM-1 CACCAAAAGGACCTAGTTCTGT TGCCAGAGATCTTGAATAGCCT157 bp
SDF-1α GATTCTTCGAAAGCCATGTTGC TCAATGCACACTTGTCTGTTGT121 bp
VCAM-1 AGTTCTTGTTTGCCGAGCTAAA AAATCTCTGGAGCTGGTAGACC197 bp
VLA-4 AACATGAGCAGATTGGTAAGGC CAGACAGAAGCTCCAAAGTACG112 bp
GAPDH CAAATTCCATGGCACCGTCA AGCATCGCCCCACTTGATTT109 bp
Western blotting

After corresponding treatment for 48 h, MV-4-11 or U937 cells were collected from each group. Total protein was extracted from cells using RIPA buffer (Changsha Auragene Biological Technology Co., Ltd.) and quantified using a BCA Protein Assay kit (Beijing Dingguo Changsheng Biotechnology, Co., Ltd.). The lysates were incubated at 95˚C for 5 min, an equal amount of total protein (30 µg/lane), separated using 10% SDS-PAGE (Bio-Rad Laboratories) and transferred onto PVDF membranes (MilliporeSigma).

After being blocked with 5% skimmed milk at room temperature for 2 h, the membrane was incubated with primary antibodies CXCR4 (1:1,000; cat. no. ab16502), SDF-1α (1:1,000; cat. no. ab25117), VLA-4 (1:1,000; ProteinTech Group, Inc., cat. no. 19676-1-AP), VCAM1 (1:1,000; cat. no. ab134047), LFA-1 (1:1,000; cat. no. ab235456), FLT3 (1:1,000; cat. no. ab52648), NPM-1 (1:1,000; cat. no. ab10530), C/EBPA (1:1,000; cat. no. ab40761), AKT (1:1,000; cat. no. ab179463), mTOR(1:1,000; cat. no. ab2732), p-AKT(1:1,000; cat. no. ab81283), p-mTOR(1:1,000; cat. no. ab109268) or GAPDH (1:500; cat. no. ab8245) at 4˚C overnight. The membrane was washed with TBST three times for 10 min. Subsequently, HRP-labeled goat anti-rabbit IgGⅡ antibody (1:5,000) was added for incubation at room temperature for 1 h, and the membrane was washed with TBST three times for 10 min. Finally, Proteins were visualized using an ECL reagent kit (Shanghai Yeasen Biotech Co., Ltd.) and were semi-quantified using ImageJ software (1.46r; National Institutes of Health).

Statistical analysis

Experiments were performed in triplicate at minimum. Data are presented as the mean ± standard deviation and were analyzed using GraphPad Prism 8.0 (GraphPad Software, Inc.). For statistical analysis, pairwise comparisons between two groups were analyzed using the unpaired Student's t-test. One-way ANOVA followed by Tukey's post hoc test was used for comparisons between >2 groups. P<0.05 was considered to indicate a statistically significant difference.

Results

Different Cim concentrations suppresses AML cell proliferation protected by BMSC

Compared with NC group, EdU positive cell rates of BMSC and BMSC + DMSO groups were significantly upregulated in MV-4-11 and U937 cell lines (P<0.001, respectively, Fig. 1A and B). With Cim supplement, compared with BMSC group, EdU positive cell rates of Cim treated groups were significantly depressed in MV-4-11 and U937 cell lines (P<0.05, P<0.01 or P<0.001, respectively, Fig. 1A and B) and was dose-dependent (P<0.05, respectively, Fig. 1A and B).

Different Cim concentrations increase AML cell apoptosis protected by BMSC by flow cytometry

Compared with NC group, apoptosis rates of BMSC and BMSC + DMSO groups were significantly downregulated in MV-4-11 and U937 cell lines (P<0.001, respectively, Fig. 2A and B). With Cim supplement, compared with BMSC group, apoptosis rates of Cim treated groups were significantly increased in MV-4-11 and U937 cell lines (P<0.05, P<0.01 or P<0.001, respectively, Fig. 2A and B) with dose-dependent (P<0.05, respectively, Fig. 2A and B).

Different Cim concentrations increase number of apoptotic AML cells protected by BMSC by TUNEL assay

Compared with the NC group, the number of positive apoptotic cells in BMSC and BMSC + DMSO groups was significantly downregulated in MV-4-11 and U937 cell lines (P<0.001, respectively, Fig. 3A and B). With Cim supplement, compared with BMSC group, the number of positive apoptotic cells in Cim treated groups was significantly increased in MV-4-11 and U937 cell lines (P<0.05, P<0.01 or P<0.001, respectively; Fig. 3A and B) dose-dependently (P<0.05, respectively; Fig. 3A and B).

Different Cim concentrations affect relative gene expression by RT-qPCR assay

Compared with the NC group, CXCR4, SDF-1α, mTOR, AKT, VLA-4, VCAM-1, LFA-1 and C/EBPα mRNA expression in BMSC and BMSC + DMSO groups was significantly upregulated and FLT3 and NPM-1 mRNA expression in BMSC and BMSC + DMSO groups was significantly downregulated in MV-4-11 and U937 cell lines (P<0.001, respectively, Fig. 4A and B). With Cim, compared with the BMSC group, CXCR4, SDF-1α, mTOR, AKT, VLA-4, VCAM-1, LFA-1 and C/EBPα mRNA expression was significantly downregulated and FLT3 and NPM-1 mRNA expression was significantly upregulated in MV-4-11 and U937 cell lines (P<0.05, P<0.01 or P<0.001, respectively, Fig. 4A and B) dose-dependently (P<0.05, respectively, Fig. 4A and B).

Different Cim concentrations affect relative protein expression by western blotting

Compared with the NC group, CXCR4, SDF-1α, p-mTOR, p-AKT, VLA-4, VCAM-1, LFA-1 and C/EBPα protein expression of BMSC and BMSC + DMSO groups was significantly upregulated, and FLT3 and NPM-1 protein expression of BMSC and BMSC + DMSO groups was significantly downregulated in MV-4-11 and U937 cell lines (P<0.001, respectively, Fig. 5A and B). With Cim, compared with the BMSC group, CXCR4, SDF-1α, p-mTOR, p-AKT, VLA-4, VCAM-1, LFA-1 and C/EBPα protein expression was significantly downregulated and FLT3 and NPM-1 protein expression was significantly upregulated in MV-4-11 and U937 cell lines (P<0.05, P<0.01 or P<0.001, respectively, Fig. 5A and B) dose-dependently (P<0.05, respectively, Fig. 5A and B).

Effect of CXCR4 on the anti-tumor effects of Cim in cell proliferation

Compared with the NC group, EdU positive cell rates in BMSC groups were significantly upregulated in MV-4-11 and U937 cell lines (P<0.001, respectively, Fig. 6A and B). With BL8040 (CXCR4 inhibitor) and/or Cim treatment, compared with BMSC group, EdU positive cell rates of BMC + BL8040, BMSC + Cim and BMSC + Cim + BL8040 groups were significantly suppressed (P<0.001, respectively, Fig. 6A and B).

Effect of CXCR4 on the anti-tumor effects of Cim in cell apoptosis by flow cytometry

Compared with the NC group, apoptosis rates in BMSC groups were significantly downregulated in MV-4-11 and U937 cell lines (P<0.001, respectively, Fig. 7A and B). With BL8040 (CXCR4 inhibitor) and/or Cim treatment, compared with BMSC group, apoptosis rates of BMC + BL8040, BMSC + Cim and BMSC + Cim + BL8040 groups were significantly increased (P<0.001, respectively, Fig. 7A and B).

Effect of CXCR4 on the anti-tumor effects of Cim in cell apoptosis by TUNEL assay

Compared with the NC group, number of positive apoptotic cells in the BMSC groups was significantly downregulated in MV-4-11 and U937 cell lines (P<0.001, respectively, Fig. 8A and B). With BL8040 (CXCR4 inhibitor) and/or Cim treatment, compared with BMSC group, the number of positive apoptotic cells in the BMC + BL8040, BMSC + Cim and BMSC + Cim + BL8040 groups was significantly increased (P<0.001, respectively, Fig. 8A and B).

CXCR4 inhibitor affects relative gene expression

Compared with the NC group, CXCR4, SDF-1α, mTOR, AKT, VLA-4, VCAM-1, LFA-1 and C/EBPα mRNA expression in BMSC groups was significantly upregulated, and FLT3 and NPM-1 mRNA expression in BMSC groups was significantly downregulated in MV-4-11 and U937 cell lines (P<0.001, respectively, Fig. 9A and B). With BL8040 (CXCR4 inhibitor) and/or Cim treatment, compared with the BMSC group, CXCR4, SDF-1α, mTOR, AKT, VLA-4, VCAM-1, LFA-1 and C/EBPα mRNA expression was significantly downregulated and FLT3 and NPM-1 mRNA expression was significantly upregulated in MV-4-11 and U937 cell lines (P<0.001, respectively, Fig. 9A and B).

CXCR4 inhibitor affects relative protein expression

Compared with the NC group, CXCR4, SDF-1α, mTOR, AKT, VLA-4, VCAM-1, LFA-1 and C/EBPα protein expression of BMSC groups was significantly upregulated, and FLT3 and NPM-1 protein expression of BMSC groups was significantly downregulated in MV-4-11 and U937 cell lines (P<0.001, respectively, Fig. 10A and B). With BL8040 (CXCR4 inhibitor) and/or Cim treatment, compared with BMSC group, CXCR4, SDF-1α, mTOR, AKT, VLA-4, VCAM-1, LFA-1 and C/EBPα protein expression was significantly downregulated and FLT3 and NPM-1 protein expression was significantly upregulated in MV-4-11 and U937 cell lines (P<0.001, respectively, Fig. 10A and B).

Discussion

Under the physiological state, BMSCs in BMME can produce a variety of adhesion molecules and chemokines, thus mediating multiple signal cascades to ensure and maintain the normal localization and homeostasis of HSCs in BM. In the pathological state of AML, these products can be hijacked and shared by AML cells, so that AML cells can obtain environmental conditions conducive to their own survival, expansion and progression, finally leading to the weakening of apoptosis in AML cells (9,10). The bindings and interactions of the chemokine receptor family represented by CXCR4 and its relevant ligands play a representative role in this process. CXCR4 directly or indirectly activates a variety of signal cascades by binding with its ligands to ensure the correct localization, homeostasis maintenance and normal survival of HSCs in BMME (11). CXCR4 may eventually cause the progression, difficulties in treatment and recurrence of AML by affecting the proliferation, migration, chemotaxis and angiogenesis as well as increasing the chemotherapy resistance of leukemia cells (12).

SDF-1α is one of the members of the chemokine family in BMME and also the only ligand of CXCR4. In BMME, many components such as BMSCs, immature osteoblasts and bone marrow endothelial cells can secrete SDF-1α (13). Among these cell components, BMSCs are the main source (14). SDF-1α is often expressed in bone marrow microvascular hot spots that attract the aggregation of AML cells in BMME (15). It can promote the survival and expansion of AML cells by guiding AML cells to a favorable environment in BMME (16). In addition, the adhesion between AML cells and bone marrow stroma in BMME is vital for the survival and proliferation of AML cells and SDF-1α participates in the regulation of this mechanism. This regulation can be briefly summarized as: SDF-1α regulates the adhesion of AML cells to matrix components in BMME by activating adhesion molecules, such as integrins CD44 and VLA-4, so that AML cells can obtain ‘concealment’ and produce chemotherapy resistance (17). The binding of CXCR4 and its ligand SDF-1α can initiate multiple Ca2+-dependent or independent signal events, leading to actin cytoskeleton reorganization and activating integrins, which results in its appropriate interaction with the endothelium of BM sinus and stromal cells, finally affecting the survival, chemotaxis, homing and proliferation of cells (18). A relevant study (19) demonstrated that CXCR4 inhibitor AMD3465 can promote the peripheral mobilization of AML cells and enhance the anti-leukemia effect of chemotherapeutic drugs. A further study (3) found that blocked CXCR4/SDF-1α interaction affects the activity of related downstream signaling pathways such as PI3K/AKT and MAPK and increases the mobilization rate of AML cells, finally leading to the increase in chemotherapy sensitivity. Experiments have confirmed that other CXCR4 antagonists and monoclonal antibodies, including LY2510924, CX-01, POL6326 and NOX-A12, can also effectively inhibit the growth of AML cells and produce sustained pharmacodynamic effects on peripheral mobilization of cells (20-22). Some related studies showed that the CXCR4/SDF-1α signaling pathway stimulation could improve cancer cell biological activating in vitro and in vivo studies (23,24). The results of the present study showed that after AML cells were co-cultured with BMSCs, the apoptosis of AML cells MV-4-11 and U937 was protected, the proliferation was increased and the CXCR4/SDF-1α signaling pathway was activated. Therefore, it was hypothesized that BMSCs possess a protective effect on AML cells.

Cim has an inhibitory effect on the activity of tumor cells in vivo and in vitro (5-7). The present study showed that Cim also had a certain killing effect on AML cells under the protection of BMSCs and that its mechanism might be through the inhibition of the CXCR4/SDF-1α signaling pathway, which further inhibited downstream AKT and mTOR activities and acted on the terminal VLA-4(25), VCAM-1(26), LFA-1(27), FLT3(28), NPM-1(29) and c/EBPα (30). Therefore, it is hypothesized that Cim can promote the apoptosis and inhibit the proliferation of AML cells by inhibiting the CXCR4/SDF-1α signaling pathway.

However, there were some limitations to the present study. It only studied the effect of cimigenol on AML cell lines via CXCR4/SDF-1α pathway; the anti-tumor effects of cimigenol might be regulated by other pathways, meanwhile there were some differences between in vitro in AML. Future in vivo studies will address the bioavailability of cimigenol.

Acknowledgements

Not applicable.

Funding

Funding: The present study was supported by the Youth Fund of Natural Science Foundation of Jiangsu Province (grant no. BK20201096).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

BM, HD and XMS contributed to the conceptualization and the design of the present study. BM, HD and XD performed the experiments and analyzed the data. XD, SQ, XCS and XMS were responsible for the acquisition, analysis and interpretation of the data. HD and XCS contributed to the drafting of the manuscript. XD and SQ confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the ethics committee of the Affiliated Hospital of Nanjing University of Chinese Medicine (approval no. 2021010606).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Kumar B, Garcia M, Weng L, Jung X, Murakami JL, Hu X, McDonald T, Lin A, Kumar AR, DiGiusto DL, et al: Acute myeloid leukemia transforms the bone marrow niche into a leukemia-permissive microenvironment through exosome secretion. Leukemia. 32:575–587. 2018.PubMed/NCBI View Article : Google Scholar

2 

Hanahan D and Coussens LM: Accessories to the crime: Functions of cells recruited to the tumor microenvironment. Cancer Cell. 21:309–322. 2012.PubMed/NCBI View Article : Google Scholar

3 

Uy GL, Rettig MP, Motabi IH, McFarland K, Trinkaus KM, Hladnik LM, Kulkarni S, Abboud CN, Cashen AF, Stockerl-Goldstein KE, et al: A phase 1/2 study of chemosensitization with the CXCR4 antagonist plerixafor in relapsed or refractory acute myeloid leukemia. Blood. 119:3917–3924. 2012.PubMed/NCBI View Article : Google Scholar

4 

Schmid D, Woehs F, Svoboda M, Thalhammer T, Chiba P and Moeslinger T: Aqueous extracts of Cimicifuga racemosa and phenolcarboxylic constituents inhibit production of proinflammatory cytokines in LPS-stimulated human whole blood. Can J Physiol Pharmacol. 87:963–972. 2009.PubMed/NCBI View Article : Google Scholar

5 

Sakurai N, Kozuka M, Tokuda H, Mukainaka T, Enjo F, Nishino H, Nagai M, Sakurai Y and Lee KH: Cancer preventive agents. Part 1: Chemopreventive potential of cimigenol, cimigenol-3,15-dione, and related compounds. Bioorg Med Chem. 13:1403–1408. 2005.PubMed/NCBI View Article : Google Scholar

6 

Jöhrer K, Stuppner H, Greil R and Çiçek SS: Structure-guided identification of black cohosh (actaea racemosa) triterpenoids with in vitro activity against multiple myeloma. Molecules. 25(766)2020.PubMed/NCBI View Article : Google Scholar

7 

Li X, Wang W, Fan Y, Wei Y, Yu LQ, Wei JF and Wang YF: Anticancer efficiency of cycloartane triterpenoid derivatives isolated from Cimicifuga yunnanensis Hsiao on triple-negative breast cancer cells. Cancer Manag Res. 10:6715–6729. 2018.PubMed/NCBI View Article : Google Scholar

8 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001.PubMed/NCBI View Article : Google Scholar

9 

Zaitseva L, Murray MY, Shafat MS, Lawes MJ, MacEwan DJ, Bowles KM and Rushworth SA: Ibrutinib inhibits SDF1/CXCR4 mediated migration in AML. Oncotarget. 5:9930–9938. 2014.PubMed/NCBI View Article : Google Scholar

10 

Peled A and Tavor S: Role of CXCR4 in the pathogenesis of acute myeloid leukemia. Theranostics. 3:34–39. 2013.PubMed/NCBI View Article : Google Scholar

11 

Sugiyama T, Kohara H, Noda M and Nagasawa T: Maintenance of the hematopoietic stem cell pool by CXCL12-CXCR4 chemokine signaling in bone marrow stromal cell niches. Immunity. 25:977–988. 2006.PubMed/NCBI View Article : Google Scholar

12 

Konoplev S, Rassidakis GZ, Estey E, Kantarjian H, Liakou CI, Huang X, Xiao L, Andreeff M, Konopleva M and Medeiros LJ: Overexpression of CXCR4 predicts adverse overall and event-free survival in patients with unmutated FLT3 acute myeloid leukemia with normal karyotype. Cancer. 109:1152–1156. 2007.PubMed/NCBI View Article : Google Scholar

13 

Jiang Z, Zhou W, Guan S, Wang J and Liang Y: Contribution of SDF-1α/CXCR4 signaling to brain development and glioma progression. Neurosignals. 21:240–258. 2013.PubMed/NCBI View Article : Google Scholar

14 

Greenbaum A, Hsu YM, Day RB, Schuettpelz LG, Christopher MJ, Borgerding JN, Nagasawa T and Link DC: CXCL12 in early mesenchymal progenitors is required for haematopoietic stem-cell maintenance. Nature. 495:227–230. 2013.PubMed/NCBI View Article : Google Scholar

15 

Sipkins DA, Wei X, Wu JW, Runnels JM, Côté D, Means TK, Luster AD, Scadden DT and Lin CP: In vivo imaging of specialized bone marrow endothelial microdomains for tumour engraftment. Nature. 435:969–973. 2005.PubMed/NCBI View Article : Google Scholar

16 

Tavor S, Eisenbach M, Jacob-Hirsch J, Golan T, Petit I, Benzion K, Kay S, Baron S, Amariglio N, Deutsch V, et al: The CXCR4 antagonist AMD3100 impairs survival of human AML cells and induces their differentiation. Leukemia. 22:2151–5158. 2008.PubMed/NCBI View Article : Google Scholar

17 

Avigdor A, Goichberg P, Shivtiel S, Dar A, Peled A, Samira S, Kollet O, Hershkoviz R, Alon R, Hardan I, et al: CD44 and hyaluronic acid cooperate with SDF-1 in the trafficking of human CD34+ stem/progenitor cells to bone marrow. Blood. 103:2981–2989. 2004.PubMed/NCBI View Article : Google Scholar

18 

Zhan T, Cao C, Li L, Gu N, Civin CI and Zhan X: MIM regulates the trafficking of bone marrow cells via modulating surface expression of CXCR4. Leukemia. 30:1327–1334. 2016.PubMed/NCBI View Article : Google Scholar

19 

Zeng Z, Shi YX, Samudio IJ, Wang RY, Ling X, Frolova O, Levis M, Rubin JB, Negrin RR, Estey EH, et al: Targeting the leukemia microenvironment by CXCR4 inhibition overcomes resistance to kinase inhibitors and chemotherapy in AML. Blood. 113:6215–6224. 2009.PubMed/NCBI View Article : Google Scholar

20 

Galsky MD, Vogelzang NJ, Conkling P, Raddad E, Polzer J, Roberson S, Stille JR, Saleh M and Thornton D: A phase I trial of LY2510924, a CXCR4 peptide antagonist, in patients with advanced cancer. Clin Cancer Res. 20:3581–3588. 2014.PubMed/NCBI View Article : Google Scholar

21 

Karpova D, Bräuninger S, Wiercinska E, Krämer A, Stock B, Graff J, Martin H, Wach A, Escot C, Douglas G, et al: Mobilization of hematopoietic stem cells with the novel CXCR4 antagonist POL6326 (balixafortide) in healthy volunteers-results of a dose escalation trial. J Transl Med. 15(2)2017.PubMed/NCBI View Article : Google Scholar

22 

Hoellenriegel J, Zboralski D, Maasch C, Rosin NY, Wierda WG, Keating MJ, Kruschinski A and Burger JA: The spiegelmer NOX-A12, a novel CXCL12 inhibitor, interferes with chronic lymphocytic leukemia cell motility and causes chemosensitization. Blood. 123:1032–1039. 2014.PubMed/NCBI View Article : Google Scholar

23 

Gros SJ, Graeff H, Drenckhan A, Kurschat N, Blessmann M, Rawnaq T and Izbicki JR: CXCR4/SDF-1α-mediated chemotaxis in an in vivo model of metastatic esophageal carcinoma. In Vivo. 26:711–718. 2012.PubMed/NCBI

24 

Zhu H, Sun Q, Tan C, Xu M, Dai Z, Wang Z, Fan J and Zhou J: Tacrolimus promotes hepatocellular carcinoma and enhances CXCR4/SDF-1α expression in vivo. Mol Med Rep. 10:585–592. 2014.PubMed/NCBI View Article : Google Scholar

25 

Holt RU, Fagerli UM, Baykov V, Rø TB, Hov H, Waage A, Sundan A and Børset M: Hepatocyte growth factor promotes migration of human myeloma cells. Haematologica. 93:619–622. 2008.PubMed/NCBI View Article : Google Scholar

26 

Fan X, Chen X, Feng Q, Peng K, Wu Q, Passerini AG, Simon SI and Sun C: Downregulation of GATA6 in mTOR-inhibited human aortic endothelial cells: Effects on TNF-α-induced VCAM-1 expression and monocytic cell adhesion. Am J Physiol Heart Circ Physiol. 316:H408–H420. 2019.PubMed/NCBI View Article : Google Scholar

27 

Wagner JA, Rosario M, Romee R, Berrien-Elliott MM, Schneider SE, Leong JW, Sullivan RP, Jewell BA, Becker-Hapak M, Schappe T, et al: CD56bright NK cells exhibit potent antitumor responses following IL-15 priming. J Clin Invest. 127:4042–4058. 2017.PubMed/NCBI View Article : Google Scholar

28 

Chen W, Drakos E, Grammatikakis I, Schlette EJ, Li J, Leventaki V, Staikou-Drakopoulou E, Patsouris E, Panayiotidis P, Medeiros LJ and Rassidakis GZ: mTOR signaling is activated by FLT3 kinase and promotes survival of FLT3-mutated acute myeloid leukemia cells. Mol Cancer. 9(292)2010.PubMed/NCBI View Article : Google Scholar

29 

Marzec M, Kasprzycka M, Liu X, El-Salem M, Halasa K, Raghunath PN, Bucki R, Wlodarski P and Wasik MA: Oncogenic tyrosine kinase NPM/ALK induces activation of the rapamycin-sensitive mTOR signaling pathway. Oncogene. 26:5606–5614. 2007.PubMed/NCBI View Article : Google Scholar

30 

Watanabe M, Hisatake M and Fujimori K: Fisetin suppresses lipid accumulation in mouse adipocytic 3T3-L1 cells by repressing GLUT4-mediated glucose uptake through inhibition of mTOR-C/EBPα signaling. J Agric Food Chem. 63:4979–4987. 2015.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

February-2023
Volume 25 Issue 2

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ma B, Dai H, Dai X, Qian S, Sha X and Sun X: Cimigenol depresses acute myeloid leukemia cells protected by breaking bone marrow stromal cells via CXCR4/SDF‑1α. Exp Ther Med 25: 80, 2023
APA
Ma, B., Dai, H., Dai, X., Qian, S., Sha, X., & Sun, X. (2023). Cimigenol depresses acute myeloid leukemia cells protected by breaking bone marrow stromal cells via CXCR4/SDF‑1α. Experimental and Therapeutic Medicine, 25, 80. https://doi.org/10.3892/etm.2022.11779
MLA
Ma, B., Dai, H., Dai, X., Qian, S., Sha, X., Sun, X."Cimigenol depresses acute myeloid leukemia cells protected by breaking bone marrow stromal cells via CXCR4/SDF‑1α". Experimental and Therapeutic Medicine 25.2 (2023): 80.
Chicago
Ma, B., Dai, H., Dai, X., Qian, S., Sha, X., Sun, X."Cimigenol depresses acute myeloid leukemia cells protected by breaking bone marrow stromal cells via CXCR4/SDF‑1α". Experimental and Therapeutic Medicine 25, no. 2 (2023): 80. https://doi.org/10.3892/etm.2022.11779