cells - Bosnian Journal of Basic Medical Sciences

21 downloads 0 Views 2MB Size Report
Sep 15, 2017 - Medical Biology Department, Ege University Medicine Faculty, Izmir, Turkey. ABSTRACT .... WST-1 assay (Roche Applied Science, Mannheim,.
BOSNIAN JOURNAL OF BASIC MEDICAL SCIENCES

RESEARCH ARTICLE

WWW.BJBMS.ORG

Matrine induced G0/G1 arrest and apoptosis in human acute T-cell lymphoblastic leukemia (T-ALL) cells Aslı Tetik Vardarlı*, Zekeriya Düzgün, Ceren Erdem, Burçin Tezcanli Kaymaz, Zuhal Eroglu, Vildan Bozok Çetintas Medical Biology Department, Ege University Medicine Faculty, Izmir, Turkey

ABSTRACT Matrine, a natural product extracted from the root of Sophora flavescens, is a promising alternative drug in different types of cancer. Here, we aimed to investigate the therapeutic effects and underlying molecular mechanisms of matrine on human acute lymphoblastic leukemia (ALL) cell line, CCRF-CEM. Cell viability and IC50 values were determined by WST-1 cell cytotoxicity assay. Cell cycle distribution and apoptosis rates were analyzed by flow cytometry. Expression patterns of 44 selected miRNAs and 44 RNAs were analyzed by quantitative reverse transcription polymerase chain reaction (qRT-PCR) using the Applied Biosystems 7500 Fast Real-Time PCR System. Matrine inhibited cell viability and induced apoptosis of CCRF-CEM cells in a dose-dependent manner. Cell cycle analysis demonstrated that matrine-treated CCRF-CEM cells significantly accumulated in the G0/G1 phase compared with the untreated control cells. hsa-miR-376b-3p (-37.09 fold, p = 0.008) and hsa-miR106b-3p (-16.67 fold, p = 0.028) expressions were decreased, whereas IL6 (95.47 fold, p = 0.000011) and CDKN1A (140.03 fold, p = 0.000159) expressions were increased after matrine treatment. Our results suggest that the downregulation of hsa-miR-106b-3p leads to the upregulation of target p21 gene, CDKN1A, and plays a critical role in the cell cycle progression by arresting matrine-treated cells in the G0/G1 phase. KEY WORDS: Sophora flavescens; matrine; acute lymphoblastic leukemia; T-ALL; CCRF-CEM; miR-376b; miR-106b; p21; CDKN1A; cell cycle; G0/G1 arrest; apoptosis; autophagy DOI: http://dx.doi.org/10.17305/bjbms.2017.2457

Bosn J Basic Med Sci. xxxx;xx(x):1-9. © 2018 ABMSFBIH

INTRODUCTION

oncogenes, deletions, somatic gene mutations, as well as by alterations of signaling pathways and dysregulation of microRNAs (miRNAs) [2]. miRNAs are small noncoding RNAs about 22 nucleotides long which downregulate gene expression at the post-transcriptional level; they can inhibit translation or induce mRNA degradation by binding to the 3’-UTR region of the targeted mRNA. miRNAs regulate different cellular processes, including cell proliferation, transformation, and death; in addition, they control cell development and metabolism [3]. Consequently, miRNAs play an important role in the regulation of many diseases such as viral infections, cardiovascular diseases, pulmonary artery hypertension, and cancer [4]. Furthermore, cell cycle regulators have a crucial role in the development and progression of cancer and are closely linked to cell death and proliferation [5]. The mechanisms of cell death are evolutionarily conserved processes important for vital functions in all multicellular organisms, and are classified into three types: apoptosis, necrosis, and autophagy [6-8]. Apoptosis and autophagy are the most important for maintaining tissue homeostasis, and their dysregulation can lead to cancer development. The two cell death processes can be regulated by the same or different signaling pathways. Understanding these pathways will help

T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive form of leukemia characterized by an increased proliferation of lymphoblasts. T-ALL constitutes 10-15% of childhood ALLs and 25% of adult ALLs. An effective chemotherapy combination for T-ALL has not been developed yet, and firstline therapy fails in 25% of pediatric and more than 50% of adult cases [1,2]. Recent studies have demonstrated that new treatments targeting specific genes or molecular pathways in cancer patients are more efficient and less toxic. Thus, targeted treatment and the determination of related oncogenes have a priority in terms of patient survival and occurrence of toxicity [2]. Therefore, new treatment options should be developed for these cases by determining new molecular targets. T-ALL is characterized by different gene and chromosome aberrations, including chromosomal translocations in the T-cell receptor (TCR) genes, abnormal expression of *Corresponding author: Aslı Tetik Vardarlı, Medical Biology Department, Ege University Medicine Faculty, Izmir, Turkey. Phone: +905067158158; Fax: +902323420542. E-mail: [email protected], [email protected] Submitted: 15 September 2017/Accepted: 09 October 2017

1

Aslı Tetik Vardarlı, et al.: Molecular mechanisms of matrine in T-ALL cells

MATERIALS AND METHODS

develop new strategies for inhibiting the proliferation of cancer cells. The progression through the cell cycle is controlled by cell cycle checkpoints, i.e.,  G1, S, G2 and M, and depends on the action of cyclins, cyclin-dependent kinases (CDKs) and cyclin-dependent kinase inhibitors (CDKIs) [5]. The proliferation or death of cells can be induced at any stage of cell cycle. Recent studies have demonstrated that several miRNAs (let‑7, miR-15a, miR16-1, miR-34, miR-106, miR-221, miR-222, and miR-124a clusters) regulate gene expression at different stages of cell cycle progression [9-14]. For example, dysregulation of miR34 causes a decreased expression of E2F family members and arrests cells in the G0/G1 phase [15]. Overexpression of miR-221 and miR-222 inhibit the expression of P27Kip1 protein of CDKI family and promotes cell proliferation [13,16]. Furthermore, miR-16 family members contribute to apoptosis during cell cycle progression by silencing BCL2 gene expression [11,17]. The expression of miR-106 has been studied in renal cell carcinoma (RCC) [18], osteosarcoma [19], and gastric [20], lung [21], and colorectal [22] cancers. Moreover, the diagnostic and prognostic significance of miR-106 has also been explored in several cancer types. It was reported that miR-106 is one of four miRNAs whose serum level is closely associated with the overall survival of gastric cancer patients who received adjuvant chemotherapy [20]. In another study, serum levels of miR-106a were higher in RCC patients compared to controls, but a decrease in miR-106a levels in the patients was observed one month after surgery [18]. High-mobility group AT-hook-2 (HMGA2), E1A binding protein p300 (EP300), DLC1 Rho GTPase activating protein (DLC1), and RE-1 silencing transcription factor (REST) have been reported as target genes of miR-106 [19,22-24]. CDKI 1 (also known as p21 and CDKN1A) is a cell cycle inhibitor protein that controls cell cycle arrest, apoptosis and DNA repair, and it can be regulated by miRNAs, RNA-binding proteins, phosphorylation and ubiquitination [25]. Numerous studies have demonstrated that dysregulation of miR-17, miR20, and miR-93 leads to aberrant p21 expression, which causes enhanced cell proliferation and inhibits progression through the G0/G1 and G1/S phases [12,25-28]. Matrine (dodecahydro-3a, 7a-diaza-benzo[de]anthracen8-one; molecular formula: C15H24N2O) is an alkaloid compound obtained from Sophora flavescens plant, that acts as a κ-opioid and μ-opioid receptor agonist. Although in vivo and in vitro studies have shown that matrine has strong antitumor effects, such as inhibiting cell proliferation and inducing apoptosis, the molecular mechanisms by which matrine exerts its effects have not been fully explained [29-33]. In this study, we aimed to investigate the effects and underlying molecular mechanisms of matrine on human T-ALL cell line, CCRF-CEM.

Chemicals and materials Matrine (molecular formula: C15H24N2O; molecular weight: 248.36) was purchased from Sigma-Aldrich (Germany), and 50  mg/mL stock solution was prepared with ultrapure water. Fetal bovine serum (FBS), trypsin, L-glutamine, and RPMI-1640 medium were purchased from Biological Industrie s (USA). Total RNA and miRNA were isolated using RNeasy Mini Kit (Qiagen, Germany). The primers for mRNA and miRNA were obtained from Stellarray and GeneCopoeia (USA). The mRNAs and miRNAs analyzed in this study are listed in Table  1 and 2. Human T-ALL cell line CCRF-CEM was obtained from the American Type  Culture Collection (ATCC, Manassas, VA, USA) and cultured in RPMI-1640 medium supplemented with 10% FBS, 100 units/mL penicillin, and 100 μg/mL streptomycin and L-glutamine. Untreated CCRF-CEM line was used as a control group, while CCRF-CEM cell lines treated with different concentrations of matrine were defined as matrine-treated groups.

Cytotoxicity assay WST-1 assay (Roche Applied Science, Mannheim, Germany) was used to determine the cytotoxic effects of matrine at different concentrations. CCRF-CEM cell suspension was placed into a 96-well plate at a density of 1 × 104  cells in 100 µl per well. The cells were treated with matrine at a concentration of 0.5, 0.75, 1, 1.5, 2, 2.5, or 3 mg/ml for 72 hours. At the end of the experiment, 10 μl WST1 solution was added to each well and the plate was incubated for 1 hour. The absorbance of each well was measured spectrophotometrically at 450  nm with an enzyme linked immunosorbent assay (ELISA) reader (Thermo Fisher Scientific, Massachusetts, USA). All experiments were performed in triplicate.

Apoptosis analysis Apoptosis analysis was performed using BD Annexin V/ propidium iodide (PI) apoptosis detection kit (BD Biosciences, USA) and ApoDirect in situ DNA fragmentation assay kit (Biovision, USA). For the Annexin V/PI staining, the cells were seeded in 6-well plates and treated with matrine at a concentration of 2.4 mg/ml for 48 hours. After the incubation period, the cells were collected and washed twice with cold PBS and then resuspended in binding buffer to a concentration of 1 × 106 cells/ml. An aliquot of 100 μl of the resulting solution (1 × 105 cells) was transferred to a 5 ml culture tube, after which 5 μl of Annexin V and 5 μl of PI were added to each tube. The

2

Aslı Tetik Vardarlı, et al.: Molecular mechanisms of matrine in T-ALL cells TABLE  1. The effects of matrine treatment on gene expression profile of CCRF‑CEM cells Gene symbol CDKN1A IL6 KDR VEGFC NRP1 FLT1 JUNB STAT1 JUND CASP8 CDKN2B NRP2 FLT4 REL NFKBIA PDGFC MGMT BCL2L1 JAK2 KIT STAT3 PGF BAX RUNX3 BCL2 CDKN3 E2F1

Fold change 140.03 95.47 78.19 17.77 11.92 11.78 9.02 8.76 6.67 6.02 5.25 4.86 4.73 4.56 4.45 4.20 4.16 3.95 3.89 3.72 3.62 3.15 2.85 2.39 −4.68 −4.88 −5.38

TABLE 2. The effects of matrine treatment on miRNA expression in CCRF‑CEM cells

p 0.000159 0.000011 0.001288 0.000004 0.000908 0.000023 0.000138 0.000088 0.000511 0.000007 0.001667 0.000076 0.005928 0.000417 0.000087 0.0079 0.000345 0.000479 0.001661 0.000045 0.000549 0.005472 0.000548 0.000081 0.002893 0.000024 0.000128

miRNA hsa‑miR‑376b‑3p hsa‑miR‑106b‑3p hsa‑miR‑20a‑3p hsa‑miR‑29a‑5p hsa‑miR‑101‑5p hsa‑miR‑143‑5p hsa‑miR‑519a‑3p hsa‑miR‑143‑3p hsa‑miR‑148a‑5p hsa‑miR‑204‑5p hsa‑miR‑15a‑5p hsa‑miR‑24‑2‑5p hsa‑miR‑148a‑3p hsa‑miR‑30b‑5p hsa‑miR‑15b‑5p hsa‑miR‑106b‑5p hsa‑miR‑885‑3p hsa‑miR‑30a‑5p hsa‑miR‑24‑1‑5p hsa‑miR‑34a‑3p hsa‑miR‑20a‑5p hsa‑miR‑204‑3p hsa‑miR‑130a‑3p hsa‑miR‑181a‑5p hsa‑miR‑21‑3p hsa‑miR‑195‑5p hsa‑miR‑30b‑3p hsa‑miR‑195‑3p hsa‑miR‑181a‑3p hsa‑miR‑181b‑5p hsa‑miR‑15a‑3p hsa‑miR‑153‑3p hsa‑miR‑30a‑3p hsa‑miR‑519a‑5p hsa‑miR‑374a‑5p hsa‑miR‑15b‑3p hsa‑miR‑29a‑3p hsa‑miR‑374a‑3p hsa‑miR‑34a‑5p hsa‑miR‑101‑3p hsa‑miR‑24‑3p hsa‑miR‑630 hsa‑miR‑130a‑5p hsa‑miR‑320a

cells were incubated for 15 minutes at room temperature in the dark and then, within 30 minutes, analyzed by flow cytometry. ApoDirect in situ DNA fragmentation assay is based on the principle that terminal deoxynucleotidyl transferase (TdT) catalyzes the addition of FITC-labeled deoxyuridine triphosphates (FITC-dUTP) to the 3’-hydroxyl ends of double- and single-stranded DNA. For fixation of 0.5, 0.75, 1.0, 1.5, 2.0, 2.5, and 3.0  mg/ml matrine-treated CCRF-CEM cells and untreated control CCRF-CEM cells, they were resuspended in 1% paraformaldehyde on ice for 15 minutes. The cells were washed twice with PBS and treated with 70% alcohol while on ice, then kept at −20°C until analysis. For staining, the cells were washed with buffer containing 0.05% sodium azide, then resuspended with staining solution and incubated for 60 minutes at 37°C. After washing, the cells were incubated with PI/RNase A solution at room temperature for 30 minutes. The analysis was performed with BD Accuri C6 Flow Cytometer (BD Biosciences).

Fold change −37.09 −16.67 −15.92 −15.27 −12.18 −11.90 −11.73 −11.60 −11.20 −10.95 −10.62 −10.33 −9.06 −9.06 −8.89 −8.85 −8.61 −8.59 −8.55 −8.03 −7.69 −7.55 −7.44 −7.36 −7.16 −6.94 −6.93 −6.86 −6.57 −6.46 −6.24 −6.13 −6.09 −5.91 −5.85 −5.72 −5.5 −5.54 −4.31 −3.73 −3.10 −2.55 −1.32 −1.239

p 0.008595 0.028296 0.014856 0.083151 0.144244 0.097206 0.00534 0.150177 0.05312 0.001278 0.080913 0.128857 0.065481 0.022123 0.033958 0.021004 0.00006 0.009001 0.198366 0.116312 0.00001 0.023191 0.057459 0.0746 0.023407 0.098425 0.139264 0.020987 0.013863 0.083719 0.12063 0.236792 0.027658 0.051431 0.005774 0.066376 0.001615 0.093776 0.000157 0.116745 0.040189 0.26145 0.161284 0.878023

1  mL of buffer solution by vortexing. Cell concentration was adjusted with buffer solution to 1 × 106 cells/µL. For staining, the supernatant was removed and each tube was incubated at room temperature for 10  minutes with the addition of 250 μL solution-A (trypsin buffer). The tubes were vortexed gently after the addition of 200 μL of solution-B (trypsin inhibitor and RNase buffer) and incubated for 10 minutes at room temperature. Another 200 μL of cold (2–8°C) solution-C (PI lectin) was added to each vial and carefully vortexed. The cell suspension was incubated in the refrigerator for 10 minutes in the dark and analyzed with a flow cytometer within 30 minutes.

Cell cycle analysis Cell cycle analysis was performed in untreated and matrine-treated CCRF-CEM cells (2.4  mg/ml of matrine for 48 hours) using BD Cycletest assay according to the manufacturer’s instructions (BD Biosciences). Briefly, the cells were centrifuged at 300 × g for 5 minutes at room temperature and the supernatant was discarded. The pellet was resuspended in

3

Aslı Tetik Vardarlı, et al.: Molecular mechanisms of matrine in T-ALL cells

RESULTS

mRNA expression analysis Total RNA was isolated from untreated control CCRFCEM cells and matrine-treated CCRF-CEM cells (2.5 mg/ml of matrine for 48 hours) using the MagNA Pure LC RNA Isolation Kit (Roche Applied Science, Germany) and was then reverse transcribed into cDNA using the Transcriptor First Strand cDNA Synthesis Kit (Roche Applied Science). According to the manufacturer’s instructions, 1 μg total RNA was mixed with 50 pmol anchored oligo (dT)18 primer and incubated at 65°C for 10 minutes. Reactions were carried out for 30 minutes at 55°C for the reverse transcription step and 5 minutes at 85°C for the inactivation of transcriptor reverse transcriptase. All cDNA samples were 1:  4 diluted with RNase-free water and used as a template in real-time polymerase chain reaction (real-time PCR). The mRNA expression levels of genes associated with apoptosis and cell cycle were detected by quantitative reverse transcription PCR (qRT-PCR) method using SYBR Green all-in-one qRT-PCR master mix (Biomatic, USA/ Canada) and ABI-7500 instrument (Applied Biosystems, USA). For each reaction, 5 μL of ×2 all-in-one qRT-PCR master mix, 0.2 μL of ×50 ROX Dye, 5 μM of each primer, and 1 μL of diluted cDNA were mixed. Conditions for the qRT-PCR reactions were as follows: denaturation step, 95°C for 10  minutes; amplification step, 40  cycles at 95°C for 15 seconds, 60°C for 1 minute; and melting curve step, 95°C for 15 seconds, 60°C for 1 minute, 95°C for 30 seconds, and 60°C for 15  seconds. The relative expression of the genes was calculated using the 2−ΔΔCt method as described previously [34].

To analyze the cytotoxic effects of matrine, CCRF-CEM cells were treated with different concentrations of matrine (0.5, 0.75, 1, 1.5, 2, 2.5, and 3 mg/ml). After incubation for 24, 48, and 72 hours, the viability was evaluated by WST cytotoxicity assay. Our results indicate that matrine at concentrations of 0.5-2.0 mg/ml does not affect the viability of CCRF-CEM cells. However, the higher doses of matrine decreased the cell viability, and inhibitor concentration (IC50) values were 2.898, 2.400, and 2.390 mg/ml at 24, 48, and 72 hours, respectively (Figure 1). To evaluate the effects of matrine treatment on apoptosis, CCRF-CEM cells were treated with matrine at the IC50 dose (2.898, 2.400, and 2.390 mg/ml) for 72 hours, then analyzed using Annexin V-PI staining and DNA fragmentation assay. The apoptotic cell ratios of untreated and matrine-treated CCRF-CEM cells were 0.8% and 5.4%, respectively (Figure 2). The increase in matrine-induced apoptosis in CCRF-CEM cells was more prominent with 2.4 mg/ml dose of matrine. For the DNA fragmentation assay, CCRF-CEM cells were exposed to various concentrations of matrine (0.5, 0.75, 1, 1.5, 2, 2.5, and 3  mg/ml) for 48 hours. An average of 0.1% DNA fragmentation was detected for untreated control CCRFCEM cells and 0.5, 0.75, and 1  mg/ml matrine-treated cells. The ratios of DNA fragmentation were dramatically increased at 2, 2.5 and 3  mg/ml of matrine, i.e.,  12.7%, 19.4% and 61.2%, respectively (Figure  3). These results indicated that matrine induced apoptosis in CCRF-CEM cells in a dose-dependent manner. The cell cycle distribution of untreated and 2.4  mg/ml matrine-treated CCRF-CEM cells was determined by BD Cycletest assay. The distributions of cells in the G0/G1, S, and G2/M phases were 61.0% versus 85.5%, 25.9% versus 6.6%, and 14.2% versus 8.3% in untreated control CCRF-CEM cells and the matrine-treated cells, respectively (Figure 4). These results showed that matrine significantly arrested the CCRF-CEM cells in the G0/G1 phase, resulting in the majority of matrinetreated cells remained at that restriction point. After demonstrating the apoptotic and cell cycle arrest effects of matrine, we determined the mRNA expression levels of 49 candidate apoptotic genes by qRT-PCR analysis, in 2.5  mg/ml matrine-treated and untreated CCRF-CEM cells. We observed the upregulation of the CDKN1A (140.03 fold, p = 0.000159), IL6 (95.47 fold, p = 0.000011), KDR (78.19 fold, p = 0.001288), and VEGFC (17.7728 fold, p = 0.000004) genes, and downregulation of BCL2 (−4.68 fold, p = 0.002893), CDKN3 (−4.88 fold, p = 0.000024), and E2F1 (−5.38 fold, p = 0.000128) after matrine treatment (Table 1 and Figure 5). To determine the candidate miRNAs involved in the mechanism of matrine-induced apoptosis and cell cycle

miRNA expression analysis An miRNA isolation kit (Genoid, Hungary) was used to purify miRNA and an all-in-one first strand cDNA synthesis kit (Genoid) was used to obtain cDNA from untreated control CCRF-CEM cells and matrine-treated CCRF-CEM cells (2.5 mg/ml matrine for 48 hours). All qRT-PCR analyses were performed using the ABI 7500 Fast Real-Time PCR System (Applied Biosystems). The expression levels of miRNAs were assessed by relative quantification, and the fold expression changes were determined by the 2−ΔΔCT method [34].

Statistical analysis Matrine IC50 concentrations were calculated with the GraphPad Prism Software version  5.01 (GraphPad Software, Inc., La Jolla, CA, USA). The average of relative expression level, determined by the 2−ΔΔCT method for each mRNA and miRNA, was compared between untreated control CCRFCEM cells and matrine-treated groups using Student’s t-test. A p < 0.05 was accepted as statistically significant.

4

Aslı Tetik Vardarlı, et al.: Molecular mechanisms of matrine in T-ALL cells

arrest, we determined the expression levels of 44 different miRNAs by qRT-PCR analysis, in 2.5 mg/ml matrine-treated and untreated CCRF-CEM cells. Our results indicated that the relative expression levels of several miRNAs were decreased after matrine treatment, as shown in Table 2 and Figure  6. The miRNAs showing the greatest decrease in matrine-treated CCRF-CEM cells were as follows: hsa-miR376b-3p (−37.09 fold, p = 0.008), hsa-miR-106b-3p (−16.67 fold, p = 0.028), hsa-miR-20a-3p (−15.92 fold p = 0.014), hsamiR-519a-3p (−11.73 fold, p = 0.00534), and hsa-miR-204-5p (−10.95 fold, p = 0.001).

been investigated in human T-ALL cells. Our results indicated that matrine triggers cell cycle arrest and apoptosis in CCRF-CEM cells in a dose-dependent manner. Moreover, our study is the first to investigate potential miRNAs and target genes involved in the mechanism of matrine-induced cell death in T-ALL. Studies have shown that miRNAs regulate various cellular processes, including cell division, proliferation and apoptosis [40]. miRNAs are usually located in genomic regions involved in cancers and at chromosome fragile sites [41], and aberrant expression levels of miRNAs have been associated with different cancer types in humans [41-45]. Apoptosis is programmed cell death that occurs in both normal physiological and pathologic conditions. The disruption of this delicate balance can lead to the development of cancer. Autophagy is closely associated with eukaryotic cell death and apoptosis. In some cases, autophagy and apoptosis are controlled by the same proteins. Apoptotic signals can regulate autophagy, and autophagy can control apoptosis as well as other death mechanisms. The molecular interactions between autophagy and cell death are complex and, depending on physiological conditions, autophagy may promote or inhibit cell death [46]. In our study, of the 42 miRNAs that were downregulated after matrine treatment, maximum fold changes were observed for hsa-miR-376b-3p (−37.09 fold, p = 0.008595) and hsa-miR106b-3p (−16.67 fold, p = 0.028). Recent studies have revealed that miR-376b is downregulated in different carcinomas, sarcomas, solid tumors, and leukemias (e.g., uterine leiomyomas, ovarian cancer, non-small cell lung cancer [NSCLC], clear cell renal carcinoma, and chondrosarcomas) [41,45,47-50]. Furthermore, it was shown that the expression levels of miR376 family members were associated with tumor grade, invasion, metastasis, and/or response to chemotherapy [40]. miR-376b modulates autophagy by regulating the intracellular levels of the autophagy related 4C cysteine peptidase (ATG4C) and coiled-coil myosin-like BCL2-interacting protein (BECN1),

DISCUSSION Natural, non-toxic and chemopreventive agents, including matrine, are promising new therapies for the treatment of cancer. The antitumor activities of matrine have been reported in esophageal cancer [29], myeloid leukemia [30], gastric cancer [31], melanoma [32], hepatocellular carcinoma [33], and lung [35], breast [36], prostate [37], pancreatic [38], and colon cancers [39]. Up until now, the anticancer effects of matrine have not

FIGURE 1. Dose-response curves for cell viability of matrine‑treated CCRF-CEM cells after 24, 48, and 72 hours of exposure.

FIGURE 2. Apoptosis in untreated control CCRF-CEM cells and matrine-treated CCRF-CEM cells was assessed by Annexin V/propidium iodide staining. The apoptotic cell ratio of matrine-treated CCRF-CEM cells was increased significantly at 2.4 mg/ml.

5

Aslı Tetik Vardarlı, et al.: Molecular mechanisms of matrine in T-ALL cells

FIGURE 3. DNA fragmentation ratios of untreated control CCRF-CEM cells and matrine-treated CCRF-CEM cells. DNA fragmentation increased in the matrine-treated cells in a dose-dependent manner.

FIGURE 4. Distribution of G0/G1, S, and G2/M phases in untreated control CCRF-CEM cells and matrine-treated CCRF-CEM cells. The matrine-treated cells were arrested at the G0/G1 phase. 

key proteins that are directly involved in autophagy [40]. Lu et al. [51] investigated the effects of dysregulated miR-376b on liver regeneration and reported that the downregulation of miR-376b causes the overexpression of interleukin 6 (IL-6), which contributes to liver regeneration by inhibiting proliferation and inducing apoptosis in Hepa1-6 cells [51]. Similarly, our results showed that miR-376b was downregulated (−37.09 fold, p = 0.0008) and IL6 was upregulated (95.47 fold, p = 0.000011) in CCRF-CEM cells after matrine treatment compared to untreated control CCRF-CEM cells. These results demonstrate that the dysregulation of miR-376b and IL6 can promote cell death by both autophagy and apoptosis. Cell cycle checkpoints are controlled by specific miRNAs. Recent studies reported that the dysregulation of miRNAs

such as the members of miR-15b, miR-17, and miR-34 precursor families, as well as miR-106, miR-221 and miR-222 disrupts the progression through all cell cycle checkpoints [52]. In this study, we showed that the downregulation of hsamiR-106b-3p in matrine-treated CCRF-CEM cells led to cell cycle arrest in the G0/G1 phase by increasing the level of p21. Our findings are in agreement with those of Ivanovska et al. [12], who reported that the inhibition of miR-106 leads to the accumulation of tumor cells in G1 phase, also by increasing p21 levels. Moreover, the authors found that the upregulation of miR-106b inhibits the expression of p21 and causes G2/M arrest in doxorubicin-treated breast cancer cells [12]. Another study similarly showed that the overexpression of miR-106 suppressed the activation of p21 and led to G2/M

6

Aslı Tetik Vardarlı, et al.: Molecular mechanisms of matrine in T-ALL cells

FIGURE 6. Hierarchical clustering of the miRNA expression profiles of CCRF-CEM cells after matrine treatment. From the total of 44 miRNAs, 42 were downregulated and exhibited ≥2-fold change in expression relative to untreated control CCRF-CEM cells. All experiments were performed in triplicate and RNU6-2 was used as an endogenous control. For each treatment group, the cells were treated with 2.5 mg/ml matrine for 48 hours. Hsa‑miR‑376b-3p (p = 0.008), hsa-miR-106b-3p (p = 0.028), hsa-miR-20a-3p (p = 0.014), hsa-miR-519a-3p (p = 0.00534), and hsa-miR-204-5p (p = 0.001) expression levels were significantly decreased in the matrine-treated compared to untreated control cells.

FIGURE 5. Hierarchical clustering of the RNA expression profiles of CCRF-CEM cells after matrine treatment. From the total of 49 RNAs, 24 were upregulated and 3 were downregulated and exhibited ≥2-fold change in expression relative to the untreated CCRFCEM control cells. All experiments were performed in triplicate and Hs18s, GAPDH, and HPRT1 were used as housekeeping controls. For each treatment group, the cells were treated with 2.5 mg/ml matrine for 48 hours. CDKN1A (p = 0.000159), IL6 (p = 0.000011), KDR (p = 0.001288), and VEGFC (p = 0.000004) gene expressions were prominently upregulated, whereas BCL2 (p = 0.002893), CDKN3 (p = 0.000024), and E2F1 (p = 0.000128) gene expressions were downregulated in matrine-treated CCRF‑CEM cells compared with untreated control CCRF-CEM cells.

pathway and indirectly affects the expression of p21 and Bcl‑2-like protein 11 (Bim). Due to the suppression of miR106, the upregulation of E2F1 activates the TGFb tumor suppressor pathway and blocks cell cycle transition at the G1/S stage in gastric cancer cells [28]. Furthermore, An et al. [54] investigated the therapeutic effects and related molecular mechanisms of matrine on A549 cell line and reported that

arrest in radiation-treated prostate cancer cells [53]. In gastric cancer cells, the E2F1 gene was identified as the main target of miR-106b-25, which expression is regulated at the posttranscriptional level. It was reported that the dysregulation of E2F1 impairs transforming growth factor beta (TGFb)

7

Aslı Tetik Vardarlı, et al.: Molecular mechanisms of matrine in T-ALL cells

dose-dependent overexpression of miR-126 inhibits vascular endothelial growth factor gene expression and induces G0/G1 arrest in NSCLC cells [54]. Although molecular mechanisms of miR-106b-mediated cell cycle arrest have been investigated in several studies, it remains unclear what causes arrest at the checkpoint. Moreover, the discrepancies between different studies might be due to the differences in selected cell lines and/or treatments.

[8] Noguchi M, Hirata N, Edamura T, Ishigaki S, Suizu F. Intersection of apoptosis and autophagy cell death pathways. Austin J Mol Cell Biol 2015;2(2):1004. [9] Johnson CD, Esquela-Kerscher A, Stefani G, Byrom M, Kelnar K, Ovcharenko D, et al. The let-7 microRNA represses cell proliferation pathways in human cells. Cancer Res 2007;67(16):7713-22. https://doi.org/10.1158/0008-5472.CAN-07-1083. [10] Calin GA, Cimmino A, Fabbri M, Ferracin M, Wojcik SE, Shimizu M, et al. MiR-15a and miR-16-1 cluster functions in human leukemia. Proc Natl Acad Sci U S A 2008;105(13):5166-71. https://doi.org/10.1073/pnas.0800121105. [11] Linsley PS, Schelter J, Burchard J, Kibukawa M, Martin MM, Bartz  SR, et al. Transcripts targeted by the microRNA-16 family cooperatively regulate cell cycle progression. Mol Cell Biol 2007;27(6):2240-52. https://doi.org/10.1128/MCB.02005-06. [12] Ivanovska I, Ball AS, Diaz RL, Magnus JF, Kibukawa M, Schelter JM, et al. MicroRNAs in the miR-106b family regulate p21/CDKN1A and promote cell cycle progression. Mol Cell Biol 2008;28(7):2167‑74. https://doi.org/10.1128/MCB.01977-07. [13] Gillies JK, Lorimer IA. Regulation of p27Kip1 by miRNA 221/222 in glioblastoma. Cell Cycle 2007;6(16):2005-9. https://doi.org/10.4161/cc.6.16.4526. [14] Lujambio A, Esteller M. CpG island hypermethylation of tumor suppressor microRNAs in human cancer. Cell Cycle 2007;6(12):1455-9. https://doi.org/10.4161/cc.6.12.4408. [15] Yu Z, Baserga R, Chen L, Wang C, Lisanti MP, Pestell RG. MicroRNA, cell cycle, and human breast cancer. Am J Pathol 2010;176(3):1058‑64. https://doi.org/10.2353/ajpath.2010.090664. [16] le Sage C, Nagel R, Egan DA, Schrier M, Mesman E, Mangiola A, et al. Regulation of the p27(Kip1) tumor suppressor by miR221 and miR-222 promotes cancer cell proliferation. EMBO J 2007;26(15):3699‑708. https://doi.org/10.1038/sj.emboj.7601790. [17] Cimmino A, Calin GA, Fabbri M, Iorio MV, Ferracin M, Shimizu M, et al. MiR-15 and miR-16 induce apoptosis by targeting BCL2. Proc Natl Acad Sci U S A 2005;102(39):13944-9. https://doi.org/10.1073/pnas.0506654102. [18] Tusong H, Maolakuerban N, Guan J, Rexiati M, Wang WG, Azhati B, et al. Functional analysis of serum microRNAs miR-21 and miR-106a in renal cell carcinoma. Cancer Biomark 2017;18(1):79-85. https://doi.org/10.3233/CBM-160676. [19] He QY, Wang GC, Zhang H, Tong DK, Ding C, Liu K, et al. MiR-106a-5p suppresses the proliferation, migration, and invasion of osteosarcoma cells by targeting HMGA2. DNA Cell Biol 2016;35(9):506-20. https://doi.org/10.1089/dna.2015.3121. [20] Song J, Yin J, Bai Z, Zhang J, Meng H, Cai J, et al. The profile of serum microRNAs predicts prognosis for resected gastric cancer patients receiving platinum-based chemotherapy. Dig Dis Sci 2017;62(5):1223-34. https://doi.org/10.1007/s10620-017-4513-2. [21] Fan L, Qi H, Teng J, Su B, Chen H, Wang C, et al. Identification of serum miRNAs by nano-quantum dots microarray as diagnostic biomarkers for early detection of non-small cell lung cancer. Tumour Biol 2016;37(6):7777-84. https://doi.org/10.1007/s13277-015-4608-3. [22] Zhang GJ, Li JS, Zhou H, Xiao H, Li Y, Zhou T, et al. MicroRNA106b promotes colorectal cancer cell migration and invasion by directly targeting DLC1. J Exp Clin Cancer Res 2015;34(1):73. https://doi.org/10.1186/s13046-015-0189-7. [23] Hu Y, Li K, Asaduzzaman M, Cuella R, Shi H, Raguz S, et al. MiR106b~25 cluster regulates multidrug resistance in an ABC transporter-independent manner via downregulation of EP300. Oncol Rep 2016;35(2):1170-8. https://doi.org/10.3892/or.2015.4412. [24] Liang H, Studach L, Hullinger RL, Xie J, Andrisani OM. Downregulation of RE-1 silencing transcription factor (REST) in advanced prostate cancer by hypoxia-induced miR-106b~25. Exp

CONCLUSION In summary, our findings revealed that matrine suppresses cell growth and has an apoptotic effect in CCRF-CEM human T-ALL cell line. miR-106 affected cell cycle progression by blocking the transition of matrine-treated CCRF-CEM cells from G0 to G1 phase. Further in vivo studies are necessary to determine the functional role of matrine in T-ALL patients.

ACKNOWLEDGMENTS The authors wish to thank Jacqueline Renee Gutenkunst and Çağdaş Aktan (Department of Medical Biology, Beykent University, Istanbul, Turkey) for proofreading and valuable contributions to the manuscript, respectively.

DECLARATION OF INTERESTS The authors declare no conflict of interests.

REFERENCES [1] Evangelisti C, Evangelisti C, Chiarini F, Lonetti A, Buontempo  F, Neri  LM, et al. Autophagy in acute leukemias: a double-edged sword with important therapeutic implications. Biochim Biophys Acta 2015;1853(1):14-26. https://doi.org/10.1016/j.bbamcr.2014.09.023. [2] Richard NP, Pippa R, Cleary MM, Puri A, Tibbitts D, Mahmood S, et al. Combined targeting of SET and tyrosine kinases provides an effective therapeutic approach in human T-cell acute lymphoblastic leukemia. Oncotarget 2016;7(51):84214-27. https://doi.org/10.18632/oncotarget.12394. [3] Jovanovic M, Hengartner MO. MiRNAs and apoptosis: RNAs to die for. Oncogene 2006;25(46):6176-87. https://doi.org/10.1038/sj.onc.1209912. [4] Jing Z, Han W, Sui X, Xie J, Pan H. Interaction of autophagy with microRNAs and their potential therapeutic implications in human cancers. Cancer Lett 2015;356(2B):332-8. https://doi.org/10.1016/j.canlet.2014.09.039. [5] Alenzi FQ. Links between apoptosis, proliferation and the cell cycle. Br J Biomed Sci 2004;61(2):99-102. https://doi.org/10.1080/09674845.2004.11732652. [6] Wang RC, Levine B. Autophagy in cellular growth control. FEBS Lett 2010;584(7):1417-26. https://doi.org/10.1016/j.febslet.2010.01.009. [7] Eisenberg-Lerner A, Bialik S, Simon HU, Kimchi A. Life and death partners: Apoptosis, autophagy and the cross-talk between them. Cell Death Differ 2009;16(7):966-75. https://doi.org/10.1038/cdd.2009.33.

8

Aslı Tetik Vardarlı, et al.: Molecular mechanisms of matrine in T-ALL cells Cell Res 2014;320(2):188-99. https://doi.org/10.1016/j.yexcr.2013.09.020. [25] Fontana L, Fiori ME, Albini S, Cifaldi L, Giovinazzi S, Forloni M, et  al. Antagomir-17-5p abolishes the growth of therapy-resistant neuroblastoma through p21 and BIM. PLoS One 2008;3(5):e2236. https://doi.org/10.1371/journal.pone.0002236. [26] Dutto I, Tillhon M, Cazzalini O, Stivala LA, Prosperi E. Biology of the cell cycle inhibitor p21CDKN1A: Molecular mechanisms and relevance in chemical toxicology. Arch Toxicol 2015;89(2):155-78. https://doi.org/10.1007/s00204-014-1430-4. [27] Kim YK, Yu J, Han TS, Park SY, Namkoong B, Kim DH, et al. Functional links between clustered microRNAs: suppression of cell-cycle inhibitors by microRNA clusters in gastric cancer. Nucleic Acids Res 2009;37(5):1672-81. https://doi.org/10.1093/nar/gkp002. [28] Petrocca F, Visone R, Onelli MR, Shah MH, Nicoloso MS, de Martino I, et al. E2F1-regulated MicroRNAs ımpair TGFβdependent cell-cycle arrest and apoptosis in gastric cancer. Cancer Cell 2008;13(3):272-86. https://doi.org/10.1016/j.ccr.2008.02.013. [29] Wang Q, Du H, Geng G, Zhou H, Xu M, Cao H, et al. Matrine inhibits proliferation and induces apoptosis via BID-mediated mitochondrial pathway in esophageal cancer cells. Mol Biol Rep 2014;41(5):3009-20. https://doi.org/10.1007/s11033-014-3160-3. [30] Zhang LP, Jiang JK, Tam JW, Zhang Y, Liu XS, Xu XR, et al. Effects of matrine on proliferation and differentiation in K-562 cells. Leuk Res 2001;25(9):793-800. https://doi.org/10.1016/S0145-2126(00)00145-4. [31] Hu MJ, Zeng H, Wu YL, Zhang YP, Zhang S, Qiao MM, et al. Synergistic effects of matrine and 5-fluorouracil on tumor growth of the implanted gastric cancer in nude mice. Chin J Dig Dis 2005;6(2):68-71. https://doi.org/10.1111/j.1443-9573.2005.00201.x. [32] Liu XY, Fang H, Yang ZG, Wang XY, Ruan LM, Fang DR, et al. Matrine inhibits invasiveness and metastasis of human malignant melanoma cell line A375 in vitro. Int J Dermatol 2008;47(5):448-56. https://doi.org/10.1111/j.1365-4632.2008.03627.x. [33] Ma L, Wen S, Zhan Y, He Y, Liu X, Jiang J. Anticancer effects of the Chinese medicine matrine on murine hepatocellular carcinoma cells. Planta Med 2008;74(3):245-51. https://doi.org/10.1055/s-2008-1034304. [34] Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods 2001;25(4):402-8. https://doi.org/10.1006/meth.2001.1262. [35] Niu H, Zhang Y, Wu B, Zhang Y, Jiang H, He P. Matrine induces the apoptosis of lung cancer cells through downregulation of inhibitor of apoptosis proteins and the Akt signaling pathway. Oncol Rep 2014;32(3):1087-93. https://doi.org/10.3892/or.2014.3273. [36] Li LQ, Li XL, Wang L, Du WJ, Guo R, Liang HH, et al. Matrine inhibits breast cancer growth via miR-21/PTEN/Akt pathway in MCF-7 cells. Cell Physiol Biochem 2012;30(3):631-41. https://doi.org/10.1159/000341444. [37] Zhang P, Wang Z, Chong T, Ji Z. Matrine inhibits proliferation and induces apoptosis of the androgen-independent prostate cancer cell line PC-3. Mol Med Rep 2012;5(3):783-7. https://doi.org/10.3892/mmr.2011.701. [38] Liu T, Song Y, Chen H, Pan S, Sun X. Matrine inhibits proliferation and induces apoptosis of pancreatic cancer cells in vitro and in vivo. Biol Pharm Bull 2010;33(10):1740-5. https://doi.org/10.1248/bpb.33.1740. [39] Chang C, Liu SP, Fang CH, He RS, Wang Z, Zhu YQ, et al. Effects of matrine on the proliferation of HT29 human colon cancer cells and

its antitumor mechanism. Oncol Lett 2013;6(3):699-704. https://doi.org/10.3892/ol.2013.1449. [40] Korkmaz G, le Sage C, Tekirdag KA, Agami R, Gozuacik D. MiR376b controls starvation and mTOR inhibition-related autophagy by targeting ATG4C and BECN1. Autophagy 2012;8(2):165-76. https://doi.org/10.4161/auto.8.2.18351. [41] Calin GA, Sevignani C, Dumitru CD, Hyslop T, Noch E, Yendamuri S, et al. Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc Natl Acad Sci U S A 2004;101(9):2999-3004. https://doi.org/10.1073/pnas.0307323101. [42] Croce CM. Causes and consequences of microRNA dysregulation in cancer. Nat Rev Genet 2009;10(10):704-14. https://doi.org/10.1038/nrg2634. [43] Esquela-Kerscher A, Slack FJ. Oncomirs-microRNAs with a role in cancer. Nat Rev Cancer 2006;6(4):259-69. https://doi.org/10.1038/nrc1840. [44] Pencheva N, Tavazoie SF. Control of metastatic progression by microRNA regulatory networks. Nat Cell Biol 2013;15(6):546-54. https://doi.org/10.1038/ncb2769. [45] Wang T, Zhang X, Obijuru L, Laser J, Aris V, Lee P, et al. A microRNA signature associated with race, tumor size, and target gene activity in human uterine leiomyomas. Genes Chromosomes Cancer 2007;46(4):336-47. https://doi.org/10.1002/gcc.20415. [46] Yonekawa T, Thorburn A. Autophagy and cell death. Essays Biochem 2013;55:105-17. https://doi.org/10.1042/bse0550105. [47] Zhang L, Volinia S, Bonome T, Calin GA, Greshock J, Yang N, et al. Genomic and epigenetic alterations deregulate microRNA expression in human epithelial ovarian cancer. Proc Natl Acad Sci U S A 2008;105(19):7004-9. https://doi.org/10.1073/pnas.0801615105. [48] Son JW, Kim YJ, Cho HM, Lee SY, Jang JS, Choi JE, et al. MicroRNA expression profiles in Korean non-small cell lung cancer. Tubercu Respir Dis 2009;67(5):413-21. https://doi.org/10.4046/trd.2009.67.5.413. [49] Nakada C, Matsuura K, Tsukamoto Y, Tanigawa M, Yoshimoto T, Narimatsu T, et al. Genome-wide microRNA expression profiling in renal cell carcinoma: significant down-regulation of miR-141 and miR-200c. J Pathol 2008;216(4):418-27. https://doi.org/10.1002/path.2437. [50] Yoshitaka T, Kawai A, Miyaki S, Numoto K, Kikuta K, Ozaki T, et al. Analysis of microRNAs expressions in chondrosarcoma. J Orthop Res 2013;31(12):1992-8. https://doi.org/10.1002/jor.22457. [51] Lu S, Jiao H, Xu J, Zheng Y, Sun Y, Chen H. Downregulation of IL6 targeted MiR-376b may contribute to a positive IL6 feedback loop during early liver regeneration in mice. Cell Physiol Biochem 2015;37(1):233-42. https://doi.org/10.1159/000430348. [52] Chen D, Farwell MA, Zhang B. MicroRNA as a new player in the cell cycle. J Cell Physiol 2010;225(2):296-301. https://doi.org/10.1002/jcp.22234. [53] Li B, Shi XB, Nori D, Chao CK, Chen AM, Valicenti R, et al. Downregulation of microRNA 106b is involved in p21-mediated cell cycle arrest in response to radiation in prostate cancer cells. Prostate 2011;71(6):567-74. https://doi.org/10.1002/pros.21272. [54] An Q, Han C, Zhou Y, Li F, Li D, Zhang X, et al. Matrine induces cell cycle arrest and apoptosis with recovery of the expression of miR126 in the A549 non-small cell lung cancer cell line. Mol Med Rep 2016;14(5):4042-8. https://doi.org/10.3892/mmr.2016.5753.

9