<<

Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Chikusetsusaponin Ⅳa butyl ester (CS-Ⅳa-Be), a novel IL-6R antagonist, inhibits IL-6/STAT3 signaling pathway and induces cancer cell Jie Yang 1, 2, Shihui Qian 2, Xueting Cai 1, 2, Wuguang Lu 1, 2, Chunping Hu 1, 2, * Xiaoyan Sun1, 2, Yang Yang1, 2, Qiang Yu 3, S. Paul Gao 4, Peng Cao 1, 2

1. Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China 2. Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China 3. Shanghai Institute of Materia Medical, Chinese Academy of Sciences, Shanghai, 201203, China 4. Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY10065, USA title: CS-Ⅳa-Be, a novel IL-6R antagonist, inhibits IL-6/STAT3 Keywords: Chikusetsusaponin Ⅳ a butyl ester (CS- Ⅳ a-Be), STAT3, IL-6R, antagonist, cancer Grant support: P. Cao received Jiangsu Province Funds for Distinguished Young Scientists (BK20140049) grant, J. Yang received National Natural Science Foundation of China (No. 81403151) grant, and X.Y. Sun received National Natural Science Foundation of China (No. 81202576) grant. Corresponding author: Peng Cao Institute: Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu, China Mailing address: 100#, Shizi Street, Hongshan , Nanjing, Jiangsu, China Tel: +86-25-85608666 Fax: +86-25-85608666 Email address: [email protected] The first co-authors: Jie Yang and Shihui Qian The authors disclose no potential conflicts of interest.

1

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Abbreviations

IL-6: 6; STAT: Signal transducer and activator of transcription; GP130:

Glycoprotein 130; IL-6Rα: receptor alpha; LIFR: Leukemia inhibitory

factor receptor; LeptinR: ; :

(TNF)-related apoptosis-inducing ; DR: Death receptor; LIF: Leukemia

inhibitory factor; OSM: ; JAK: ; c-FLIP: cellular

FLICE-like inhibitory ; Bcl-xL: B-cell lymphoma-extra large; Mcl-1: Myeloid

cell leukemia 1; IAP: Inhibitor of apoptosis protein; MTT: Methyl thiazolyl

tetrazolium; TNF-α: Tumor necrosis factor alpha; EGF: Epidermal ;

IFN-γ: gamma; SDS-PAGE: Sodium dodecyl sulfate polyacrylamide gel

electrophoresis; PVDF: polyvinylidene fluoride; GM-CSF: Granulocyte-macrophage

colony-stimulating factor; NF-κB: Nuclear factor kappa--chain-enhancer of

activated B cells; ROS: Reactive oxygen species; TYK2: kinase 2; EGFR:

Epidermal growth factor receptor; IGF1R: Insulin-like growth factor 1 receptor;

PDGFRα: Platelet-derived growth factor receptor α; FGFR: Fibroblast growth factor

receptor; ELISA: Enzyme linked immunosorbent assay; EMSA: Electrophoretic

mobility shift assay; MST: Microscale thermophoresis; JNK: c-Jun N-terminal kinase;

ERK: Extracellular signal-regulated kinases; PARP: Poly ADP ribose polymerase;

MAPK: Mitogen-activated protein kinase; TNF-α: Tumor necrosis factor alpha;

PTPase: Protein tyrosine phosphatase; XIAP: X-linked inhibitor of apoptosis.

2

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Abstract

The activation of interleukin-6 (IL-6)/signal transducer and activator of transcription 3

(STAT3) signaling is associated with the pathogenesis of many cancers. Agents that

suppress IL-6/STAT3 signaling have cancer therapeutic potential. In this study, we

found that chikusetsusaponin Ⅳa butyl ester (CS-Ⅳa-Be), a triterpenoid saponin

extracted from Acanthopanas gracilistylus W.W.Smith, induced cancer cell apoptosis.

CS-Ⅳa-Be inhibited constitutive and IL-6-induced STAT3 activation, repressed

STAT3 DNA binding activity, STAT3 nuclear translocation, IL-6-induced STAT3

luciferase reporter activity, IL-6-induced STAT3 regulated anti-apoptosis genes

expression in MDA-MB-231 cells and IL-6-induced TF-1 cell proliferation.

Surprisingly, CS-Ⅳa-Be inhibited IL-6 family rather than other growth

factors induced STAT3 activation. Further studies indicated that CS-Ⅳa-Be is an

antagonist of IL-6 receptor via directly binding to the IL-6Rα with a Kd of 663±74

nM and the GP130 (IL-6Rβ) with a Kd of 1660±243 nM, interfering the binding of

IL-6 to IL-6R (IL-6Rα and GP130) in vitro and in cancer cells. The inhibitory effect

of CS-Ⅳa-Be on the IL-6-IL-6Rα-GP130 interaction was relatively specific as

CS-Ⅳa-Be showed higher affinity to IL-6Rα than to LIFR (Kd: 4990±915 nM) and

LeptinR (Kd: 4910±1240 nM). We next demonstrated that CS-Ⅳa-Be not only

directly induced cancer cell apoptosis but also sensitized MDA-MB-231 cells to

TRAIL-induced apoptosis via up-regulating DR5. Our findings suggest that

CS-Ⅳa-Be as a novel IL-6R antagonist inhibits IL-6/STAT3 signaling pathway and

sensitizes the MDA-MB-231 cells to TRAIL induced cell death.

3

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Introduction

Interleukin 6 (IL-6) is a pleiotropic mediating cancer associated chronic

inflammation and inflammatory response, plays important roles in the tumorigenesis

(1). IL-6 possesses three topologically distinct receptor binding sites: site 1 for

binding to the 80 kDa chain IL-6Rα, and sites 2 and 3 for interacting with the two

subunits of the signaling chain (GP130). The IL-6 signaling

transduces after the homodimerization of GP130, which becomes associated with

IL-6-binding chain (IL-6Rα) in the presence of IL-6. (2). As a common receptor,

GP130 also transduces signals delivered by the leukemia inhibitory factor (LIF), the

IL-11, the oncostatin M (OSM) and the others (3).

Signal transducer and activator of transcription 3 (STAT3), the key factor mediating

the IL-6-induced signaling becomes tyrosine phosphorylated by IL-6 induced

-associated kinases, the Janus kinase (JAK) family (4). The

IL-6/STAT3 signaling pathway mediates tumor immune suppression (5), tumor cell

survival, pre-metastatic niche formation and chemotherapy resistance (6).

Breast cancer is the most common malignancy and the primary cause of

cancer-related death in women worldwide (7). IL-6/JAK/STAT3 signaling plays a

critical and pharmacologically targetable role in orchestrating the composition of the

breast tumor microenvironment that promotes cancer cell growth, invasion and

metastasis (8) (9). Increasing clinical data have indicated that the circulating levels of

4

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

IL-6 in breast cancer patients correlate with clinical tumor stage and poor prognosis.

IL-6 plays two distinct roles in breast cancer: the systemic IL-6, reflecting whole body

metabolic and inflammatory status, is correlated with poor prognosis, advanced

disease and metastasis; the paracrine and autocrine IL-6 signaling controls cancer cell

growth, cancer renewal and metastasis (10) (11). In summary, the IL-6

mediated excessive STAT3 activation plays important roles in breast cancer.

Inhibitors of the IL-6/STAT3 signaling pathway should be effective against the

cancers with high levels of STAT3 activation. As a matter of fact, JAK3 inhibitor

(12, 13) and IL-6Rα monoclonal (14) have been used

in clinical trial. However, no natural IL-6/STAT3 inhibitory compound has been used

in cancer clinical trial. Identifying novel compounds that suppress the JAKs signaling

or antagonize IL-6R is a pressing issue to prevent and treat the cancers with aberrant

activated IL-6/STAT3 signaling.

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a

promising target for cancer therapy because it preferentially induces cancer cell

apoptosis with little or no effects on normal cells (15). However, some highly

malignant cancers, including breast cancer, (16) are resistant to TRAIL induced

apoptosis. The resistance of cancer cells to TRAIL occur at various points of the

TRAIL signaling pathways, dysfunctions of the death receptors DR4 and DR5, over

expression of cellular FLICE-like inhibitory protein (c-FLIP), B-cell lymphoma-extra

5

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

large (Bcl-xL), Myeloid cell leukemia 1 (Mcl-1) and the inhibitor of apoptosis

proteins (IAP) all lead to TRAIL resistance (17). TRAIL resistance can occur through

activating STAT3 in cancer cells (18). Therefore, a combination of TRAIL with

STAT3 inhibitor may prove to be a sound strategy to overcome the resistance.

Identifying novel agents that inhibit STAT3 activation not only can directly induce

apoptosis but also sensitize cancer cells to TRAIL-mediated apoptosis.

CS-Ⅳa-Be (Fig. 1A), a triterpenoid saponin extracted from a traditional Chinese

medicine (TCM) herb Acanthopanas gracilistylus W.W.Smith, exhibits

immunomodulation (19) and antithrombotic effects (20). The pharmacological effects

of CS-Ⅳa-Be have not been reported yet. In this study, we demonstrated that as an

IL-6R antagonist, CS-Ⅳa-Be inhibited IL-6/STAT3 signaling, repressed cancer cell

growth and synergized with TRAIL to induce breast cancer cell apoptosis.

Materials and Methods

Cell lines and cultures Human breast cancer MDA-MB-231 and MCF-7 cells, human hepatocellular cancer

HepG2 and MHCC97L cells, cervical cancer Hela cells, leukemic U937 and TF-1

cells, gastric cancer SGC-7901 cells were purchased from Cell Bank of the Shanghai

Institute of Biochemistry and in June 2013, Human fetal cell line

LO2 was purchased from Kunming Institute of Zoology, Chinese Academy of

Sciences in June 2013. All cell lines had been authenticated by provider through short

tandem repeat (STR) profiling. All cell lines were preserved in liquid N2 after receipt

6

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

and passaged for fewer than 6 months when we use them to do experiments. The cell

lines were last tested in June 2013 by above cell bank through STR profiling.

MDA-MB-231, MCF-7 and HepG2 cells were cultured in MEM medium

supplemented with 10 % fetal bovine serum (FBS). Hela and MHCC97L cells were

cultured in DMEM media (Invitrogen, Carlsbad, CA) supplemented with 10 % fetal

bovine serum (FBS) (Invitrogen, Carlsbad, CA). U937, SGC-7901 and LO2 cells

were cultured in RPMI-1640 media supplemented with 10 % FBS, TF-1 cells were

cultured in RPMI-1640 media supplemented with 10 % FBS and 2 ng/mL GM-CSF.

All cell lines were cultured in a humidified atmosphere with a 5 % CO2 incubator at

37 °C.

Agents and

CS- Ⅳ a-Be was identified and purified from the fruit of the Acanthopanar

gracilistμlusW.W.Smith, the purity of CS-Ⅳa-Be is 98.8 %, which was determined by

HPLC-ELSD method. A 20 mM solution of CS-Ⅳa-Be was prepared in dimethyl

sulfoxide (DMSO), stored as small aliquots at 4 °C. STAT3, phospho-STAT3 (Tyr705),

phosphor-STAT1(Tyr701), STAT1, phosphor-STAT5(Tyr694), STAT5,

phosphor-ERK1/2(Thr201/Tyr204), ERK1/2, phosphor-AKT(Ser473), AKT,

phosphor-JAK1(Tyr1022/1023), phosphor-JAK2(Tyr1007/1008), phosphor-Src(Tyr416),

Bcl-xL, Mcl-1, Survivin, XIAP, -3, Caspase-8, Caspase-9, PARP, DR5, DR4,

c-FLIP, IL-6Rα, GAPDH antibodies were purchased from Technology

(MA, USA). Anti-IL-6 and anti-GP130 antibodies were purchased from

Abcam((Massachusetts, US). IL-6 ELISA purchased from R&D system (MN,

USA). His-probe, NE-PER® Nuclear and Cytoplasmic Extraction and LightShift 7

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Chemiluminescent EMSA kit were obtained from Santa Cruz Biotechnology (CA,

USA). MTT (methyl thiazolyl tetrazolium), NAC(N-acetyl-L-cysteine), SP600125,

PD98059, SB203580 and Z-VAD-FMK were purchased from Sigma-Aldrich (MO,

USA). Recombinant human TNF-α, IL-6, EGF, IFN-γ were obtained from Tocris

Bioscience (Bristol, UK). Recombinant human IL-6Rα, GP130, LIFR and Leptin

receptor were purchased from Sino biological inc (Beijing, China). PierceTM Classic

Magnetic IP/Co-IP Kit were purchased from Thermo Fisher Scientific (MA, USA)

Immunoblot Assay

Cells were plated in six-well plate at a density of 5 × 106 cells/mL overnight before

being treated with CS-Ⅳa-Be. The total cell lysates were prepared as previously

described (21). The equal amounts of proteins from each sample were subjected to

sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) followed by

transfer to polyvinylidene fluoride (PVDF) membranes. After being blocked with 1%

(w/v) albumin from bovine serum (BSA) in TBST for 2 h, the membranes were

incubated with primary antibody overnight at 4 °C then washed and incubated with

secondary antibody conjugated with IgG DyLightTM for 1 h at room temperature.

Afterwards, membranes were washed and scanned with an Odyssey infrared

fluorescent scanner (LI-COR) and analyzed with Odyssey software version 3.

Cytotoxicity Assay

LO2, HepG2, MHCC97L, SGC-7901, TF-1, U937, MDA-MB-231, MCF-7 cells were

8

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

plated in triplicate in a 96-well plate at a density of 1 × 104 cells with 100 μL culture

medium per well in the absence or presence of indicated concentrations of CS-Ⅳa-Be

for 24 h. Cell viability was then determined by the MTT assay as described earlier

(22).

Apoptosis Assay Apoptotic cells were determined by using an FITC Annexin V Apoptosis Detection

Kit (BD Biosciences, USA) according to the manufacturer’s protocol. The cells were

washed and incubated with binding buffer containing Annexin V-FITC and PI in the

dark at room temperature for 15 min. Afterwards, the degree of apoptosis was

analyzed by FACScan laser flowcytometer (FACS Calibur, Becton Dickinson, USA).

The data were analyzed using the software CELLQuest.

JC-1 staining

Mitochondrial membrane potential was examined via JC-1 staining (Beyotime

Institute of Biotechnology, China). MDA-MB-231 cells were incubated with JC-1

working solution at 37°C in the dark for 20 min and observed by fluorescence

microscopy. In healthy cells, JC-1 forms complexes of J-aggregates showing punctate

red fluorescence at 590nm emission wavelength; however in apoptotic cells, JC-1

remains in the monomeric form showing diffused green fluorescence at 530nm

emission wavelength.

Immunofluorescence staining

MDA-MB-231 cells were seeded at the density of 70 ~ 80 % confluence per well into

9

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

24-well chamber slides. After being treated with test drugs for the indicated times,

cells were fixed with cold 4 % (w/v) paraformaldehyde for 20 min, rehydrated in PBS

for 15 min, and permeabilized in 0.1 % (w/v) TritonX-100 at room temperature for 10

min. After being washed with PBS, the cells were blocked with 3 % BSA for 1 hour

and then incubated with primary antibody at 4 °C overnight followed by

FITC-conjugated secondary antibody for 1 h at room temperature. The images green

fluorescence stained pSTAT3 were captured using a fluorescence microscope.

Luciferase reporter gene activity assay

We used the luciferase reporter assay (the Dual-Luciferase Reporter Assay System,

Promega, UK) to investigate the IL-6-induced transcriptional activity of STAT3.

Transient transfection was performed in 96-well plates at a cell density of 50% ~ 70%

confluence per well. The STAT3 luciferase reporter plasmid was co-transfected with

the pRL-TK plasmid (which encodes Renilla luciferase as an internal control for

transfection efficiency) using Lipofectamine 2000 (Invitrogen, Carlsbad, CA, USA)

according to the manufacturer’s instructions. The transfected cells were treated with

CS-Ⅳa-Be for various periods of time, and the cell lysates were prepared for

assessment of luciferase activity. Firefly and Renilla luciferase activities were

measured using a luminometer (Centro XS3 LB960, Berthold, Germany) according to

the manufacturer’s instructions. Relative firefly luciferase activity normalized by

Renilla luciferase activity was expressed as fold induction after treatment with CS-Ⅳ

a-Be compared with vehicle control (DMSO).

10

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Electrophoretic Mobility Shift Assay (EMSA) STAT3-DNA binding activity was analyzed by EMSA using biotin-labeled,

double-stranded STAT3 consensus-binding motif probe. STAT3 consensus oligo:

5'-GATCCTTCTGGGAATTCCTAGATC-3',3'-CTAGGAAGACCCTTAAGGATCT

AG-5'. Nuclear protein extracts were prepared from CS- Ⅳ a-Be treated

MDA-MB-231 cells and incubated with biotin-labeled probe. STAT3-DNA complex

was separated from free oligonucleotide on 5 % nondenaturing polyacrylamide gels,

then transferred to nylon membranes, and cross-linked for 15 min under a hand-held

UV lamp. Cross-linked, biotin-labeled DNA was detected using the

Chemiluminescent Nucleic Acid Detection Module (Pierce Biotechnology, inc).

IL-6 inhibitory bioassay

After being starved in media without GM-CSF for 24 h, various concentrations of CS-

Ⅳa-Be (2.5, 5, 7.5, 10, 12.5) μM in the presence of IL-6 (25 ng/ml) were added to the

TF-1 cells and incubated for 24 or 48 h, cell viability was then determined by the

MTT assay.

In vitro kinase assay

In vitro JAKs kinase (JAK2, JAK3, TYK2) assay for a series of CS-Ⅳa-Be

concentrations was performed by BPS Bioscience Service using Kinase-Glo Plus

luminescence kinase assay kit (Promega). In vitro panel kinases (JAK1, JAK2, JAK3,

TYK2, EGFR, IGF1R, PDGFRα, FGFR1) assay with two concentrations (25 μM and

11

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

50 μM) of CS-Ⅳa-Be was performed by Eurofins’ Kinase Profiler service according

to Cerep’s validation Standard Operating Procedure.

IL-6 receptor binding assay in cell level

The cell-based IL-6-receptor binding was performed using the kit of Fluorokine®

Biotinylated Human Interleukin 6 (R&D Systems) followed by the manufacturer’s

protocol. Firstly, U937, MDA-MB-231, Hela and HepG2 cells were treated with the

test compound for various periods of time and the cells were harvested and washed

twice with PBS to remove any residual growth factors that may be present in the

culture media. Next, the cells were resuspended in PBS to a final concentration of 4×

106 cells/mL, 10 μL of biotinylated IL-6 was added to a 25 μL aliquot of the cell

suspension for a total reaction volume of 35 μL. As a negative staining control, an

aliquot of cells were stained with 10 μL of negative control reagent (a soybean trypsin

inhibitor protein biotinylated as the IL-6). The cells were then incubated for 30 min at

4 °C followed by the addition of 10 μL Avidin-FITC reagent and another 30 min

incubation at 4 °C in the dark before being analyzed by flow cytometry.

Analysis of IL-6Rα, DR4 and DR5 surface expression

Cells were treated with CS-Ⅳa-Be for various periods of times before being detached

with EDTA and washed twice with staining buffer. Cells were then incubated with

mouse monoclonal anti-human DR5, DR4 or IL-6Rα antibodies for 45 min at 4 °C,

washed twice, incubated with FITC-conjugated secondary antibody for 30 min at 4 °C

12

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

and washed again, then re-suspended in staining buffer for flow cytometric analysis.

IL-6 receptor binding assay in vitro

IL-6 receptor binding was performed following the method described earlier (23).

Briefly, Ninety-six well-plates (Nunc) were coated with 500 ng/mL of human

recombinant soluble IL-6 receptor (sIL-6R), washed and blocked. The sIL-6R coated

96-well plates were incubated with 6.2 nM of recombinant human IL-6, or 6.2 nM

IL-6 plus one of six concentrations of CS-Ⅳa-Be, in triplicates, and washed to

remove unbound IL-6, the wells then were incubated with a monoclonal rabbit

anti-human IL-6 antibody (200 ng/mL), washed and incubated with goat anti-rabbit

IgG-HRP (40 ng/mL, Santa Cruz). Sample wells were washed and developed with

tetramethylbenzidine (100 μL/well, 10 min at RT and stopped with 2 M H2SO4).

Optical density in each well was determined using a microplate reader (Multiskan

Ascent, Thermo) 450 nm with a correction wavelength of 630 nm. Non-specific

binding of IL-6 is determined by the reading of wells without sIL-6R. Data were

analyzed by non-linear regression analysis using GraphPad Prism4 (Graph Pad, San

Diego, CA). Maximum binding of IL-6 (100 % bound) is defined as the absorbance in

the absence of CS-Ⅳa-Be. The absorbance in the presence of various concentrations

of CS-Ⅳa-Be is calculated as percent of maximum binding of IL-6.

Microscale Thermophoresis (MST) assay

Binding assay of labeled recombinant human IL-6Rα, GP130, LIFR, and LeptinR to

13

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

CS-Ⅳa-Be was performed using the Monolith NT.115 kit (NanoTemper

Technologies, Germany). Briefly, 10 μM recombinant human IL-6Rα, GP130, LIFR,

and LeptinR were labeled in PBS buffer using the Protein Labeling Kit RED-NHS

followed by the manufacturer’s protocol (NanoTemper Technologies, Germany). A

16-point titration series of CS-Ⅳa-Be were added to the labeled protein (200 nM) in

PBST buffer (PBS + 0.05 % Tween-20), the total volume of mixed sample was 20 μL

and the volume ratio of CS-Ⅳa-Be to labeled protein is 1:1, the final DMSO

concentration for each protein-compound sample was 2 %. The protein-compound

samples were equilibrated for 5 min at room temperature, loaded into the MST

Premium Coated capillaries (NanoTemper Technologies, Germany) and assayed by

MST. The thermophoresis data was analyzed using NT Analysis 1.5.41 (NanoTemper

Technologies, Germany) to compute Kd value according to the law of mass action.

The experiment was performed in triplicate, and the fits are represented as mean ± SD.

Data normalization and curve fitting were performed using GraphPad Prism 5.

Co-immunoprecipitation (Co-IP) assay

The interaction of IL-6Rα with GP130 in the presence of IL-6 was assayed by Co-IP

assay according to the manufacturer’s protocol of PierceTM Classic Magnetic IP/Co-IP

Kit. At room temperature, 10 μg of recombinant human IL-6Rα was pre-incubated

with 0, 5 or 10 μM of CS-Ⅳa-Be for 2 h in IP wash buffer, added 10 μg of

recombinant human IL-6 and GP130 incubation for another 2 h, then added 2 μg of

His antibody and incubated for 2 h to form immune complex. Place 25 μL of

14

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

pre-washed Pierce Protein A/G Magnetic Beads into the above immune complex and

incubated for 1 h with mixing. Then the beads were washed and the target antigen was

eluted with Alternative Elution, the target antigen and binding proteins were

immuno-blotted with indicated antibodies by western blot assay.

Small interfering RNA (siRNA)-mediated Gene Silencing

DR5 specific siRNA was obtained from RiboBio (Guangzhou, China). Transient

transfection was performed using Lipofectamine 2000. Briefly, cells were transfected

with indicated concentration of siRNAs for 24 h, followed by treatment with CS-Ⅳ

a-Be for indicated time, the cells were then harvested for flow cytometry analysis.

Data analysis

The experimental results presented in the figures are representative of three or more

independent experiments. The data are presented as the mean values ± standard error.

Statistical comparisons between the groups were done using one-way analysis of

variance. Values of P < 0.05 were considered to be statistically significant.

Results

CS-Ⅳa-Be induces cancer cell apoptosis

CS- Ⅳ a-Be (Fig. 1A), a triterpenoid saponin extracted from Acanthopanas

gracilistylus W.Smith, was identified as a cell death inducer. We investigated the

effects of CS-Ⅳa-Be on the viability of cancer cells and found that CS-Ⅳa-Be

15

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

inhibits the viability of various kinds of cancer cells (including hepatocellular, gastric,

leukemia and breast cancers). Breast cancer MDA-MB-231 and MCF-7 cells were

more sensitive to the CS-Ⅳa-Be-induced cell viability attenuation than other cancer

cells (Fig. 1B). We next investigated whether the CS-Ⅳa-Be-induced breast cancer

cell inhibition due to apoptosis. We then found that CS-Ⅳa-Be induced pro-Caspase 3,

8 and 9 cleavage (Fig. 1C). Furthermore, Z-VAD-FMK, a pan-Caspase inhibitor,

significantly offset the CS-Ⅳa-Be-induced MDA-MB-231 cell viability attenuation

and apoptosis (Fig. 1D and E). In addition, CS-Ⅳa-Be induced a decrease of

mitochondrial membrane potential (Fig. 1F). The results indicate that CS-Ⅳa-Be

promotes breast cancer cell apoptosis.

CS-Ⅳa-Be selectively inhibits constitutive and IL-6 family member-induced

STAT3 activation in MDA-MB-231 cells

To identify the signaling pathways that were involved in the CS-Ⅳa-Be-induced cell

apoptosis, we investigated the effects of CS-Ⅳa-Be on STAT3, (nuclear factor

kappa-light-chain-enhancer of activated B) NF-κB, Mitogen-activated protein kinase

(MAPK) and AKT signaling. CS-Ⅳa-Be was found to inhibit the constitutive and

IL-6-induced STAT3 and JAK1, JAK2, Src activation rather than constitutive and

IL-6-induced ERK1/2 and AKT activation (Fig. 2A and B). Other IL-6 family

cytokines (IL-11, LIF and OSM , which share the same GP130 receptor to transducer

signal) induced STAT3 activation was also inhibited by CS-Ⅳa-Be (Fig. 2C). But

interestingly, constitutive and EGF-induced STAT3, STAT5 activation or

16

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

IFN-γ-induced STAT3, STAT1 activation were not inhibited by CS-Ⅳa-Be (Fig. 2A

and D). In addition, ROS production was observed upon CS-Ⅳa-Be treatment

(Supplementary Fig. S1A), but treatment with ROS, c-Jun N-terminal

kinase (JNK1/2/3), extracellular signal-regulated kinases (ERK1/2) or p38 specific

inhibitor did not reverse the CS- Ⅳ a-Be-induced cell viability attenuation in

MDA-MB-231 cells (Supplementary Fig. S1B). CS-Ⅳa-Be had no inhibitory effects

on the TNF-α-induced NF-κB transcription activity (Supplementary Fig. S1C) and

NF-κB-DNA binding activity (Supplementary Fig. S1D).

CS-Ⅳa-Be abrogates IL-6/STAT3 downstream signaling

We further investigated the effects of CS-Ⅳa-Be on constitutive and IL-6-induced

STAT3 downstream signaling in MDA-MB-231 cells. CS-Ⅳa-Be was found to

inhibit the pSTAT3 nuclear translocation (Fig. 3A), the STAT3-DNA binding activity

(Fig. 3B), IL-6 induced STAT3 luciferase activity (Fig. 3C) in MDA-MB-231 cells.

CS-Ⅳa-Be also down-regulated the constitutive and IL-6-induced Survivin, XIAP,

Bcl-xL and Mcl-1 expression (Fig. 3D and E). IL-6 induced TF-1 cell proliferation

was also inhibited by CS-Ⅳa-Be (Fig. 3F). These findings suggest that CS-Ⅳa-Be

inhibit IL-6/STAT3 downstream signaling.

CS-Ⅳa-Be doses not inhibit the enzyme activity of non-receptor or receptor

tyrosine kinases of STAT3 in vitro

As STAT3 was mediated by several non-receptor tyrosine kinases

17

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

(24) and receptor kinases (25). We investigated whether CS-Ⅳa-Be impact the

activity of upstream kinases. CS-Ⅳa-Be has showed inhibitory effects on the

IL-6-induced phosphorylation of JAK1, JAK2 and Src (Fig. 2B) in MDA-MB-231

cells. The inhibitory effect of CS-Ⅳa-Be on IL-6-induced STAT3 activation might be

through its repression on JAK1, JAK2 and Src kinase activity. However, by in vitro

kinase assay, we found no inhibitory effect of CS-Ⅳa-Be on the enzyme activity of

tyrosine kinases such as JAKs family kinases (JAK1, JAK2, JAK3 and TYK2) (Fig.

4A and B), Src, epidermal growth factor receptor (EGFR), insulin-like growth factor 1

receptor (IGF1R), platelet-derived growth factor receptor α (PDGFRα) and fibroblast

growth factor receptor 1 (FGFR1) (Fig. 4B), even at 50 μM, the drug concentration

much higher than the IC50 of CS-Ⅳa-Be in MDA-MB-231 cells. Therefore, we

suggest that CS-Ⅳa-Be is not a inhibitor.

CS-Ⅳa-Be blocks IL-6 binding to its cell surface receptor

We had shown CS-Ⅳa-Be inhibited IL-6-induced JAKs and STAT3 activation in

breast cancer cells, without affecting JAKs enzyme activity in vitro. These findings

made us speculate that CS-Ⅳa-Be might disrupt the upstream signaling such as the

interaction of IL-6 to its receptor. To test this hypothesis, we investigated whether

CS-Ⅳa-Be affect the binding of IL-6 to cell surface IL-6R in cancer cells using

biotin-labled IL-6 by flow cytometry analysis. To rule out the possibility that CS-Ⅳ

a-Be might down-regulate IL-6R protein levels, we also measured the cell surface

IL-6Rα expression upon CS-Ⅳa-Be treatment. The results demonstrated that CS-Ⅳ

18

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

a-Be indeed lowered biotin-labled IL-6 binding to IL-6R in various cancer cells tested

(U937, MDA-MB-231, Hela, HepG2) in a dose dependent manner, without

decreasing IL-6Rα cell surface expression (Fig. 5 A, B and Supplementary Fig. S2).

To further confirm our hypothesis that CS-Ⅳa-Be inhibits IL-6-IL-6R binding, we

investigated the effects of CS-Ⅳa-Be on IL-6-IL-6Rα interaction using an in vitro

ELISA assay. Our results showed that CS- Ⅳ a-Be significantly inhibited the

IL-6/IL-6Rα interaction with maximal inhibition rate less than 20 % (Fig. 5C). As CS-

Ⅳa-Be can not antagonize IL-6-IL-6Rα interaction completely, we considered CS-Ⅳ

a-Be could be a non-competitive IL-6Rα antagonist.

Biochemical characterization of CS-Ⅳa-Be/ IL-6R interaction

Our results have illustrated that CS-Ⅳa-Be inhibited the IL-6-IL-6R interaction, we

next studied the binding specificity of CS-Ⅳa-Be to IL-6 or IL-6R. We first measured

the binding affinity (Kd: 48 ± 2 nM) of IL-6 to IL-6Rα (Supplementary Fig. S3A)

using microscale thermophoresis (MST) analysis, which allows a sensitive detection

of small-molecule binding to a protein target. Next, we measured the binding

characteristics of CS-Ⅳa-Be to IL-6 or IL-6R and found that CS-Ⅳa-Be can bind to

IL-6Rα (Kd: 663 ± 74 nM) and IL-6Rβ (GP130) (Kd: 1660 ± 243 nM) (Fig. 5D and E)

while had no binding affinity to IL-6 (Supplementary Fig. S3B). The affinity of CS-

Ⅳa-Be to IL-6R was relatively higher than other IL-6 family receptors such as

LeptinR (Kd: 4910 ± 1240 nM) and LIFR (Kd: 4990 ± 915 nM) (Fig. 5E and

19

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Supplementary Fig. S3). IL-6Rα pretreatment with CS-Ⅳa-Be also inhibited

IL-6Rα/GP130 interaction in the presence of IL-6 (Fig. 5F). The expression of IL-6R,

IL-6, pSTAT3 in different cancer cell lines were compared (Supplementary Fig. S4A,

B and C). Since MDA-MB-231 cells secret the highest levels of IL-6 (Supplementary

Fig. S4B), we also considered if CS-Ⅳa-Be could inhibit IL-6 secretion, however,

CS-Ⅳa-Be exhibited little inhibitory effect on IL-6 secretion (Supplementary Fig.

S4D). Overall, these results suggest that CS-Ⅳa-Be binds to IL-6R (IL-6Rα and

GP130) and disrupts IL-6/IL-6Rα/GP130 interaction.

CS-Ⅳa-Be synergized with TRAIL to induce apoptosis in breast cancer cells

Resistance to TRAIL induced apoptosis in many cancers limits its clinical use as an

anticancer agent (26), and aberrant STAT3 activation has been suggested to be part of

the causes(27). We have proved that CS-Ⅳa-Be is an effective IL-6/STAT3 inhibitor,

we next determined whether CS-Ⅳa-Be can synergize with TRAIL to induce

apoptosis in breast cancer cells. We treated the breast cancer MDA-MB-231 cells

with CS-Ⅳa-Be alone or combined with TRAIL and analyzed cell apoptosis. The

cells treated by TRAIL/CS-Ⅳa-Be combination demonstrated obvious apoptotic

morphology (Fig. 6A) and increased cell apoptosis (Fig. 6B and C).

CS-Ⅳa-Be up-regulates DR5 synergized with TRAIL

With our data showing CS-Ⅳa-Be activating Caspase-8, we hypothesized that the

death receptor pathways could be involved in the CS-Ⅳa-Be-induced apoptosis. We

20

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

then investigated the effects of CS-Ⅳa-Be on the expression of death receptors and

found that CS-Ⅳa-Be increased DR5 protein levels in breast cancer MDA-MB-231

and MCF-7 cells (Fig. 6D). We next analyzed the cell surface DR5 expression levels

in CS-Ⅳa-Be treated MDA-MB-231 cells, which was also increased upon CS-Ⅳa-Be

treatment. In contrast, the levels of DR4 didn’t increase by CS-Ⅳa-Be (Fig. 6D). It

has previously been suggested that the up-regulation of DR5 synergizes with TRAIL

to induce apoptosis. We then examined whether CS-Ⅳa-Be induced up-regulation of

DR5 synergized with TRAIL. Transfection of MDA-MB-231 cells with DR5 specific

siRNA resulted in a marked inhibition of DR5 surface expression (data not shown).

After the combined treatment with CS-Ⅳa-Be and TRAIL, the apoptosis rate in the

DR5-knocked down cells was significantly reduced compared with the control (Fig.

6E). We suggest that the synergized induction of apoptosis by CS-Ⅳa-Be and TRAIL

was dependent on the DR5 up-regulation, at least partially.

Discussion

CS-Ⅳa-Be, a triterpenoid saponin from TCM Acanthopanas gracilistylus W.W.Smith,

has previously been shown to possess cytotoxic activity in many cancer cells (28)

although the mechanism remains elusive. Herein, we uncovered that CS-Ⅳa-Be

decrease cell viability in various cancer cells by inducing apoptosis. Numerous

signaling pathways, such as IL-6/STAT3, ROS/MAPK, TNF-α/NF-κB and AKT, are

involved in regulating cell proliferation and survival (29, 30). Though CS-Ⅳa-Be

treatment induced ROS production, we demonstrated that oxidative stress is not the

21

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

primary cause for CS-Ⅳa-Be induced cell apoptosis. TNF-α/NF-κB and AKT are also

not involved in CS-Ⅳa-Be mediated cell apoptosis. We next discovered CS-Ⅳa-Be

inhibited IL-6 family cytokines induced STAT3 activation. Especially, CS-Ⅳa-Be

showed more potential inhibitory effect on IL-6 induced STAT3 activation than other

family cytokines (Fig. 2B and C). Interestingly, CS-Ⅳa-Be showed no inhibitory

effect on EGF or IFN-γ induced STAT3, STAT5 or STAT1 activation as well as

constitutive STAT5 or STAT1 activation.

Tyrosine kinases (JAKs, Src, EGFR, FGFR1, IGF1R and PDGFRα) are not involved

in the mechanism of CS-Ⅳa-Be mediated IL-6/STAT3 inhibition cause CS-Ⅳa-Be

showed no inhibitory effect on the enzyme activity of any of the kinases. We further

found that CS-Ⅳa-Be directly binds to IL-6R (IL-6Rα and GP130) and disrupts the

interaction of IL-6/IL-6Rα/GP130 in vitro and in cancer cells. Since CS-Ⅳa-Be

showed higher affinity to IL-6Rα than other IL-6 family receptors (LIFR and

LeptinR), we suggest that CS-Ⅳa-Be is an relatively specific IL-6R antagonist,

interfering IL-6 induced STAT3 activation. In view of the fact that GP130 is a

common subunit of IL-6 family receptors and CS-Ⅳa-Be also can directly bind

GP130, CS-Ⅳa-Be exhibit inhibitory effect on other IL-6 family cytokines (IL-11,

LIF and OSM) induced STAT3 activation. The evidences that U937 cells express

relatively high levels of cell surface IL-6Rα than other cancer cells (Fig. 5A and

Supplementary Fig. S4A) and IL-6Rα knockdown partly reversed the CS-Ⅳa-Be

induced U937 cell apoptosis (Supplementary Fig. S4E and F) also demonstrate the

22

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

key role of IL-6Rα in mediating CS-Ⅳa-Be induced cancer cell apoptosis.

Many kinds of natural compounds have been reported to abolish IL-6/STAT3

signaling through different mechanisms(31) such as targeting IL-6Rα (32) or

GP130(33), directly inhibiting JAKs (34), inducing PTPase expression (35), inhibiting

STAT3 translocation (36) and blocking JAK-STAT3 interaction (37). The natural

antagonist of IL-6R is rarely reported except for the ERBF, which is isolated from

bufadienolide and sensitize breast cancer cells to TRAIL-induced apoptosis via

inhibition of STAT3/Mcl-1 pathway (38). In accordance with their results, our data

suggest CS-Ⅳa-Be is a novel natural IL-6R antagonist and shows anti-cancer activity.

To further confirm the inhibitory effect of CS-Ⅳa-Be on cancer cells was via

IL-6/STAT3 inhibition, we investigated the GP130, pSTAT3, STAT3, IL-6 secretion

levels and cell surface IL-6Rα expression in different cancer cells (Supplementary Fig.

S4A, B and C) and compared the sensitivity of different cancer cells to CS-Ⅳa-Be.

MDA-MB-231 cells express the highest levels of GP130, pSTAT3 and IL-6 than

other cancer cells determined MDA-MB-231 cell line is sensitive to CS-Ⅳa-Be. We

found that MCF-7 cell line expresses low level of IL-6 and p-STAT3, but it is also

sensitive to CS-Ⅳa-Be. It has been reported that low level of IL-6 is indispensible to

maintain the growth of MCF-7 breast cancer cells (39). In addition, CS-Ⅳa-Be also

can bind GP130, the expression of GP130 in MCF-7 cells is higher than other cancer

cells except MDA-MB-231. We speculate that MCF-7 cells are more dependent on

23

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

GP130 mediated other IL-6 family cytokines induced signaling to survive and

antagonize GP130 by CS-Ⅳa-Be can effectively inhibit MCF-7 viability.

TRAIL resistance in many cancers limits its therapeutic use in clinical scenarios. The

STAT3 trans-regulated pro-survival genes Survivin, c-FLIP, XIAP, Bcl-xL and Mcl-1

are involved in regulating cell apoptosis and TRAIL sensitivity to cancer cells (40).

The expression levels of these genes were attenuated by CS-Ⅳa-Be, which exhibits

enhanced apoptosis in breast cancer cells via up-regulating DR5 expression, which is

consistent with the report that up-regulation of the cell surface DR5 was dependent on

the suppression of STAT3 activation (41). These studies suggest that DR5 is

negatively regulated by STAT3, inhibition of STAT3 phosphorylation by CS-Ⅳa-Be

accounts for DR5 induction and the apoptosis enhancing synergy with TRAIL.

In summary, we characterized CS-Ⅳa-Be as a novel natural IL-6R antagonist, that

inhibits IL-6/STAT3 signaling, induces cancer cell apoptosis and synergizes with

TRAIL in breast cancer cells. Therefore, synergistic combination of CS-Ⅳa-Be with

TRAIL has potential to be a more effective strategy to treat cancers with aberrant

IL-6/STAT3 activation.

Acknowledgement We thank Xiaojun Xu and Ping Zhou in State Key Laboratory of Natural Medicine

(China Pharmaceutical University) for providing us with the help for MST analysis.

24

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

References

1. Rattigan Y, Hsu JM, Mishra PJ, Glod J, Banerjee D. Interleukin 6 mediated recruitment of mesenchymal stem cells to the hypoxic tumor milieu. Experimental cell research. 2010;316:3417-24. 2. Kishimoto T, Akira S, Taga T. Interleukin-6 and its receptor: a paradigm for cytokines. Science. 1992;258:593-7. 3. Kishimoto T, Taga T, Akira S. Cytokine . Cell. 1994;76:253-62. 4. Schindler C, Levy DE, Decker T. JAK-STAT signaling: from to cytokines. The Journal of biological chemistry. 2007;282:20059-63. 5. Ferguson SD, Srinivasan VM, Heimberger AB. The role of STAT3 in tumor-mediated immune suppression. Journal of neuro-oncology. 2015;123:385-94. 6. Spitzner M, Ebner R, Wolff HA, Ghadimi BM, Wienands J, Grade M. STAT3: A Novel Molecular Mediator of Resistance to Chemoradiotherapy. Cancers. 2014;6:1986-2011. 7. Place AE, Jin Huh S, Polyak K. The microenvironment in breast cancer progression: biology and implications for treatment. Breast cancer research : BCR. 2011;13:227. 8. Senst C, Nazari-Shafti T, Kruger S, Honer Zu Bentrup K, Dupin CL, Chaffin AE, et al. Prospective dual role of mesenchymal stem cells in breast tumor microenvironment. Breast cancer research and treatment. 2013;137:69-79. 9. Chang Q, Bournazou E, Sansone P, Berishaj M, Gao SP, Daly L, et al. The IL-6/JAK/Stat3 feed-forward loop drives tumorigenesis and metastasis. Neoplasia. 2013;15:848-62. 10. Fuksiewicz M, Kowalska M, Kotowicz B, Rubach M, Chechlinska M, Pienkowski T, et al. Serum soluble tumour necrosis factor receptor type I concentrations independently predict prognosis in patients with breast cancer. Clinical chemistry and laboratory medicine : CCLM / FESCC. 2010;48:1481-6. 11. Dethlefsen C, Hojfeldt G, Hojman P. The role of intratumoral and systemic IL-6 in breast cancer. Breast cancer research and treatment. 2013;138:657-64. 12. Cutolo M, Meroni M. Clinical utility of the oral JAK inhibitor tofacitinib in the treatment of rheumatoid arthritis. Journal of inflammation research. 2013;6:129-37. 13. Vyas D, O'Dell KM, Bandy JL, Boyce EG. Tofacitinib: The First Janus Kinase (JAK) inhibitor for the treatment of rheumatoid arthritis. The Annals of pharmacotherapy. 2013;47:1524-31. 14. Hashizume M, Tan SL, Takano J, Ohsawa K, Hasada I, Hanasaki A, et al. Tocilizumab, A Humanized Anti-IL-6R Antibody, as an Emerging Therapeutic Option for Rheumatoid Arthritis: Molecular and Cellular Mechanistic Insights. International reviews of immunology. 2015;34:265-79. 15. Walczak H, Miller RE, Ariail K, Gliniak B, Griffith TS, Kubin M, et al. Tumoricidal activity of tumor necrosis factor-related apoptosis-inducing ligand in vivo. Nature medicine. 1999;5:157-63. 16. Rahman M, Pumphrey JG, Lipkowitz S. The TRAIL to targeted therapy of breast cancer. Advances in cancer research. 2009;103:43-73. 17. Zhang LD, Fang BL. Mechanisms of resistance to TRAIL-induced apoptosis in cancer. Cancer Gene Ther. 2005;12:228-37. 18. Chen KF, Tai WT, Liu TH, Huang HP, Lin YC, Shiau CW, et al. overcomes TRAIL resistance of hepatocellular carcinoma cells through the inhibition of STAT3. Clinical cancer research : an official journal of the American Association for Cancer Research. 2010;16:5189-99. 19. Spelman K, Burns J, Nichols D, Winters N, Ottersberg S, Tenborg M. Modulation of cytokine expression by traditional medicines: a review of herbal immunomodulators. Alternative medicine

25

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

review : a journal of clinical therapeutic. 2006;11:128-50. 20. Chen XC, Xia L, Hu S, Huang G. Inhibitory effects of Acanthopanax gracilistylus saponins on human platelet aggregation and liberation in vitro. Zhongguo yao li xue bao = Acta pharmacologica Sinica. 1996;17:523-6. 21. Isomoto H, Mott JL, Kobayashi S, Werneburg NW, Bronk SF, Haan S, et al. Sustained IL-6/STAT-3 signaling in cholangiocarcinoma cells due to SOCS-3 epigenetic silencing. Gastroenterology. 2007;132:384-96. 22. Takada Y, Ichikawa H, Badmaev V, Aggarwal BB. Acetyl-11-keto-beta-boswellic acid potentiates apoptosis, inhibits invasion, and abolishes osteoclastogenesis by suppressing NF-kappa B and NF-kappa B-regulated gene expression. J Immunol. 2006;176:3127-40. 23. Vardanyan M, Melemedjian OK, Price TJ, Ossipov MH, Lai J, Roberts E, et al. Reversal of pancreatitis-induced pain by an orally available, interleukin-6 receptor antagonist. Pain. 2010;151:257-65. 24. Imada K, Leonard WJ. The Jak-STAT pathway. Molecular immunology. 2000;37:1-11. 25. Song LX, Turkson J, Karras JG, Jove R, Haura EB. Activation of Stat3 by receptor tyrosine kinases and cytokines regulates survival in human non-small cell carcinoma cells. Oncogene. 2003;22:4150-65. 26. Dimberg LY, Anderson CK, Camidge R, Behbakht K, Thorburn A, HL. On the TRAIL to successful cancer therapy? Predicting and counteracting resistance against TRAIL-based therapeutics. Oncogene. 2013;32:1341-50. 27. Lanuti P, Bertagnolo V, Pierdomenico L, Bascelli A, Santavenere E, Alinari L, et al. Enhancement of TRAIL cytotoxicity by AG-490 in human ALL cells is characterized by downregulation of cIAP-1 and cIAP-2 through inhibition of Jak2/Stat3. Cell Res. 2009;19:1079-89. 28. Wang J, Lu J, Lv C, Xu T, Jia L. Three new triterpenoid saponins from root of Gardenia jasminoides Ellis. Fitoterapia. 2012;83:1396-401. 29. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646-74. 30. Sung B, B, Yadav VR, Aggarwal BB. Celastrol, a triterpene, enhances TRAIL-induced apoptosis through the down-regulation of cell survival proteins and up-regulation of death receptors. The Journal of biological chemistry. 2010;285:11498-507. 31. Sansone P, Bromberg J. Targeting the interleukin-6/Jak/stat pathway in human malignancies. J Clin Oncol. 2012;30:1005-14. 32. Hayashi M, Rho MC, Fukami A, Enomoto A, Nonaka S, Sekiguchi Y, et al. Biological activity of a novel nonpeptide antagonist to the interleukin-6 receptor 20S,21-epoxy-resibufogenin-3-formate. The Journal of pharmacology and experimental therapeutics. 2002;303:104-9. 33. Hong SS, Choi JH, Lee SY, Park YH, Park KY, Lee JY, et al. A Novel Small-Molecule Inhibitor Targeting the IL-6 Receptor beta Subunit, Glycoprotein 130. J Immunol. 2015;195:237-45. 34. Wang Y, Ma X, Yan S, Shen S, Zhu H, Gu Y, et al. 17-hydroxy-jolkinolide B inhibits signal transducers and activators of transcription 3 signaling by covalently cross-linking Janus kinases and induces apoptosis of human cancer cells. Cancer research. 2009;69:7302-10. 35. Yang J, Cai X, Lu W, Hu C, Xu X, Yu Q, et al. Evodiamine inhibits STAT3 signaling by inducing phosphatase shatterproof 1 in hepatocellular carcinoma cells. Cancer letters. 2013;328:243-51. 36. Bharadwaj U, Eckols TK, Kolosov M, Kasembeli MM, Adam A, Torres D, et al. Drug-repositioning screening identified piperlongumine as a direct STAT3 inhibitor with potent activity against breast cancer. Oncogene. 2015;34:1341-53.

26

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

37. Shin DS, Jung SN, Yun J, Lee CW, Han DC, Kim B, et al. Inhibition of STAT3 activation by KT-18618 via the disruption of the interaction between JAK3 and STAT3. Biochemical pharmacology. 2014;89:62-73. 38. Dong Y, Yin S, Li J, Jiang C, Ye M, Hu H. Bufadienolide compounds sensitize human breast cancer cells to TRAIL-induced apoptosis via inhibition of STAT3/Mcl-1 pathway. Apoptosis : an international journal on programmed cell death. 2011;16:394-403. 39. Jiang XP, Yang DC, Elliott RL, Head JF. Down-regulation of expression of interleukin-6 and its receptor results in growth inhibition of MCF-7 breast cancer cells. Anticancer Res. 2011;31:2899-906. 40. Krueger A, Schmitz I, Baumann S, Krammer PH, Kirchhoff S. Cellular FLICE-inhibitory protein splice variants inhibit different steps of caspase-8 activation at the CD95 death-inducing signaling complex. The Journal of biological chemistry. 2001;276:20633-40. 41. Ivanov VN, Zhou H, Partridge MA, Hei TK. Inhibition of ataxia telangiectasia mutated kinase activity enhances TRAIL-mediated apoptosis in human melanoma cells. Cancer research. 2009;69:3510-9.

Figure legend

Figure 1. CS-Ⅳa-Be induces cancer cell apoptosis. (A) The chemical structure of

CS-Ⅳa-Be. (B) LO2, HepG2, MHCC97L, SGC-7901, TF-1, U937, MDA-MB-231,

MCF-7 cells were treated with various concentrations of CS-Ⅳa-Be for 24 h and then

subjected to MTT assay, the value of IC50 was calculated. (C) Cell lysates from

MDA-MB-231 treated with various concentrations (0, 5, 7.5, 10) μM of CS-Ⅳa-Be

for 24 h were analyzed for apoptosis proteins. MDA-MB-231 cells were treated with

various concentrations (0, 7.5, 10) μM of CS-Ⅳa-Be for 24 h following being treated

by Z-VAD-FMK for 30 min, and subjected to MTT assay (D) and apoptosis assay (E).

(F) MDA-MB-231 cells treated with various concentrations (0, 5, 7.5, 10) μM of CS-

Ⅳa-Be for 24 h were stained with JC-1 and then subjected to cytometry assay. This is

a representative result of three repetitive experiments with similar results and error

bars mark SDs (*, P < 0.05).

27

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 2. CS-Ⅳa-Be inhibits IL-6/STAT3 activation in MDA-MB-231 cells. (A)

Cell lysates from MDA-MB-231 cells treated with various concentrations (0, 5, 7.5,

10) μM of CS-Ⅳa-Be for 24 h were analyzed for pSTAT3 (Tyr705), STAT3,

pSTAT1(Tyr701), STAT1, pSTAT5, STAT5, pAKT, AKT, pERK1/2 and ERK1/2,

GAPDH as a loading control. (B) Cell lysates from MDA-MB-231 cells treated with

various concentrations (0, 2.5, 5, 7.5, 10) μM of CS-Ⅳa-Be for 8 h, followed by 15

min IL-6 (25 ng/mL) stimulation were analyzed for pSTAT3 (Tyr705), STAT3, pJAK1,

pJAK2, pSrc, pAKT, AKT, pERK1/2 and ERK1/2, GAPDH as loading control. (C)

Cell lysates from MDA-MB-231 cells treated with 10 μM of CS-Ⅳa-Be for 8 h,

followed by 15 min stimulation of 25 ng/mL IL-11, LIF, OSM respectively were

analyzed for pSTAT3 (Tyr705), STAT3, GAPDH as loading control. (D) Cell lysates

from MDA-MB-231 cells treated with 10 μM of CS-Ⅳa-Be for 8 h, followed by 15

min stimulation of 100 ng/mL EGF and IFN-γ respectively were analyzed for

pSTAT3 (Tyr705), STAT3, pSTAT1(Tyr701), STAT1, pSTAT5 and STAT5, GAPDH

as loading control. This is a representative result of three repetitive experiments with

similar results.

Figure 3. CS-Ⅳa-Be inhibits IL-6/STAT3 downstream signaling.

(A) Immunofluorescence staining of pSTAT3 (Tyr705). MDA-MB-231 cells were

treated with 5 μM CS-Ⅳa-Be for 8 hours and then pSTAT3(Tyr705) was labeled

through immunofluorescence staining, scale bar indicates 20 μm. (B) MDA-MB-231

cells were treated with various concentrations (0, 5, 7.5, 10) μM of CS-Ⅳa-Be for 24

28

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

h, after which nuclear proteins were extracted and subjected to EMSA with STAT3

probe. (C) MDA-MB-231 cells transfected with STAT3 luciferase reporter vector for

24 h, and then treated with various concentrations (0, 2.5, 5, 7.5, 10) μM of CS-Ⅳ

a-Be for 8 h, the STAT3 luciferase activity were measured following stimulation of

IL-6 (25 ng/mL) for 8 h. (D) Cell lysates from MDA-MB-231 cells treated with

various concentrations (0, 5, 7.5, 10) μM of CS-Ⅳa-Be for 24 h were analyzed for

Survivin, c-FLIP, XIAP, Bcl-xL and Mcl-1, GAPDH as a loading control. (E) Cell

lysates from MDA-MB-231 cells treated with various concentrations (0, 2.5, 5, 7.5,

10) μM of CS-Ⅳa-Be for 24 h, followed by IL-6 (25 ng/mL) stimulation for 15 min

were analyzed for Survivin, c-FLIP, XIAP, Bcl-xL and Mcl-1, GAPDH as a loading

control. (F) Inhibition of IL-6-induced proliferation of TF-1 cells by CS-Ⅳa-Be. TF-1

cells were incubated with IL-6 (25 ng/ml) in the presence of various concentrations (0,

2.5, 5, 7.5, 10, 12.5) μM of CS-Ⅳa-Be 24 h or 48 h, cell viability was measured by

MTT assay. This is a representative result of three repetitive experiments with similar

results and error bars mark SDs (*, P < 0.05).

Figure 4. The effect of CS-Ⅳa-Be on JAKs and other growth factor receptors in

vitro kinase enzymatic activity. (A) The effect of CS-Ⅳa-Be on JAK2, JAK3, TYK2

kinase enzymatic activity in vitro. (B) The effect of CS-Ⅳa-Be ( 25 μM and 50 μM )

on JAK1, JAK2, JAK3, Tyk2, Src, EGFR, FGFR1 and PDGFRα kinase activity in

vitro.

29

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 5. CS-Ⅳa-Be disrupts IL-6/IL-6Rα/GP130 interaction. Cytometry analysis

of the inhibitory effect of CS-Ⅳa-Be on IL-6-IL-6R interaction in U937 cells.

Biotinated-IL-6 was used to label cell surface IL-6R. (A) Left panel: the levels of cell

surface IL-6Rα in cells with CS-Ⅳa-Be treatment or not; (B) right panel: cell surface

IL-6-IL-6R binding levels in cells with CS-Ⅳa-Be treatment or not. (C) ELISA

analysis of the inhibitory effect of CS-Ⅳa-Be on IL-6-IL-6Rα binding in vitro. The

maximum binding of IL-6 (100 % bound) is defined as without CS-Ⅳa-Be while in

the effects of various concentrations of CS-Ⅳa-Be calculated as percentages of

maximum binding of IL-6. Microscale thermophoresis analysis of CS-Ⅳa-Be binding

to IL-6Rα, GP130, LeptinR and LIFR. (D) CS-Ⅳa-Be binding affinity to IL-6Rα. (E)

CS- Ⅳ a-Be binding affinity to GP130, LeptinR and LIFR respectively. (F)

Recombinant human IL-6Rα with His tag was incubated with 0, 5 or 10 μM CS-Ⅳa

-Be for 2 h, then mixed with recombinant human GP130 and IL-6,

immunoprecipitated with anti-His antibody. The precipitates were washed, suspended

in reducing sample buffer and immunoblotted with anti-His or anti-GP130 antibody.

These are representative results of three repetitive experiments with similar results.

Figure 6. CS- Ⅳ a-Be sensitized to TRAIL-induced cancer cell apoptosis.

MDA-MB-231 cells were treated with 7.5 μM CS-Ⅳa-Be, TRAIL (60 ng/mL) or

combination for 24 h, the cell morphology was recorded (A) cell apoptosis was

analyzed by Annexin V/PI double staining assay (B) and Western blot with PARP

antibody (C). (D) MDA-MB-231 and MCF-7 cells were treated with different

30

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

concentrations (0, 5, 7.5, 10) μM of CS-Ⅳa-Be for 24 h, and then subjected to

Western blot assay with DR5 antibody (up panel). MDA-MB-231 cells treated with

7.5 μM of CS-Ⅳa-Be for 24 h were subjected to cytometry assay with DR5 or DR4

antibodies, mouse IgG as an isotype control (down panel). (E) MDA-MB-231 cells

were transfected with scramble siRNA or DR5 siRNA for 24 h, treated with 7.5 μM

CS-Ⅳa-Be, 30 ng/mL TRAIL or in combination with for 24 h, cell apoptosis was

analyzed by Annexin V/PI double staining assay. These are representative results of

three repetitive experiments with similar results and error bars mark SDs (*, P <

0.05).

31

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 29, 2016; DOI: 10.1158/1535-7163.MCT-15-0551 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Chikusetsusaponin ?a butyl ester (CS-?a-Be), a novel IL-6R antagonist, inhibits IL-6/STAT3 signaling pathway and induces cancer cell apoptosis

Jie Yang, Shihui Qian, Xueting Cai, et al.

Mol Cancer Ther Published OnlineFirst February 29, 2016.

Updated version Access the most recent version of this article at: doi:10.1158/1535-7163.MCT-15-0551

Supplementary Access the most recent supplemental material at: Material http://mct.aacrjournals.org/content/suppl/2016/02/27/1535-7163.MCT-15-0551.DC1

Author Author manuscripts have been peer reviewed and accepted for publication but have not yet been Manuscript edited.

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://mct.aacrjournals.org/content/early/2016/02/27/1535-7163.MCT-15-0551. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from mct.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research.