Open Access

Expression and role of fibroblast activation protein α in acute myeloid leukemia

  • Authors:
    • Shuchong Mei
    • Yuan Zhang
    • Li Yu
    • Guoan Chen
    • Fuming Zi
  • View Affiliations

  • Published online on: November 30, 2020     https://doi.org/10.3892/or.2020.7874
  • Pages: 641-651
  • Copyright: © Mei 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

Currently, the prognosis of acute myeloid leukemia (AML) is poor. In the AML microenvironment, bone marrow (BM) mesenchymal stem cells (BMMSCs) serve an important role in protecting AML cells from chemotherapy‑induced apoptosis. The present study aimed to evaluate the expression of fibroblast activation protein α (FAPα) in BMMSCs and BM biopsy samples via flow cytometry, reverse transcription‑quantitative PCR and immunohistochemistry, as well as to identify the correlation between the expression of FAPα in BM with clinical parameters and survival of newly diagnosed patients with AML. Subsequently, the protective effect of FAPα on Cytosine arabinoside (Ara‑C)‑induced apoptosis in Kasumi‑1 cells was investigated via small interfering (si)RNA, and its underlying mechanism was examined by western blotting. The results demonstrated significant differences in FAPα expression in BMMSCs and BM biopsy samples between patients with AML and healthy donors. Furthermore, BMMSCs protected Ara‑C‑induced Kasumi‑1 cells from apoptosis, and knockdown of FAPα using siRNA decreased this protection. It was found that Kasumi‑1 cells expressed β‑catenin, which could be inhibited by Ara‑C, and β‑catenin expression was significantly activated when co‑cultured with BMMSCs, even in the presence of Ara‑C. Knockdown of FAPα with siRNA significantly suppressed the expression of β‑catenin. The present results indicated that FAPα serves an important role in the AML BM microenvironment, and that increased expression of FAPα in BM may be a poor prognostic factor in patients with AML. Moreover, the current findings demonstrated that BMMSCs protected AML cells from apoptosis, which was in part contributed by FAPα, and may occur via the β‑catenin signaling pathway.

Introduction

Acute myeloid leukemia (AML) is a common hematologic malignancy with a statistical incidence rate of 38 case per million individuals in the United States between 2001 and 2007 (1). In the last 20 years, no significant improvement has been achieved in the prognosis or long-term outcomes of AML, and a considerable number of patients still experience induction failure or relapse after complete remission (CR) (2). The interaction between tumor cells and the tumor microenvironment is one of the key mechanisms of chemoresistance in tumor cells, including AML (3,4). Bone marrow (BM) mesenchymal stem cells (BMMSCs) serve an important role in protecting AML cells from chemotherapy-induced apoptosis (5,6). Previous studies have reported that tumor cells recruit BMMSCs by secreting cytokines or chemokines, and culturing BMMSCs to become cancer-related fibroblasts (CAFs) supports the progression of malignant cells (7,8). CAFs have been considered as central components in the tumor microenvironment and serve vital roles in tumor features, such as proliferation, angiogenesis, invasion and metastasis (911). However, the exact origin of CAFs remains unknown (12).

BMMSCs have recently been recognized as one of the origins of CAFs (13,14). However, a precise molecular definition of CAFs has not yet been elucidated. Previous studies have revealed that CAFs express cell surface markers, including fibroblast activation protein (FAPα) (15). Across a wide range of human cancer types, such as gastric carcinoma, breast cancer and colon cancer, the expression of FAPα has been reported to correlate with a higher tumor grade and worse overall survival in solid tumors (1620). Considering the important role of FAPα in tumor progression and its rare expression in healthy tissues, FAPα has become a key target in tumor therapy (21,22). Previously, it was observed that tumor cells can reprogram BMMSCs to evolve into CAFs, thereby further promoting the progression of hematological malignancies (23). The expression of FAPα in BMMSCs moderately increases the number of tumor cells in conditioned medium from myeloma cells and those co-cultured with multiple myeloma (MM) cells (24). Furthermore, knockdown of FAPα with small interfering (si)RNA decreases the protection of bortezomib-induced apoptosis in MM cells (24). However, it remains unknown whether the expression of FAPα in BMMSCs is different between patients with AML and healthy donors, nor has it been elucidated whether FAPα serves an important role in mediating AML cell features. Therefore, the aim of the present study was to investigate the expression of FAPα in BMMSCs and BM, as well as to identify the role of FAPα in BMMSCs in protecting AML cells from apoptosis.

Materials and methods

Cell culture

The human AML cell line, Kasumi-1, was obtained from the Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic. Cells were cultured in RPMI-1640 (Thermo Fisher Scientific, Inc.) medium containing 10% heat-inactivated FBS (Thermo Fisher Scientific, Inc.), 1% l-glutamine, 100 U/ml penicillin and 100 µg/ml streptomycin at 37°C and 5% CO2.

BMMSCs were cultured according to a previously published paper (25). In total, 15 newly diagnosed patients with AML were selected, including six males and nine females (median age 56 years; age range 26-78 years). The inclusion criteria for patients with AML were as follows: i) Age ≥18 years old; ii) non-acute promyelocytic leukemia; ii) the diagnostic criteria were based on the 2016 World Health Organization diagnostic criteria for AML (26); and iv) no other malignant tumors-present, except AML. The control group consisted of healthy adults who were matched for age and sex with patients with AML. After informed consent was obtained, in keeping with ethical guidelines of The Second Affiliated Hospital of Nanchang University and the Declaration of Helsinki, BMMNCs were obtained from posterior superior iliac crest bone marrow of 15 healthy donors and 15 newly diagnosed patients with AML from The Second Affiliated Hospital of Nanchang University between January 2018 and January 2020 via lymphocyte separation medium (Tianjin Hao Yang Biological Manufacture Co., Ltd.), and were cultured. Half of the medium was replaced every 3-7 days. After culture for 2-4 weeks, cells became adherent and fibroblast-like, reaching >90% confluency. Then, cells were digested with 0.25% trypsin-EDTA and subcultured in RPMI-1640 (Thermo Fisher Scientific, Inc.) medium containing 10% heat-inactivated FBS (Thermo Fisher Scientific, Inc), 1% l-glutamine, 100 U/ml penicillin and 100 µg/ml streptomycin at 37°C and 5% CO2. MSCs of passage 2-6 were used in experiments. Clinical and demographic parameters of 15 newly diagnosed patients with AML and healthy donors are presented in Table I.

Table I.

Clinical and demographic parameters of participants in the study.

Table I.

Clinical and demographic parameters of participants in the study.

ParameterPatients with acute myeloid leukemiaHealthy donorsP-value
Number (untreated)15 (8)15 (15)>0.05
Age, years56 (26–78)47 (25–65)>0.05
Sex >0.05
  Male67
  Female98
FAB system classification
  M290
  M420
  M530
  M610
WBC, 109/l21.28 (1.03-39.04)5.67 (3.34-9.67)<0.05
HGB, g/l83 (36–150)128 (115–158)<0.05
PLT, 109/l32 (5–124)198 (119–279)<0.05
Myeloblasts in marrow, %50±22.55

[i] Data are presented as the number, median and interquartile range or mean ± SD. FAB, French-American-British; WBC, white blood cell; HGB, hemoglobin; PLT, platelet.

Identification of BMMSCs

BMMSCs were detached from the culture flasks using Accutase™ solution (EMD Millipore). Cells were washed twice with PBS, non-specific antigens were blocked with 5% goat serum (Beijing Biosynthesis Biotechnology Co., Ltd.) for 1 h at room temperature, and then incubated with anti-CD14-FITC (cat. no. 561712; BD Pharmingen; BD Biosciences), anti-CD34-FITC (cat. no. 560942; BD Pharmingen; BD Biosciences), anti-CD90-APC (cat. no. 561971; BD Pharmingen; BD Biosciences), anti-CD105-PE (cat. no. 560839; BD Pharmingen; BD Biosciences) and anti-CD45-PE-Cy7 (cat. no. 560915; BD Pharmingen; BD Biosciences) antibodies for 15 min at 4°C according to the manufacturer's instructions. Mouse IgG1K (cat. no. 562438; BD Pharmingen; BD Biosciences; 1:500) was incubated with cells at 4°C as an isotype control. Cells were analyzed via flow cytometry (Gallios; Beckman Coulter, Inc.) and the FlowJo software program (FlowJo7.6; FlowJo LLC).

Detection of FAPα expression and myeloblasts in marrow via flow cytometry

BMMSCs were detached from culture flasks using Accutase™ solution, washed twice with PBS and non-specific antigens were blocked with 5% goat serum (Beijing Biosynthesis Biotechnology Co., Ltd.) for 1 h at room temperature. Then, cells were incubated with mouse anti-FAPα (cat. no. sc-100528; Santa Cruz Biotechnology, Inc.; 1:500) for 2 h at 4°C. Subsequently, cells were washed twice with PBS and incubated with secondary antibody Alexa Fluor 488-labeled goat anti-mouse IgG (cat. no. A11001; Invitrogen; Thermo Fisher Scientific, Inc.; 1:500) for 30 min at 4°C. Cells were analyzed via flow cytometry (Gallios; Beckman Coulter, Inc.) and the FlowJo7.6 software program (FlowJo LLC). The steps to detect the myeloblasts in marrow were as follows: Leukemia-related phenotype was assess using antibodies (CD7-FITC, cat. no. 561933; CD117-PE, cat. no. 562407; CD19-APC, cat. no. 560727; HLA-DR-PE, cat. no. 560651; CD15-FITC, cat. no. 560997; CD34-APC,cat. no. 560940; CD56-FITC, cat. no. 562794; CD13-PE, cat. no. 560998; CD11b-APC,cat. no. 561690; CD64-PE, cat. no. 561926; CD14-FITC, cat. no. 561712; CD33-APC, cat. no. 561817; cCD22-PE, cat. no. 563941; cCD3-APC, cat. no. 561800; BD Pharmingen; BD Biosciences; 1:500. MPO-FITC, cat. no. 130-107-177; Miltenyi Biotec; 1:500.), followed by the addition of 50-100 µl bone marrow. This was mixed well and incubated in dark at room temperature for 15-20 min. Then, 1 ml hemolysin was added and incubated in dark for 12 min. Centrifugation with 200 × g at room temperature for 5 min was conducted, the supernatant was removed and 300 µl PBS was added for suspension. Finally, flow cytometry (Gallios; Beckman Coulter, Inc.) was used for detection.

Immunofluorescence

Before use in experiments, BMMSCs were seeded (60%) in 6-well plates and cultured for 24 h. Then, cells were washed three times with PBS for 5 min each and fixed in 4% paraformaldehyde for 30 min at room temperature. Cells were washed three times with PBS and permeabilized within 0.3% Triton X-100 for 10 min. After being washed three times with PBS, non-specific antigens were blocked with 5% goat serum (Beijing Biosynthesis Biotechnology Co., Ltd.) for 1 h at room temperature. After being washed three times with PBS, cells were and incubated with anti-FAPα (cat. no. sc-100528; Santa Cruz Biotechnology, Inc.; 1:500) at 4°C overnight. Then, cells were incubated in the dark with Alexa Fluor 488-labeled goat anti-mouse IgG (cat. no. A11001; Invitrogen; Thermo Fisher Scientific, Inc.; 1:500) at 4°C for 30 min. After being washed three times with PBS, cells were incubated with DAPI (1:500) for 5 min at room temperature in the dark. Then, cells were visualized using a fluorescence microscope with ×10 and ×40 magnification and images were collected.

Immunohistochemistry

A small piece of cylindrical bone marrow tissue, 0.5-1 cm long, was collected from the puncture needle. After sampling, tissues were immediately placed into 10% neutral buffered formalin solution and fixed at room temperature for 6-24 h. Then, 4% bone marrow decalcification solution (Vignes; http://www.wexisgp.com/) was used for 2 h at 37°C. The slices were prepared with a thickness of 3 µm and baked at 68°C for 1 h. The antigen was dewaxed with xylene and then repaired with citric acid antigen repair solution (cat. no. G1202; Wuhan Servicebio Technology Co., Ltd.). The slices were placed into 3% hydrogen peroxide solution (cat. no. G0115; Wuhan Servicebio Technology Co., Ltd.), incubated at room temperature and protected from light for 25 min to block endogenous peroxidase. Then, 3% BSA (cat. no. G5001, Wuhan Servicebio Technology Co., Ltd.) was added to evenly cover the tissue and slices were sealed at room temperature for 30 min. Slices were incubated overnight with anti-FAPα antibody (cat. no. ab53066; Abcam; 1:100) at 4°C. Then, slices were incubated with a secondary goat anti-rabbit antibody (cat. no. GB23303; Wuhan Servicebio Technology Co., Ltd.; 1:200) at room temperature for 50 min. After DAB (cat. no. G1211; Wuhan Servicebio Technology Co., Ltd.) color development, re-dyeing nucleus with hematoxylin for 3 min at room temperature, dehydration and sealing with neutral gum, images were captured using Nikon E100 light microscope with ×200 and ×400 magnification and analyzed using Image-pro plus 6.0 (Media Cybernetios, Inc; Version 6.0.0.260 for windows 2000/XP professional; serial no. 41M60032-00032) [optical density (OD)/area].

Reverse transcription-quantitative (RT-q)PCR

Total RNA was extracted from BMMSCs using RNAiso™ Plus (Takara Bio, Inc.) according to the manufacturer's instructions. cDNA was generated from 1 µg total RNA using Prime Script™ RT reagent kit (Perfect Real Time; Takara Bio, Inc.) with DNA Eraser according to the manufacturer's instructions. Duration of RT was 15 min at 37°C. RT-qPCR was performed using SYBR® Premix Ex Taq™ II (Tli RNaseH Plus; Takara Bio, Inc.) following the manufacturers' instructions on an iQ5 Multicolor Real-Time PCR Detection System (Bio-Rad Laboratories, Inc.). GAPDH served as an endogenous control. The primer sequences for FAPα and GAPDH amplification were as follows: FAPα forward, 5′-GTATTTGGAGTTGCCACCTCTG-3′ and reverse, 5′-GAAGGGCGTAAGACAATGCAC-3′; and GAPDH forward, 5′-AAGGTGAAGGTCGGAGTCAAC-3′ and reverse, 5′-GGGGTCATTGATGGCAACAATA-3′. The thermocycling conditions for FAPα and GAPDH amplification were as follows: Initial denaturation at 95°C for 30 sec, followed by 40 cycles of 95°C for 5 sec and 60°C for 30 sec. The primer specificity was verified using the melt curve. The relative quantification of FAPα in BMMSCs was calculated using the 2−ΔΔCq method (27).

Transfection and cell apoptosis analysis via flow cytometry

The day before transfection, BMMSCs were seeded (60%) in 12-orifice plates. When the cells reached ~90% confluency, they were transfected with FAPα small interfering (si)RNA (forward, 5′-CGCCCUUCAAGAGUUCAUATT-3′ and reverse, 5′-UAUGAACUCUUGAAGGGCGTT-3′), FAPα-Mock (only lipo2000 and no siRNA sequence) and NC siRNA (forward, 5′-UUCUCCGAACGUGUCACGUTT-3′ and reverse, 5′-ACGUGACACGUUCGGAGAATT-3′) at room temperature using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.). The mass and concentration of transfected siRNA was 5 µl and 100 pmol, respectively. After 6 h, the medium was removed, and Kasumi-1 cells were added and co-cultured with transfected BMMSCs. The co-culture ratio of AML cells and BMMSCs was 1-2:1.

A total of 100 µM Cytosine arabinoside (Ara-C; Pfizer, Inc.) was added to the well to induce apoptosis in Kasumi-1 cells (28) for 24 h at room temperature. After 24 h, co-cultured Kasumi-1 cells were isolated from the monolayer of BMMSCs, and gently transferred into the monolayer with medium. Next, cells were stained with FITC-conjugated Annexin V and PE-conjugated PI at 4°C for 30 min following the Annexin V FITC Apop Dtec Kit I (BD Biosciences) manufacturer's instructions. Annexin V-FITC and PI-PE positive cells were considered apoptotic cells. The interfering effect was determined via RT-qPCR and flow cytometry (Gallios; Beckman Coulter, Inc.) and the FlowJo7.6 software program (FlowJoLLC).

Western blot analysis

After transfection of BMMSCs with FAPα-siRNA, BMMSCs (2×105) and Kasumi-1 cells (5×105) were cocultured in 6-well plates. After 48 h, Kasumi-1 cells were collected and washed twice with PBS. Total cell lysates were obtained using lysis buffer (Beyotime Institute of Biotechnology) supplemented with 1 mM PMSF. The protein concentration was determined using the BCA (Sangon Biotech Co., Ltd.) method according to the manufacturer's guidelines. An equal amount of protein (20-40 g) was separated via 10-12% SDS-PAGE and transferred to PVDF membranes. Next, membranes were blocked with 5% non-fat milk for 2 h at room temperature. PVDF membranes were incubated with anti-actin and anti-β-catenin antibodies (cat. no. 2698S; Cell Signaling Technology, Inc.; 1:500) overnight at 4°C. Subsequently, membranes were washed and incubated with a horseradish peroxidase-conjugated antibody (cat. no. 7074S; Cell Signaling Technology, Inc.; 1:5,000) in 0.2% TBS-Tween at room temperature for 2 h. After being washed, protein bands were visualized using an ECL detection kit (Biological Industries). Semi-quantification of the western blotting was performed using the Bio-Rad imaging system and ImageJ software (29) (Life-Line Fiji versions java8; http://imagej.net/Fiji/Downloads) to detect protein expression.

Statistical analysis

All the experiments were repeated three times. All analyses were performed using SPSS 22.0 software (IBM Corp.). The χ2 test was used for categorical data. Measurement data were presented as mean ± SD. The unpaired Students t-test was used for two independent groups. Multiple comparisons were performed using one-way ANOVA (Bonferroni) statistical analysis. Pearson correlation test was used for correlation analysis. P<0.05 was considered to indicate a statistically significant difference.

Results

Identification of BMMSCs

The experimental results demonstrated that cultured BMMSCs adhered to the flasks and had a fibroblast-like morphology (Fig. 1A). BMMSCs of passage 2 had a low expression of CD14, CD34 and CD45, and a high expression of CD90 and CD105 (n=3). Analysis of these CD molecular markers on BMMSCs via flow cytometry is presented in Fig. 1B.

Expression of FAPα in BMMSCs of newly diagnosed patients with AML and healthy donors

Significant differences in the expression of FAPα between newly diagnosed patients with AML and healthy donors were identified via flow cytometry (70.92±4.38 vs. 36.74±10.37%, respectively; P=0.0072; Fig. 2B and C), RT-qPCR (24.75±2.75 vs. 9.77±1.94, respectively; P=0.0001; Fig. 2D) and immunofluorescence (Fig. 2A).

Expression of FAPα in BM biopsy samples of newly diagnosed patients with AML and healthy donors

The expression of FAPα in BM biopsy samples between newly diagnosed patients with AML and healthy donors was examined via immunohistochemistry. FAPα expression was observed in the BM mesenchymal matrix as indicated by the positively stained yellow area (Fig. 3A and B). Image-pro plus 6.0 software was used to estimate the percentage of the stained area, represented as OD/area. The OD/area value was higher in AML BM compared with BM from healthy donors (OD/area, 11.88±4.55 vs. 5.16±3.67; P<0.0001; n=15; Fig. 3C).

Correlation of FAPα expression in BM with clinical parameters and survival time of newly diagnosed patients with AML

The correlation between FAPα expression in BM and clinically parameters of newly diagnosed AML patients was analyzed, and the results demonstrated that the OD/area value of FAPα was positively correlated with the proportion of myeloblasts in marrow detected via flow cytometry (r=0.878; P<0.0001; Fig. 4A). No significant correlation was observed between the OD/area value of FAPα and leukocytes (r=0.444; P=0.0976; Fig. 4B), hemoglobin (r=0.331; P=0.2287; Fig. 4C) or platelets (r=0.278; P=0.3153; Fig. 4D). Representative flow scatter plots of myeloblasts in marrow is presented in Fig. 4E. The OD/area value of FAPα was significantly and negatively correlated with survival in eight patients who did not undergo chemotherapy (r=0.815; P=0.0137; Fig. 4F).

BMMSCs protects AML cells from Ara-C-induced apoptosis, which in part is contributed by FAPα

RT-qPCR was used to verify successful transfection prior to detecting apoptosis. The relative expression of FAPα mRNA in the FAPα-siRNA group was significantly lower compared with that in negative control (NC)-siRNA group (0.11±0.01 vs. 0.84±0.08; P=0.0001; Fig. 5A). Then, flow cytometry results demonstrated that BMMSCs protected Kasumi-1 cells from apoptosis, even in the presence of Ara-C (100 µM), and knockdown of FAPα with siRNA decreased the protective effect of BMMSCs (Fig. 5C). The proportion of apoptotic cells in the MSC-Mock group was significantly lower compared with that in the Ara-C group (12.96±0.95 vs. 25.66±1.54%; P<0.001; n=9; Fig. 5B). Knockdown of FAPα using siRNA decreased the protective effect. The proportion of apoptotic cells in the FAPα-siRNA group was significantly increased compared with the proportion in the NC-siRNA group (22.69±1.99 vs. 13.29±1.10%; P<0.001; n=9; Fig. 5B). The proportion of apoptotic cells in FAPα-siRNA group was also lower compared with that in the Ara-C group (22.69±1.99 vs. 25.66±1.54%; P=0.001; Fig. 5B).

Possible mechanism of FAPα protecting AML cells from apoptosis

The results demonstrated that Kasumi-1 cells expressed β-catenin, which could be inhibited by Ara-C. The expression of β-catenin in Kasumi-1 cells was significantly increased when Kasumi-1 cells were cocultured with BMMSCs and NC-siRNA for 48 h, and even in the presence of Ara-C the expression of β-catenin is activated. Compared with NC-siRNA, knockdown of FAPα using siRNA significantly suppressed the expression of β-catenin (Fig. 6A). The experiment was performed in triplicate, the relative expression of β-catenin was calculated and statistical analyses were performed. In the absence of BMMSCs, the expression of β-catenin was decreased from 113.20±2.77 to 44.02±0.06 in groups with and without Ara-C, respectively (P<0.0001; Fig. 6B). In addition, in the presence of Ara-C and BMMSCs, the expression of β-catenin in the NC-siRNA group and FAPα-siRNA group was 116.84±0.40 and 82.11±1.26, respectively (P<0.0001; Fig. 6B).

Discussion

In 1991, Caplan's pioneering work reported the existence of BMMSCs (30). Typically, MSCs are isolated by their ability to adhere to the plastic surface of a culture dish, which demonstrates the ability of the cells to expand in culture plates, and MSCs have specific markers of immunological characteristics (31). The International Society for Cellular Therapy suggests three minimal criteria for the characterization of MSCs: i) Adherence to plastic; ii) expression of markers associated with MCS (such as CD73, CD90 and CD105), and the lack of hematopoietic-related cell expression (including CD34, CD45, CD11b or CD14, CD19 or CD79 and human leukocyte antigen DR); and iii) potential tri-lineage differentiation into adipocytes, osteoblasts and chondrocytes (30). In the present study, cultured BMMSCs adhered to culture flasks and were fibroblast-like in morphology. In addition, BMMSCs of passage 2 had a low expression of CD14, CD34 and CD45, and high expression of CD90 and CD105. Collectively, the current findings met the minimal criteria for defining MSCs.

FAPα (also known as seprase) is a serine oligopeptidase that was originally identified in 1986 as an inducible cell surface glycoprotein F19 (32), and was renamed FAP in 1994 based upon its abundance in activated fibroblasts (33). Human FAP is comprised of M(r) 95,000 (p95, FAPα) and M(r) 105,000 (p105, FAPβ) subunits, which are conjugated by non-covalent, non-disulfide bonds (34). Immunoblot experiments have shown that FAPα, but not FAPβ, carries an epitope that is bound by monoclonal antibody F19 (35), and so the F19 surface antigen was renamed to FAPα.

Most healthy adult tissues have little or no detectable FAPα expression (36). However, FAPα expression is highly upregulated during active tissue remodeling of fibroblasts, including wound healing, fibrosis and at cancer sites (37). FAPα is also expressed in BMMSCs, and promotes the motility of human BMMSCs (38). In the context of cancer, FAPα has been widely considered as a marker of CAFs, which have multiple pro-tumorigenic functions (38). Moreover, FAPα itself has been reported to exert pro-tumorigenic activity, both via enzymatic and non-enzymatic mechanisms (36,39). Previous studies have revealed that tumor cell conditioned medium can transform BMMSCs into CAFs, thereby promoting tumor progression (40). Moreover, tumor cells can reprogram BMMSCs and promote the evolution of BMMSCs into CAFs, further facilitating the development of hematological malignancies (41). Therefore, it was hypothesized that the AML microenvironment may instruct BMMSCs to become CAFs, to further increase the expression of FAPα in BMMSCs compared with BMMSCs in a healthy environment.

The present study first evaluated the expression of FAPα on BMMSCs between newly diagnosed patients with AML and healthy donors using RT-qPCR and flow cytometry. The experimental results demonstrated that the expression of FAPα in newly diagnosed patients with AML was significantly higher compared with healthy donors. However, the specific origin of FAPα within the BM remains to be elucidated. To further confirm the expression of FAPα in BM, the expression of FAPα in the BM mesenchymal matrix was positively stained yellow. It was identified that the OD/area value of FAPα in newly diagnosed patients with AML was significantly higher compared with that of healthy donors. Previous studies have reported that FAPα expression is associated with worse prognosis in solid tumors (42). However, the relationship between FAPα expression and the prognosis in hematologic tumors remains unknown. The present study demonstrated that the OD/area value of FAPα was positively correlated with the proportion of myeloblasts in BM as detected via flow cytometry, and was significantly negatively associated with survival time in eight patients who did not undergo chemotherapy due to the costs and risks. Collectively, these results suggested that FAPα serves an important role in the AML BM microenvironment, and may be involved in the development of AML. Thus, increased expression of FAPα in BM may be a poor prognostic factor in AML.

The interaction between AML cells and BMMSCs, either via direct contact or secreted cytokines, can promote AML progression, survival, growth, chemotherapy resistance and angiogenesis (4346). However, little is known regarding the function of FAPα expression in BMMSCs and how it affects the features of AML cells. In the current study, flow cytometry results demonstrated that BMMSCs protected Ara-C (100 µM)-induced Kasumi-1 cell apoptosis in a cocultured system. Furthermore, knockdown of FAPα with siRNA decreased this protective effect. However, the proportion of apoptotic cells in the FAPα-siRNA group was lower compared with that in Ara-Cgroup. Thus, these results suggested that BMMSCs in the BM microenvironment of patients with AML were instructed to become CAFs to protect AML cells from apoptosis, which, in part was contributed by FAPα. However, other factors may be involved in the protective effect of BMMSCs on leukemia cell apoptosis.

Previous studies have revealed that, via a variety of mechanisms, BMMSCs inhibit AML cell apoptosis and promote their survival, proliferation and chemotherapy resistance (4651). In addition, it has been reported that Wnt ligands are the key drivers of most types of tissue stem cells in adult mammals, and mutated Wnt pathway components are causative in the progression of multiple growth-related pathologies and cancer (52). For instance, the Wnt/β-catenin pathway is required for the development of leukemia stem cells in AML (53). However, the current understanding of the activation of the BMMSC-mediated Wnt/β-catenin signaling pathway is limited. Furthermore, the relationship between FAPα and Wnt/β-catenin for AML cellular features remains unknown. The present study demonstrated that Kasumi-1 cells expressed β-catenin, which could be inhibited by Ara-C. However, the expression of β-catenin in Kasumi-1 cells was significantly increased when Kasumi-1 cells were cocultured with BMMSCs, even in the presence of Ara-C. The data also indicated that knockdown of FAPα using siRNA significantly suppressed the expression of β-catenin. Thus, it was suggested FAPα may be involved in BMMSC-mediated apoptosis of AML cells induced by Ara-C, which may be achieved via the β-catenin pathway.

In conclusion, the present results indicated that FAPα may serve an important role in the AML BM microenvironment and that it may be involved in the development of AML. Moreover, it was suggested that increased expression of FAPα in BM may be a poor prognostic factor in AML. Further studies demonstrated that BMMSCs could protect AML cells from apoptosis, which in part was contributed by FAPα, and likely occurs via the β-catenin signaling pathway. However, whether the activation of β-catenin was dependent on direct contact with BMMSCs or secretory of cytokines requires further research. In addition, the quantity of the samples should be expanded to clarify the relationship between FAPα expression and the prognosis of AML, and additional studies should be performed using other AML cell lines to further verify the anti-apoptotic role of FAPα in AML cells.

Acknowledgements

Not applicable.

Funding

This work was supported by the National Natural Science Foundation of China (grant no. 81560030), the Natural Science Foundation of Jiangxi Province (grant no. 20192BAB205034) and the Science and Technology Plan of Health and Family Planning Commission Jiangxi Province (grant no. 20181097).

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

SM, FZ and GC designed the research and analyzed the data. SM, YZ and LY performed the cell function experiments. SM and YZ collected the BM liquid and biopsy samples, and reviewed the clinical features. SM prepared the images and drafted the original manuscript. FZ and GC reviewed and edited the manuscript. All authors read and approved the manuscript, and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

The study was approved by the Medical Research Ethics Committee of the Second Affiliated Hospital of Nanchang University. All patients and healthy donors provided written informed consent. The study in keeping with ethical guidelines of the Second Affiliated Hospital of Nanchang University and the Declaration of Helsinki.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Dores GM, Devesa SS, Curtis RE, Linet MS and Morton LM: Acute leukemia incidence and patient survival among children and adults in the United States, 2001-2007. Blood. 119:34–43. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Montesinos P, Bergua J, Infante J, Esteve J, Guimaraes JE, Sierra J and Sanz MÁ: Update on management and progress of novel therapeutics for R/R AML: An Iberian expert panel consensus. Ann Hematol. 98:2467–2483. 2019. View Article : Google Scholar : PubMed/NCBI

3 

Liao Z, Tan ZW, Zhu P and Tan NS: Cancer-associated fibroblasts in tumor microenvironment - Accomplices in tumor malignancy. Cell Immunol. 343:1037292019. View Article : Google Scholar : PubMed/NCBI

4 

Shafat MS, Gnaneswaran B, Bowles KM and Rushworth SA: The bone marrow microenvironment - Home of the leukemic blasts. Blood Rev. 31:277–286. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Geyh S, Rodríguez-Paredes M, Jäger P, Khandanpour C, Cadeddu RP, Gutekunst J, Wilk CM, Fenk R, Zilkens C, Hermsen D, et al: Functional inhibition of mesenchymal stromal cells in acute myeloid leukemia. Leukemia. 30:683–691. 2016. View Article : Google Scholar : PubMed/NCBI

6 

von der Heide EK, Neumann M, Vosberg S, James AR, Schroeder MP, Ortiz-Tanchez J, Isaakidis K, Schlee C, Luther M, Jöhrens K, et al: Molecular alterations in bone marrow mesenchymal stromal cells derived from acute myeloid leukemia patients. Leukemia. 31:1069–1078. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Augsten M: Cancer-associated fibroblasts as another polarized cell type of the tumor microenvironment. Front Oncol. 4:622014. View Article : Google Scholar : PubMed/NCBI

8 

Chaturvedi P, Gilkes DM, Wong CC, Luo W, Zhang H, Wei H, Takano N, Schito L, Levchenko A, Semenza GL and Kshitiz: Hypoxia-inducible factor-dependent breast cancer-mesenchymal stem cell bidirectional signaling promotes metastasis. J Clin Invest. 123:189–205. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Raz Y, Cohen N, Shani O, Bell RE, Novitskiy SV, Abramovitz L, Levy C, Milyavsky M, Leider-Trejo L, Moses HL, et al: Bone marrow-derived fibroblasts are a functionally distinct stromal cell population in breast cancer. J Exp Med. 215:3075–3093. 2018. View Article : Google Scholar : PubMed/NCBI

10 

Tao L, Huang G, Song H, Chen Y and Chen L: Cancer associated fibroblasts: An essential role in the tumor microenvironment. Oncol Lett. 14:2611–2620. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Zhou P, Xiao N and Wang J, Wang Z, Zheng S, Shan S and Wang J, Du J and Wang J: SMC1A recruits tumor-associated-fibroblasts (TAFs) and promotes colorectal cancer metastasis. Cancer Lett. 385:39–45. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Komohara Y and Takeya M: CAFs and TAMs: Maestros of the tumour microenvironment. J Pathol. 241:313–315. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Chen X and Song E: Turning foes to friends: Targeting cancer-associated fibroblasts. Nat Rev Drug Discov. 18:99–115. 2019. View Article : Google Scholar : PubMed/NCBI

14 

Kalluri R: The biology and function of fibroblasts in cancer. Nat Rev Cancer. 16:582–598. 2016. View Article : Google Scholar : PubMed/NCBI

15 

LeBleu VS and Kalluri R: A peek into cancer-associated fibroblasts: origins, functions and translational impact. Dis Model Mech. 11:dmm0294472018. View Article : Google Scholar : PubMed/NCBI

16 

Higashino N, Koma YI, Hosono M, Takase N, Okamoto M, Kodaira H, Nishio M, Shigeoka M, Kakeji Y and Yokozaki H: Fibroblast activation protein-positive fibroblasts promote tumor progression through secretion of CCL2 and interleukin-6 in esophageal squamous cell carcinoma. Lab Invest. 99:777–792. 2019. View Article : Google Scholar : PubMed/NCBI

17 

Wen X, He X, Jiao F, Wang C, Sun Y, Ren X and Li Q: Fibroblast activation protein-α-positive fibroblasts promote gastric cancer progression and resistance to immune checkpoint blockade. Oncol Res. 25:629–640. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Hu M, Qian C, Hu Z, Fei B and Zhou H: Biomarkers in tumor Microenvironment? Upregulation of fibroblast activation protein-α correlates with gastric cancer progression and poor prognosis. OMICS. 21:38–44. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Byrling J, Sasor A, Nilsson J, Said Hilmersson K, Andersson R and Andersson B: Expression of fibroblast activation protein and the clinicopathological relevance in distal cholangiocarcinoma. Scand J Gastroenterol. 55:82–89. 2020. View Article : Google Scholar : PubMed/NCBI

20 

Zhang L, Yang L, Xia ZW, Yang SC, Li WH, Liu B, Yu ZQ, Gong PF, Yang YL, Sun WZ, et al: The role of fibroblast activation protein in progression and development of osteosarcoma cells. Clin Exp Med. 20:121–130. 2020. View Article : Google Scholar : PubMed/NCBI

21 

Pleshkan VV, Alekseenko IV, Tyulkina DV, Kyzmich AI, Zinovyeva MV and Sverdlov ED: Fibroblast activation protein (FAP) as a possible target of an antitumor strategy. Mol Gen Mikrobiol Virusol. 34:90–97. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Li M, Li M, Yin T, Shi H, Wen Y, Zhang B, Chen M, Xu G, Ren K and Wei Y: Targeting of cancer associated fibroblasts enhances the efficacy of cancer chemotherapy by regulating the tumor microenvironment. Mol Med Rep. 13:2476–2484. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Raffaghello L, Vacca A, Pistoia V and Ribatti D: Cancer associated fibroblasts in hematological malignancies. Oncotarget. 6:2589–2603. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Zi FM, He JS, Li Y, Wu C, Wu WJ, Yang Y, Wang LJ, He DH, Yang L, Zhao Y, et al: Fibroblast activation protein protects bortezomib-induced apoptosis in multiple myeloma cells through β-catenin signaling pathway. Cancer Biol Ther. 15:1413–1422. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Pittenger MF: Mesenchymal stem cells from adult bone marrow. Mesenchymal Stem Cells. Springer; Basel: pp. 27–44. 2008, View Article : Google Scholar

26 

Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le Beau MM, Bloomfield CD, Cazzola M and Vardiman JW: The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 127:2391–2405. 2016. View Article : Google Scholar : PubMed/NCBI

27 

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. View Article : Google Scholar : PubMed/NCBI

28 

Yang Y, Mallampati S, Sun B, Zhang J, Kim SB, Lee JS, Gong Y, Cai Z and Sun X: Wnt pathway contributes to the protection by bone marrow stromal cells of acute lymphoblastic leukemia cells and is a potential therapeutic target. Cancer Lett. 333:9–17. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Abramoff MD, Magalhaes PJ and Ram SJ: Image processing with ImageJ. Biophoton Int. 11:36–42. 2004.

30 

Caplan AI: Mesenchymal stem cells. J Orthop Res. 9:641–650. 1991. View Article : Google Scholar : PubMed/NCBI

31 

Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, Deans R, Keating A, Prockop Dj and Horwitz E: Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 8:315–317. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Rettig WJ, Chesa PG, Beresford HR, Feickert HJ, Jennings MT, Cohen J, Oettgen HF and Old LJ: Differential expression of cell surface antigens and glial fibrillary acidic protein in human astrocytoma subsets. Cancer Res. 46:6406–6412. 1986.PubMed/NCBI

33 

Rettig WJ, Su SL, Fortunato SR, Scanlan MJ, Raj BK, Garin-Chesa P, Healey JH and Old LJ: Fibroblast activation protein: Purification, epitope mapping and induction by growth factors. Int J Cancer. 58:385–392. 1994. View Article : Google Scholar : PubMed/NCBI

34 

Scanlan MJ, Raj BK, Calvo B, Garin-Chesa P, Sanz-Moncasi MP, Healey JH, Old LJ and Rettig WJ: Molecular cloning of fibroblast activation protein alpha, a member of the serine protease family selectively expressed in stromal fibroblasts of epithelial cancers. Proc Natl Acad Sci USA. 91:5657–5661. 1994. View Article : Google Scholar : PubMed/NCBI

35 

Rettig WJ, Garin-Chesa P, Healey JH, Su SL, Ozer HL, Schwab M, Albino AP and Old LJ: Regulation and heteromeric structure of the fibroblast activation protein in normal and transformed cells of mesenchymal and neuroectodermal origin. Cancer Res. 53:3327–3335. 1993.PubMed/NCBI

36 

Keane FM, Yao TW, Seelk S, Gall MG, Chowdhury S, Poplawski SE, Lai JH, Li Y, Wu W, Farrell P, et al: Quantitation of fibroblast activation protein (FAP)-specific protease activity in mouse, baboon and human fluids and organs. FEBS Open Bio. 4:43–54. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Jacob M, Chang L and Puré E: Fibroblast activation protein in remodeling tissues. Curr Mol Med. 12:1220–1243. 2012. View Article : Google Scholar : PubMed/NCBI

38 

Chung KM, Hsu SC, Chu YR, Lin MY, Jiaang WT, Chen RH and Chen X: Fibroblast activation protein (FAP) is essential for the migration of bone marrow mesenchymal stem cells through RhoA activation. PLoS One. 9:e887722014. View Article : Google Scholar : PubMed/NCBI

39 

Hamson EJ, Keane FM, Tholen S, Schilling O and Gorrell MD: Understanding fibroblast activation protein (FAP): Substrates, activities, expression and targeting for cancer therapy. Proteomics Clin Appl. 8:454–463. 2014. View Article : Google Scholar : PubMed/NCBI

40 

Zi F, He J, He D, Li Y, Yang L and Cai Z: Fibroblast activation protein α in tumor microenvironment: Recent progression and implications (Review). Mol Med Rep. 11:3203–3211. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Tian K, Yang S, Ren Q and Han Z, Lu S, Ma F, Zhang L and Han Z: p38 MAPK contributes to the growth inhibition of leukemic tumor cells mediated by human umbilical cord mesenchymal stem cells. Cell Physiol Biochem. 26:799–808. 2010. View Article : Google Scholar : PubMed/NCBI

42 

Liu F, Qi L, Liu B, Liu J, Zhang H, Che D, Cao J, Shen J, Geng J, Bi Y, et al: Fibroblast activation protein overexpression and clinical implications in solid tumors: A meta-analysis. PLoS One. 10:e01166832015. View Article : Google Scholar : PubMed/NCBI

43 

Traer E, Martinez J, Javidi-Sharifi N, Agarwal A, Dunlap J, English I, Kovacsovics T, Tyner JW, Wong M and Druker BJ: FGF2 from marrow microenvironment promotes resistance to FLT3 inhibitors in acute myeloid leukemia. Cancer Res. 76:6471–6482. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Fathi E, Sanaat Z and Farahzadi R: Mesenchymal stem cells in acute myeloid leukemia: A focus on mechanisms involved and therapeutic concepts. Blood Res. 54:165–174. 2019. View Article : Google Scholar : PubMed/NCBI

45 

Brenner AK, Nepstad I and Bruserud Ø: Mesenchymal stem cells support survival and proliferation of primary human acute myeloid leukemia cells through heterogeneous molecular mechanisms. Front Immunol. 8:1062017. View Article : Google Scholar : PubMed/NCBI

46 

Li Q, Pang Y, Liu T, Tang Y, Xie J, Zhang B and Chen H: Effects of human umbilical cord-derived mesenchymal stem cells on hematologic malignancies. Oncol Lett. 15:6982–6990. 2018.PubMed/NCBI

47 

Yang X, Sexauer A and Levis M: Bone marrow stroma-mediated resistance to FLT3 inhibitors in FLT3-ITD AML is mediated by persistent activation of extracellular regulated kinase. Br J Haematol. 164:61–72. 2014. View Article : Google Scholar : PubMed/NCBI

48 

Tian C, Zheng G, Zhuang H, Li X, Hu D, Zhu L, Wang T, You MJ and Zhang Y: MicroRNA-494 activation suppresses bone marrow stromal cell-mediated drug resistance in acute myeloid leukemia cells. J Cell Physiol. 232:1387–1395. 2017. View Article : Google Scholar : PubMed/NCBI

49 

Guo F, Wang Y, Liu J, Mok SC, Xue F and Zhang W: CXCL12/CXCR4: A symbiotic bridge linking cancer cells and their stromal neighbors in oncogenic communication networks. Oncogene. 35:816–826. 2016. View Article : Google Scholar : PubMed/NCBI

50 

Malfuson JV, Boutin L, Clay D, Thépenier C, Desterke C, Torossian F, Guerton B, Anginot A, de Revel T, Lataillade JJ, et al: SP/drug efflux functionality of hematopoietic progenitors is controlled by mesenchymal niche through VLA-4/CD44 axis. Leukemia. 28:853–864. 2014. View Article : Google Scholar : PubMed/NCBI

51 

Battula VL, Benito J, Somanchi AG, Duvvuri S, Hodgson L, Chen Y, Ma W, Davis RE, McNiece I, Shpall EJ, et al: Acute myeloid leukemia cells acquire stem cell features in the bone marrow microenvironment. Clin Lymphoma Myeloma Leuk. 14:S119–S120. 2014. View Article : Google Scholar

52 

Nusse R and Clevers H: Wnt/β-catenin signaling, disease, and emerging therapeutic modalities. Cell. 169:985–999. 2017. View Article : Google Scholar : PubMed/NCBI

53 

Wang Y, Krivtsov AV, Sinha AU, North TE, Goessling W, Feng Z, Zon LI and Armstrong SA: The Wnt/beta-catenin pathway is required for the development of leukemia stem cells in AML. Science. 327:1650–1653. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2021
Volume 45 Issue 2

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Mei S, Zhang Y, Yu L, Chen G and Zi F: Expression and role of fibroblast activation protein α in acute myeloid leukemia. Oncol Rep 45: 641-651, 2021
APA
Mei, S., Zhang, Y., Yu, L., Chen, G., & Zi, F. (2021). Expression and role of fibroblast activation protein α in acute myeloid leukemia. Oncology Reports, 45, 641-651. https://doi.org/10.3892/or.2020.7874
MLA
Mei, S., Zhang, Y., Yu, L., Chen, G., Zi, F."Expression and role of fibroblast activation protein α in acute myeloid leukemia". Oncology Reports 45.2 (2021): 641-651.
Chicago
Mei, S., Zhang, Y., Yu, L., Chen, G., Zi, F."Expression and role of fibroblast activation protein α in acute myeloid leukemia". Oncology Reports 45, no. 2 (2021): 641-651. https://doi.org/10.3892/or.2020.7874