Open Access

Glyceollins as novel targeted therapeutic for the treatment of triple-negative breast cancer

  • Authors:
    • Lyndsay V. Rhodes
    • Syreeta L. Tilghman
    • Stephen M. Boue
    • Shuchun  Wang
    • Hafez Khalili
    • Shannon E. Muir
    • Melyssa R. Bratton
    • Qiang Zhang
    • Guangdi  Wang
    • Matthew E. Burow
    • Bridgette M. Collins-Burow
  • View Affiliations

  • Published online on: October 24, 2011     https://doi.org/10.3892/ol.2011.460
  • Pages: 163-171
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The purpose of this study was to investigate the effects of glyceollins on the suppression of tumorigenesis in triple‑negative breast carcinoma cell lines. We further explored the effects of glyceollins on microRNA and protein expression in MDA‑MB‑231 cells. Triple‑negative (ER‑, PgR‑ and Her2/neu‑) breast carcinoma cells were used to test the effects of glyceollins on tumorigenesis in vivo. Following this procedure, unbiased microarray analysis of microRNA expression was performed. Additionally, we examined the changes in the proteome induced by glyceollins in the MDA‑MB‑231 cells. Tumorigenesis studies revealed a modest suppression of MDA‑MB‑231 and MDA‑MB‑468 cell tumor growth in vivo. In response to glyceollins we observed a distinct change in microRNA expression profiles and proteomes of the triple‑negative breast carcinoma cell line, MDA‑MB‑231. Our results demonstrated that the glyceollins, previously described as anti‑estrogenic agents, also exert antitumor activity in triple‑negative breast carcinoma cell systems. This activity correlates with the glyceollin alteration of microRNA and proteomic expression profiles.

Introduction

Breast cancer afflicts approximately 1 in 8 women and is a leading cause of cancer-related mortality. Expression profiles of breast cancer exhibit a systematic variation and allow for the classification of breast cancer into five main groups: two estrogen receptor (ER)-positive (luminal A and B) and three ER-negative groups (normal breast-like, HER2-positive, and ‘basal-like’) (1). The term ‘triple-negative breast cancer’ (TNBC) represents a heterogeneous group of diseases and clearly does not comprise a ‘single entity’ (1). Although triple-negative cancer is not a synonym for basal-like cancer, basal-like cancers are preferentially negative for ER and progesterone receptor (PR) and lack HER2 expression (1). Basal-like breast carcinomas consistently express genes generally found in normal basal/myoepithelial cells of the breast, including high-molecular-weight ‘basal’ cytokeratins (CK; CK5/6, CK14 and CK17), vimentin, p-cadherin, αB crystallin, fascin and caveolins 1 and 2 (1). While it is clear that not all TNBC cases are characterized by the basal-like phenotype and vice versa, microarray-based expression analysis has demonstrated a great deal of overlap (1,2). Clinical similarities also exist between triple-negative tumors and basal-like tumors, including a higher prevalence in African-American women, more frequent incidence in younger patients, and greater aggressiveness than other molecular subgroups (1,3,4).

Of all breast cancers diagnosed approximately 75–80% are positive for ER and/or PR expression and 15–20% are positive for Her2/neu (5). Although these subtypes of disease are potentially susceptible to endocrine therapy and targeted therapy, such as trastuzumab, the remaining 10–15% of breast cancers diagnosed as triple-negative [ER(−), PR(−) and Her2/neu(−)] do not have defined therapeutic targets (6). TNBC has an aggressive clinical history as is evident by its rapid progression to a metastatic phenotype as well as a shorter time to death from distant recurrence as compared to ER(+) disease (1). It is therefore critical to identify novel targets in this disease entity.

The flavonoid family of phytochemicals, particularly those derived from soy, has received attention regarding their estrogenic activity as well as their effects on human health and disease (710). The observation that soy phytochemicals decrease the risk of breast cancer indicates a potential for the anti-tumorigenic activity of these compounds (1113). Additionally, the ability of soy isoflavonoids to prevent carcinogen-induced mammary tumorigenesis further indicates the potential anti-tumorigenic effects of these compounds (1416). Notably, the amount and type of isoflavonoid present in soy can be readily altered in response to external stimuli (1720). We previously described an increased biosynthesis of the isoflavonoid phytoalexin compounds, glyceollins I, II and III, in soy plants grown under stressed conditions (elicited soy) (17,19,21).

We showed that glyceollins suppress the tumorigenesis of ER(+) and estrogen-dependent breast cancer systems, demonstrating a clear in vivo anti-estrogenic activity of glyceollins (22). Notably, during these studies we noted that in the absence of estrogen, glyceollin-treated tumors were significantly smaller than their negative control counterparts by day 14. This indicated that in addition to their anti-estrogenic activity, glyceollins may target ER-independent mechanisms regulating tumor cell proliferation and/or survival. In the present study, we evaluated from a biological approach the efficacy of glyceollins on TNBC tumorigenesis in immunocompromised Nu/Nu female mice. Additionally, we investigated the effects of glyceollins on microRNA (miR) expression in the triple-negative setting. In this study, we aimed to demonstrate that glyceollins act as a novel therapeutic agent in TNBC-suppressing tumorigenesis, regulating the expression of miR and altering the proteome of MDA-MB-231 cells.

Materials and methods

Cells and reagents

The MDA-MB-231 and MDA-MB-468 cell lines (human breast cancer negative for ER, PR and Her2/neu) were acquired from the American Type Culture Collection (Manassas, VA, USA) and cultured as previously described (2224). Glyceollin mixture was isolated as previously described (22).

Xenograft model of tumorigenesis

Nu/Nu immunocompromised female mice (4–6 weeks old) were obtained from Charles River Laboratories (Wilmington, MA, USA). The animals were allowed a period of adaptation in a sterile and pathogen-free environment with food and water ad libitum. Mice were divided into treatment groups of 5 mice each: MDA-MB-231 + dimethyl sulfoxide (DMSO), MDA-MB-231 + glyceollins, MDA-MB-468 + DMSO, MDA-MB-468 + glyceollins. MDA-MB-231 and MDA-MB-468 cells were harvested in the exponential growth phase using a phosphate-buffered saline (PBS)/EDTA solution and washed. Viable cells (5×106) in 50 μl of sterile PBS suspension were mixed with 100 μl Reduced Growth Factor Matrigel (BD Biosciences, Bedford, MA, USA). Cells were injected bilaterally into the mammary fat pad using 27½ gauge sterile syringes. All procedures in animals were carried out under anesthesia using a mix of isofluorane and oxygen delivered by mask. Drug treatment (50 mg/kg/day glyceollins in DMSO/PBS) or vehicle (DMSO/PBS) injections were administered intraperitoneally daily for 14 days after palpable tumors had formed (MDA-MB-231, day 10; MDA-MB-468, day 25).

Tumor size was measured every 2–3 days using digital calipers. The volume of the tumor was calculated using the formula: 4/3π LS2 (L = larger radius; S = shorter radius). At necroscopy animals were sacrificed by cervical dislocation after exposure to CO2. Tumors, uteri, livers, and lungs were removed and frozen in liquid nitrogen or fixed in 10% formalin for further analysis. All procedures involving these animals were conducted in compliance with State and Federal laws, standards of the US Department of Health and Human Services, and guidelines established by Tulane University Animal Care and Use Committee. The facilities and laboratory animals program of Tulane University are accredited by the Association for the Assessment and Accreditation of Laboratory Animal Care.

miR microarray

MDA-MD-231 cells were plated at a density of 2 million cells in 25 cm2 flasks in normal culture media (Dulbecco's modified Eagle's medium supplemented with 5% fetal bovine serum, 1% penicillin/streptomycin, 1% essential amino acids, 1% non-essential amino acids and 1% sodium pyruvate) and allowed to adhere overnight at 37°C, 5% CO2 and air. Cells were treated with glyceollins (10 μM) or DMSO for 18 h. Cells were harvested in PBS and collected by centrifugation, and total RNA was extracted using the miRNeasy kit (Qiagen) according to manufacturer's protocol. Enrichment for miRNA was not performed. Quantity and quality of RNA was determined by absorbance (260 and 280 nm). Microarray assay was performed by LC Sciences (Houston, TX, USA). The assay started from 5 μg total RNA sample, which was size fractionated using a YM-100 Microcon centrifugal filter (Millipore, Billerica, MA, USA) and the small RNAs (<300 nt) isolated were 3′-extended with a poly(A) tail using poly(A) polymerase. An oligonucleotide tag was then ligated to the poly(A) tail for later fluorescent dye staining; two different tags were used for the two RNA samples in dual-sample experiments. Hybridization was performed overnight on a μParaflo microfluidic chip using a micro-circulation pump (Atactic Technologies, Houston, TX, USA) (25,26). On the microfluidic chip, each detection probe consisted of a chemically modified nucleotide coding segment complementary to target miR (from miRBase, http://microrna.sanger.ac.uk/sequences/) or other RNA (control or customer defined sequences) and a spacer segment of polyethylene glycol to extend the coding segment away from the substrate. The detection probes were rendered by in situ synthesis using PGR (photogenerated reagent) chemistry. The hybridization melting temperatures were balanced by chemical modifications of the detection probes. Hybridization used 100 μl 6X SSPE buffer (0.90 M NaCl, 60 mM Na2HPO4, 6 mM EDTA, pH 6.8) containing 25% formamide at 34°C. After RNA hybridization, tag-conjugating Cy3 and Cy5 dyes were circulated through the microfluidic chip for dye staining. Fluorescence images were collected using a laser scanner (GenePix 4000B, Molecular Device, Sunnyvale, CA, USA) and digitized using Array-Pro image analysis software (Media Cybernetics, Bethesda, MD, USA). Data were analyzed by first subtracting the background and then normalizing the signals using a LOWESS (Locally-weighted Regression) filter (27). For two color experiments, the ratio of the two sets of detected signals (log2 transformed, balanced) and p-values of the t-test were calculated; differentially detected signals were those with p-values of <0.01. The array was performed using quadruplicate biological repeats. Full array data are available in Table I.

Table I

microRNA microarray results for MDA-MB-231 cells treated with glyceollin (10 μM) for 18 h.

Table I

microRNA microarray results for MDA-MB-231 cells treated with glyceollin (10 μM) for 18 h.

No.Reporter namep-valueGroup 1Group 2Log2 (G2/G1)


ControlGlyceollin


MeanMean
108hsa-miR-12681.09E-051,732918−0.92
156hsa-miR-130a3.61E-051,7112,9040.76
844hsa-miR-9404.32E-052047301.84
287hsa-miR-1974.59E-052,0181,015−0.99
277 hsa-miR-193a-5p5.45E-051,452866−0.75
332hsa-miR-228.15E-053,8075,7880.60
372hsa-miR-29b1.53E-042,6167,6921.56
351hsa-miR-251.68E-046,1614,963−0.31
808hsa-miR-8771.75E-041,424708−1.01
294hsa-miR-19b2.30E-043,1604,2530.43
345hsa-miR-23a*2.33E-041,205669−0.85
828hsa-miR-9232.80E-041,7431,243−0.49
748hsa-miR-6383.58E-042,7293,9460.53
621hsa-miR-542-5p3.66E-04430142−1.60
853hsa-miR-99a4.18E-0413,25716,3480.30
246hsa-miR-1854.43E-041,5761,182−0.41
446hsa-miR-361-5p4.92E-044,5533,574−0.35
378hsa-miR-301a5.19E-041,1761,4360.29
95hsa-miR-125b8.65E-0426,19223,016−0.19
375hsa-miR-29c1.03E-032,1574,4451.04
830hsa-miR-92a1.04E-0310,5089,547−0.14
789hsa-miR-7201.04E-032,1941,622−0.44
238hsa-miR-1821.17E-032,2161,493−0.57
492hsa-miR-423-5p1.22E-035,6313,810−0.56
78hsa-miR-12461.28E-03561368−0.61
774hsa-miR-6631.49E-031,0232,1021.04
282hsa-miR-1951.62E-032175431.33
32hsa-miR-10a2.23E-031,375762−0.85
513hsa-miR-4542.58E-031,8551,070−0.79
235hsa-miR-181c2.64E-038921,8061.02
212hsa-miR-151-5p2.96E-038,7287,418−0.23
272hsa-miR-19153.17E-031,1351,4700.37
407hsa-miR-320c3.31E-039,7128,731−0.15
397hsa-miR-30d3.51E-035,6696,7530.25
176hsa-miR-1383.83E-033,5884,7960.42
315hsa-miR-213.96E-0321,13318,983−0.15
211hsa-miR-151-3p4.09E-033,8112,996−0.35
515hsa-miR-455-3p4.21E-031,6661,068−0.64
523hsa-miR-486-5p4.50E-03777389−1.00
237hsa-miR-181d4.51E-035402,6912.32
363hsa-miR-28-5p5.90E-039831,1370.21
391hsa-miR-30a*5.98E-031,4521,015−0.52
439hsa-miR-34a6.31E-031,3071,6440.33
31hsa-miR-1076.42E-037,2878,2000.17
356hsa-miR-26b6.45E-033,0133,9020.37
405hsa-miR-320a6.58E-0310,4099,155−0.19
124hsa-miR-12807.49E-034,1555,2450.34
8hsa-let-7d*7.73E-03754497−0.60
38hsa-miR-11808.08E-03984501−0.97
292hsa-miR-19a8.08E-032086021.54
343hsa-miR-2248.83E-032,6011,362−0.93
686hsa-miR-5849.53E-032,4781,818−0.45

The following transcripts are statistically significant but have low signals (signal <500)

365hsa-miR-296-5p9.44E-041393041.13
125hsa-miR-12813.24E-032054401.10
352hsa-miR-25*3.32E-0315274−1.04
528hsa-miR-4894.29E-03460183−1.33
269hsa-miR-19134.37E-031262881.19
553hsa-miR-505*5.53E-03340130−1.39
464hsa-miR-374b6.50E-032012820.49
778hsa-miR-6658.19E-03551601.54
245hsa-miR-1849.20E-0327436−2.93
Proteomics analysis

MDA-MB-231 cells were treated with DMSO or glyceollin-mix (10 μM) for 18 h. Cells were harvested and run using 2D-electrophoresis. The first dimensional electrophoresis was performed using a Protean IEF cell unit (BioRad, Hercules, CA, USA). Precast 11-cm IPG strips with a pH range of 5–8 were used to separate the proteins based on their isoelectric pH. The second dimensional electrophoresis was carried out in a BioRad Criterion electrophoresis cell system. Stained gels were scanned with a Gel Doc-XR image system (BioRad) and analyzed with the PDQuest software (version 8.01). The proteins of interest were marked for excision and excised from gels using a Quest Spot cutter (BioRad), digested and analyzed by an LC-Nanospray-MS system. The tandem MS spectra were analyzed against the ipi.human.v3.27 database using SEQUEST software and tabulated.

Statistical analysis

Studies were analyzed by the unpaired Student's t-test (Graph Pad Prism V.4) and p-values of <0.05 were considered statistically significant.

Results and Discussion

Glyceollins partially suppress growth of triple-negative breast tumor growth in vivo

To determine the therapeutic relevance of glyceollins in the triple-negative setting, MDA-MB-231 and MDA-MB-468 cells were used in an in vivo xenograft model of tumorigenesis. Immunocompromised female nude mice were injected in the mammary fat pad (MFP) with either MDA-MB-231 (Fig. 1A) or MDA-MB-468 (Fig. 1B) cells mixed with reduced growth factor matrigel. After palpable tumor formation (MDA-MB-231, day 10; MDA-MB-468, day 25), mice were randomized into treatment groups (n=5) and treated with vehicle or glyceollins (50 mg/kg/day). Tumor volume of MDA-MB-231 and MDA-MB-468 cells treated with glyceollins showed decreased tumor growth compared to vehicle-treated control tumors at endpoint analysis (Fig. 1, 64.36±21.29 mm3 and 58.16±11.28 mm3, respectively). These results demonstrate the tumor-suppressive effects of glyceollins on triple-negative breast carcinoma cell lines and indicate the clinical significance and therapeutic potential of glyceollins in the TNBC.

Glyceollins alter the miRnome of triple-negative breast carcinoma cells consistent with tumor-suppressive effects

Altered miR expression is common among a number of types of cancer and this dysregulation is known to promote tumorigenesis, hormone independence and drug resistance, epithelial-mesenchymal transition (EMT) and metastasis (2839). miR microarray analysis of MDA-MB-231 cells treated with glyceollins for 18 h revealed a number of changes in the miRNA expression profile compared to vehicle treated cells. Fig. 2 shows a heat map of miR expression changes for 4 independent samples. Tables II and III show the miRs found to have a significantly altered expression (increased or decreased, respectively) in response to treatment with glyceollins (p<0.01). A number of the miRs demonstrating a significantly increased expression following treatment with glyceollins have been characterized as tumor suppressers inhibiting cell cycle and proliferation (miR-181c/d), EMT and metastasis (miR-22, 29b/c, 30d, 34a, 195), or directly targeting known oncogenes (miR-26b). Those miRs with a significantly decreased expression induced by glyceollins have been identified as oncomiRs with roles in promoting tumorigenesis (miR-21, 193-5p) and metastasis (miR-185, 224).

Table II

microRNA with increased expression following glyceollin treatment.

Table II

microRNA with increased expression following glyceollin treatment.

miRNAMean fold-changep-valuemiRNAMean fold-changep-value
19a2.91<0.01130a1.69<0.001
19b1.35<0.001301a1.22<0.001
221.52<0.0011381.34<0.01
26b1.29<0.01181c2.03<0.01
181d4.99<0.01
28-5p1.16<0.011952.51<0.01
29b2.95<0.0016381.44<0.001
29c2.06<0.01
30d1.19<0.016632.06<0.01
34a1.26<0.019403.58<0.001
99a1.23<0.00112801.27<0.01
1071.13<0.0119151.29<0.01

Table III

microRNA with decreased expression following glyceollin treatment.

Table III

microRNA with decreased expression following glyceollin treatment.

miRNAMean fold-changep-valuemiRNAMean fold-changep-value
10a−1.80<0.01361-5p−1.27<0.001
21−1.11<0.01423-5p−1.47<0.01
23a*−1.80<0.001454−1.73<0.01
25−1.24<0.001455-3p−1.56<0.01
30a*−1.43<0.01486-5p−2.00<0.01
92a−1.10<0.01542-5p−3.03<0.001
125b−1.14<0.001584−1.37<0.01
151-3p−1.27<0.01720−1.36<0.01
151-5p−1.17<0.01
182−1.48<0.01877−2.01<0.001
185−1.33<0.001923−1.40<0.001
193a-5p−1.68<0.0011180−1.96<0.01
197−1.99<0.0011246−1.53<0.01
224−1.91<0.011268−1.89<0.001
320a−1.14<0.01 let-7da−1.52<0.01
320c−1.11<0.01

Among the most highly expressed miRs following treatment with glyceollins were miR-19a/b, 22, 29b/c, 181c/d, 195, 663 and 940. Notably, a number of the miRs that demonstrated glyceollin-induced expression have previously been documented as having tumor-suppressive effects. For example, miR-22 has been classified as a tumor-suppressive miR in metastatic breast cancers, as it has been shown to target oncogenes EVI-1, ERBB3 and CDC25C (40), as well the pro-metastatic gene EZR in ovarian cancer (41,42). miR-26b inhibits glioma tumor cell proliferation, survival, and migration by directly targeting EPHA2 (43). Further evidence of the tumor-suppressive nature of miR-26b include its ability to induce apoptosis via repression of SLC7A11 and the decreased expression of miR-26b in breast carcinoma patient samples (44).

miR-29b/c have been shown to directly inhibit the cell cycle transcription factor MYBL2 and in turn induce tumor cell senescence (45). miR-29 also plays a role in maintaining adequate cell adhesion by regulating extracellular matrix proteins (46) including collagens (47,48) and elastin (49), and regulates cell survival by targeting the anti-apoptotic MCL1 (50). Furthermore, miR-29 has been shown to induce expression of the tumor suppressor p53 by inhibiting the Rho-GTPase CDC42 (51).

Decreased expression of miR-181c due to hypermethylation has been observed in gastric carcinoma and its targets include the oncogenes NOTCH4 and KRAS (52). Additionally, miR-195 expression is significantly decreased in breast carcinoma patient samples (53), and a decreased expression of miR-195 has been correlated with decreased survival and increased metastasis in colorectal cancers (54). Direct targets of miR-195 include the oncogene RAF1, cell cycle regulators CCND1 (53) and CCNE1 (55), as well as the anti-apoptotic BCL2 (56).

The remaining two miRs with fold changes >2 have also been found to play anti-tumorigenic roles in cancer. miR-663 inhibits AP-1 activity by directly targeting JunB and JunD (57) and has been described as a tumor suppressor in gastric cancer (58). In silico predicted targets (TargetScan and miRANDA) of miR-940 include RhoA, a prominent mediator of invasion and metastasis.

Notably, although miR-19 is often referred to as an oncomiR due to its inclusion in the miR-17–92 oncogenic cluster, Zhang et al have recently demonstrated the ability of miR-19 to directly target tissue factor (TF), a known promoter of cancer cell survival, angiogenesis, and metastasis (59). Therefore, miR-19 may also play a tumor-suppressive role in breast cancer.

Among the most downregulated miRs following treatment with glyceollins were 193a-5p, 197, 224, 486-5p, and 542-5p, all of which have been associated with cancer progression. For instance, miR-193a-5p has been shown to target pro-apoptotic p73 and limit the effects of chemotherapy (60), while the oncomiR miR-197 has been shown to directly target the tumor suppressor, FUS1 (61). miR-224 has been associated with cancer progression (62) and enhanced cell migration and invasion by increasing the expression of the pro-invasive PAK4 and MMP-9 (63). Additionally, miR-224 and miR-486-5p promote cell migration and invasion by targeting the tumor suppressor CD40 (64,65).

miR-542-5p expression has also been associated with maintenance of the mesenchymal phenotype (66), a key characteristic of the TNBC phenotype and driver of cell motility and invasiveness. The reversal of the mesenchymal phenotype to a more epithelial morphology through the process of mesenchymal-to-epithelial transition (MET) represents an area of high-impact research for the development of novel therapeutics.

Although not a marked change, treatment with glyceollins decreases the expression of miR-21. miR-21 is one of the most established and highly researched miRs for its oncogenic role in cancer (67), and has been shown to be highly overexpressed in TNBC (68). The expression of miR-21 in breast tumors has been associated with poor prognosis (31), development of drug resistance (69,70), and increased rate of recurrence (39). Targets of miR-21 include prominent tumor suppressors PTEN (71,72) and PDCD4 (73), as well as inhibitors of metastasis, such as TIMP3 (74) and TPM1 (75,76).

The function of the remaining two miRs downregulated more than 1.95-fold by glyceollins, miR-877 and 1180, has yet to be determined at the time of this publication. Although the function of these miRs is not currently known, putative targets predicted by TargetScan include p53 inducible nuclear protein 2 (TP53INP2) and the cell cycle regulator, CDC40 (putative targets for miR-877); a regulator of cell adhesion, PUNC, the pro-apoptotic gene, BAD and BAMBI, a negative regulator of TGFβ known to mediate cell transformation (putative targets of miR-1180).

Glyceollins alter the proteome of MDA-MB-231 breast carcinoma cells in a manner indicative of tumor suppressive effects

The treatment of MDA-MB-231 cells with glyceollins for 18 h generated distinct protein spot patterns as analyzed by 2D-gel electrophoresis. Sequence analysis of selected spots revealed a number of proteins up- and downregulated by glyceollins (Table IV). While each of these spots represents a target for validation and mechanistic analysis, two proteins identified are known to play key roles in breast cancer tumorigenesis and progression. We observed an almost 25-fold upregulation of NME1 (NM23-H1) by glyceollin treatment. NME1 is a known metastasis suppressor gene (77) and is a putative target of two miRs with a significantly decreased expression following treatment with glyceollins, miR-486-5p and miR-542-5p. Notably, as mentioned above, miR-542-5p expression has been linked to the maintenance of the mensenchymal phenotype. Decreased expression of miR-542-5p, as well as an increased expression of NME1, indicates a reversal of EMT and a suppression of metastasis by glyceollins. A second target identified via our proteomics approach is vimentin, whose expression was downregulated more than 13-fold by glyceollins. Vimentin is a marker for epithelial-to-mesenchymal transition and is highly expressed in numerous TNBCs and cell lines including MDA-MB-231 (78). Vimentin is a proven target of miR-30d (79) and a predicted target of miR-138, both found to be significantly increased by glyceollins. These data suggest that the effects of glyceollins on TNBC cell lines are achieved via regulation of miRs, which in turn regulate known oncogenes and tumor suppressors. Taken together, these data indicate that treatment of TNBC cells with glyceollins inhibit tumorigenesis and induce a miR expression profile correlative to a less aggressive phenotype.

Table IV

Effects of glyceollins on the MDA-MB-231 cell proteome.

Table IV

Effects of glyceollins on the MDA-MB-231 cell proteome.

SpotGene symbolGene nameMean intensity ratiop-value
203EEF1DElongation factor 1-δ36.50<0.01
1004ARHGDIARho GDP-dissociation inhibitor 1115.85<0.001
2102CLIC1Chloride intracellular channel protein 1129.94<0.001
2103TPD52L2Isoform 2 of tumor protein D5443.90<0.01
2204EIF2S1Eukaryotic translation initiation factor 2 subunit 155.62<0.01
3103CLIC4Chloride intracellular channel protein 444.37<0.01
5004NME1Non-metastatic cells 1 (NM23-H1)24.89<0.01
5304GIPC1PDZ domain-containing protein GIPC121.41<0.01
5405MAP2K2Mitogen-activated protein kinase kinase 229.96<0.01
5406TARDBPTAR DNA-binding protein 4329.96<0.01
7706VIMVimentin−13.84<0.01
9903DDX1ATP-dependent RNA helicase DDX1−34.75<0.001
9904KHSRPFar upstream element-binding protein 2−31.31<0.01
2304SEC13SEC13-related protein−10.07<0.01
5506HNRPH1Heterogeneous nuclear ribonucleoprotein H1−28.63<0.001
7202HNRPH3Heterogeneous nuclear ribonucleoprotein H3−66.35<0.01
8404HNRPDHeterogeneous nuclear ribonucleoprotein D0−15.28<0.001
3605FKBP4FK506-binding protein−17.83<0.01

In conclusion, the results from our study demonstrate the ability of glyceollins to inhibit tumor growth of the triple-negative breast carcinoma cell lines, MDA-MB-231 and MDA-MB-468. Furthermore, it is known that the dysregulation of miR expression is a characteristic of numerous cancer cancer types including breast carcinoma. miR microarray analysis of MDA-MB-231 cells treated with glyceollins revealed significant alterations of the miR expression profile consistent with a less aggressive phenotype.

Acknowledgements

This study was supported by: The US Department of Agriculture 58-6435-7-019 (S.M.B. and M.E.B.), the National Institutes of Health/National Center for Research Resources P20RR020152 (B.M.C.-B.), National Center for Research Resources RCMI program through Grant 5G12RR026260-02 (G.W.), and the Office of Naval Research Grant 09-10 N00014-10-1-0270 (M.E.B.).

References

1 

Reis-Filho JS and Tutt AN: Triple negative tumours: a critical review. Histopathology. 52:108–118. 2008. View Article : Google Scholar

2 

Hu Z, Fan C, Oh DS, et al: The molecular portraits of breast tumors are conserved across microarray platforms. BMC Genomics. 7:962006. View Article : Google Scholar : PubMed/NCBI

3 

Bauer KR, Brown M, Cress RD, Parise CA and Caggiano V: Descriptive analysis of estrogen receptor (ER)-negative, progeste- rone receptor (PR)-negative, and HER2-negative invasive breast cancer, the so-called triple-negative phenotype: a popu- lation-based study from the California cancer registry. Cancer. 109:1721–1728. 2007. View Article : Google Scholar

4 

Dent R, Trudeau M, Pritchard KI, et al: Triple-negative breast cancer: clinical features and patterns of recurrence. Clin Cancer Res. 13:4429–4434. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Gail MH, Anderson WF, Garcia-Closas M and Sherman ME: Absolute risk models for subtypes of breast cancer. J Natl Cancer Inst. 99:1657–1659. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Reis-Filho JS, Westbury C and Pierga JY: The impact of expression profiling on prognostic and predictive testing in breast cancer. J Clin Pathol. 59:225–231. 2006. View Article : Google Scholar : PubMed/NCBI

7 

Messina M: A brief historical overview of the past two decades of soy and isoflavone research. J Nutr. 140:1350S–1354S. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Murkies AL, Wilcox G and Davis SR: Clinical review 92: Phytoestrogens. J Clin Endocrinol Metab. 83:297–303. 1998. View Article : Google Scholar : PubMed/NCBI

9 

Tham DM, Gardner CD and Haskell WL: Clinical review 97: Potential health benefits of dietary phytoestrogens: a review of the clinical, epidemiological, and mechanistic evidence. J Clin Endocrinol Metab. 83:2223–2235. 1998.PubMed/NCBI

10 

Humfrey CD: Phytoestrogens and human health effects: weighing up the current evidence. Nat Toxins. 6:51–59. 1998. View Article : Google Scholar : PubMed/NCBI

11 

Collins-Burow BM, Burow ME, Duong BN and McLachlan JA: Estrogenic and antiestrogenic activities of flavonoid phytochemicals through estrogen receptor binding-dependent and -independent mechanisms. Nutr Cancer. 38:229–244. 2000. View Article : Google Scholar

12 

Collins BM, McLachlan JA and Arnold SF: The estrogenic and antiestrogenic activities of phytochemicals with the human estrogen receptor expressed in yeast. Steroids. 62:365–372. 1997. View Article : Google Scholar : PubMed/NCBI

13 

Fournier DB, Erdman JW Jr and Gordon GB: Soy, its components, and cancer prevention: a review of the in vitro, animal, and human data. Cancer Epidemiol Biomarkers Prev. 7:1055–1065. 1998.PubMed/NCBI

14 

Barnes S: The chemopreventive properties of soy isoflavonoids in animal models of breast cancer. Breast Cancer Res Treat. 46:169–179. 1997. View Article : Google Scholar : PubMed/NCBI

15 

Lamartiniere CA, Zhang JX and Cotroneo MS: Genistein studies in rats: potential for breast cancer prevention and reproductive and developmental toxicity. Am J Clin Nutr. 68:1400S–1405S. 1998.PubMed/NCBI

16 

Diel P, Smolnikar K, Schulz T, Laudenbach-Leschowski U, Michna H and Vollmer G: Phytoestrogens and carcinogenesis-differential effects of genistein in experimental models of normal and malignant rat endometrium. Hum Reprod. 16:997–1006. 2001. View Article : Google Scholar : PubMed/NCBI

17 

Boue SM, Carter CH, Ehrlich KC and Cleveland TE: Induction of the soybean phytoalexins coumestrol and glyceollin by Aspergillus. J Agric Food Chem. 48:2167–2172. 2000. View Article : Google Scholar : PubMed/NCBI

18 

Bhattacharyya M and Ward EWB: Resistance, susceptibility and accumulation of glyceollins I-III in soybean organs inoculated with Phytophthora megasperma f. sp. Glycinea. Physiol and Mol Plant Pathol. 29:227–237. 1986. View Article : Google Scholar

19 

Boue SM, Wiese TE, Nehls S, Burow ME, Elliott S, Carter-Wientjes CH, Shih BY, McLachlan JA and Cleveland TE: Evaluation of the estrogenic effects of legume extracts containing phytoestrogens. J Agric Food Chem. 51:2193–2199. 2003. View Article : Google Scholar : PubMed/NCBI

20 

Graham TL and Graham MY: Glyceollin elicitors induce major but distinctly different shifts in isoflavonoid metabolism in proximal and distal soybean cell populations. Mol Plant Microbe Interact. 4:60–68. 1991. View Article : Google Scholar

21 

Burow ME, Boue SM, Collins-Burow BM, et al: Phytochemical glyceollins, isolated from soy, mediate antihormonal effects through estrogen receptor alpha and beta. J Clin Endocrinol Metab. 86:1750–1758. 2001.

22 

Salvo VA, Boue SM, Fonseca JP, et al: Antiestrogenic glyceollins suppress human breast and ovarian carcinoma tumorigenesis. Clin Cancer Res. 12:7159–7164. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Rhodes LV, Muir SE, Elliott S, et al: Adult human mesenchymal stem cells enhance breast tumorigenesis and promote hormone independence. Breast Cancer Res Treat. 121:293–300. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Burow ME, Tang Y, Collins-Burow BM, et al: Effects of environmental estrogens on tumor necrosis factor alpha-mediated apoptosis in MCF-7 cells. Carcinogenesis. 20:2057–2061. 1999. View Article : Google Scholar : PubMed/NCBI

25 

Gao X, Gulari E and Zhou X: In situ synthesis of oligonucleotide microarrays. Biopolymers. 73:579–596. 2004. View Article : Google Scholar : PubMed/NCBI

26 

Zhu Q, Hong A, Sheng N, et al: microParaflo biochip for nucleic acid and protein analysis. Methods Mol Biol. 382:287–312. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Bolstad BM, Irizarry RA, Astrand M and Speed TP: A comparison of normalization methods for high density oligonucleotide array data based on variance and bias. Bioinformatics. 19:185–193. 2003. View Article : Google Scholar : PubMed/NCBI

28 

Farazi TA, Spitzer JI, Morozov P and Tuschl T: miRNAs in human cancer. J Pathol. 223:102–115. 2011. View Article : Google Scholar

29 

Winter J and Diederichs S: MicroRNA biogenesis and cancer. Methods Mol Biol. 676:3–22. 2011. View Article : Google Scholar

30 

Allen KE and Weiss GJ: Resistance may not be futile: microRNA biomarkers for chemoresistance and potential therapeutics. Mol Cancer Ther. 9:3126–3136. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Gandellini P, Profumo V, Folini M and Zaffaroni N: MicroRNAs as new therapeutic targets and tools in cancer. Expert Opin Ther Targets. 15:265–279. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Nana-Sinkam SP and Croce CM: MicroRNA dysregulation in cancer: opportunities for the development of microRNA-based drugs. IDrugs. 13:843–846. 2011.PubMed/NCBI

33 

Ma L and Weinberg RA: MicroRNAs in malignant progression. Cell Cycle. 7:570–572. 2008. View Article : Google Scholar

34 

O'Day E and Lal A: MicroRNAs and their target gene networks in breast cancer. Breast Cancer Res. 12:2012011.

35 

Gregory PA, Bracken CP, Bert AG and Goodall GJ: MicroRNAs as regulators of epithelial-mesenchymal transition. Cell Cycle. 7:3112–3118. 2008. View Article : Google Scholar : PubMed/NCBI

36 

Korpal M and Kang Y: The emerging role of miR-200 family of microRNAs in epithelial-mesenchymal transition and cancer metastasis. RNA Biol. 5:115–119. 2008. View Article : Google Scholar : PubMed/NCBI

37 

Chen J, Wang L, Matyunina LV, Hill CG and McDonald JF: Overexpression of miR-429 induces mesenchymal-to-epithelial transition (MET) in metastatic ovarian cancer cells. Gynecol Oncol. 121:200–205. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Tryndyak VP, Beland FA and Pogribny IP: E-cadherin transcriptional down-regulation by epigenetic and microRNA-200 family alterations is related to mesenchymal and drug-resistant phenotypes in human breast cancer cells. Int J Cancer. 126:2575–2583. 2011.PubMed/NCBI

39 

Ota D, Mimori K, Yokobori T, et al: Identification of recurrence-related microRNAs in the bone marrow of breast cancer patients. Int J Oncol. 38:955–962. 2011.PubMed/NCBI

40 

Patel JB, Appaiah HN, Burnett RM, et al: Control of EVI-1 oncogene expression in metastatic breast cancer cells through microRNA miR-22. Oncogene. 30:1290–1301. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Li J, Liang S, Yu H, Zhang J, Ma D and Lu X: An inhibitory effect of miR-22 on cell migration and invasion in ovarian cancer. Gynecol Oncol. 119:543–548. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Li J, Liang SH and Lu X: Potential role of ezrin and its related microRNA in ovarian cancer invasion and metastasis. Zhonghua Fu Chan Ke Za Zhi. 45:787–792. 2011.PubMed/NCBI

43 

Wu N, Zhao X, Liu M, et al: Role of microRNA-26b in glioma development and its mediated regulation on EphA2. PLoS One. 6:e162642011. View Article : Google Scholar : PubMed/NCBI

44 

Liu XX, Li XJ, Zhang B, et al: MicroRNA-26b is underexpressed in human breast cancer and induces cell apoptosis by targeting SLC7A11. FEBS Lett. 585:1363–1367. 2011. View Article : Google Scholar : PubMed/NCBI

45 

Martinez I, Cazalla D, Almstead LL, Steitz JA and DiMaio D: miR-29 and miR-30 regulate B-Myb expression during cellular senescence. Proc Natl Acad Sci USA. 108:522–527. 2011. View Article : Google Scholar : PubMed/NCBI

46 

Villarreal G Jr, Oh DJ, Kang MH and Rhee DJ: Coordinated Regulation of Extracellular Matrix Synthesis by the MicroRNA-29 Family in the Trabecular Meshwork. Invest Ophthalmol Vis Sci. 52:3391–3397. 2011. View Article : Google Scholar : PubMed/NCBI

47 

Maurer B, Stanczyk J, Jungel A, et al: MicroRNA-29, a key regulator of collagen expression in systemic sclerosis. Arthritis Rheum. 62:1733–1743. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Li Z, Hassan MQ, Jafferji M, et al: Biological functions of miR-29b contribute to positive regulation of osteoblast differentiation. J Biol Chem. 284:15676–15684. 2009. View Article : Google Scholar : PubMed/NCBI

49 

Ott CE, Grunhagen J, Jager M, et al: MicroRNAs differentially expressed in postnatal aortic development downregulate elastin via 3′ UTR and coding-sequence binding sites. PLoS One. 6:e162502011.PubMed/NCBI

50 

Mott JL, Kobayashi S, Bronk SF and Gores GJ: mir-29 regulates Mcl-1 protein expression and apoptosis. Oncogene. 26:6133–6140. 2007. View Article : Google Scholar : PubMed/NCBI

51 

Park SY, Lee JH, Ha M, Nam JW and Kim VN: miR-29 miRNAs activate p53 by targeting p85 alpha and CDC42. Nat Struct Mol Biol. 16:23–29. 2009. View Article : Google Scholar : PubMed/NCBI

52 

Hashimoto Y, Akiyama Y, Otsubo T, Shimada S and Yuasa Y: Involvement of epigenetically silenced microRNA-181c in gastric carcinogenesis. Carcinogenesis. 31:777–784. 2011. View Article : Google Scholar : PubMed/NCBI

53 

Li D, Zhao Y, Liu C, et al: Analysis of MiR-195 and MiR-497 expression, regulation and role in breast cancer. Clin Cancer Res. 7:1722–1730. 2011. View Article : Google Scholar : PubMed/NCBI

54 

Wang X, Wang J, Ma H, Zhang J and Zhou X: Downregulation of miR-195 correlates with lymph node metastasis and poor prognosis in colorectal cancer. Med Oncol. March 10–2011.(E-pub ahead of print).

55 

Sekiya Y, Ogawa T, Iizuka M, Yoshizato K, Ikeda K and Kawada N: Down-regulation of cyclin E1 expression by microRNA-195 accounts for interferon-beta-induced inhibition of hepatic stellate cell proliferation. J Cell Physiol. 226:2535–2542. 2011. View Article : Google Scholar : PubMed/NCBI

56 

Liu L, Chen L, Xu Y, Li R and Du X: microRNA-195 promotes apoptosis and suppresses tumorigenicity of human colorectal cancer cells. Biochem Biophys Res Commun. 400:236–240. 2011. View Article : Google Scholar : PubMed/NCBI

57 

Tili E, Michaille JJ, Adair B, et al: Resveratrol decreases the levels of miR-155 by upregulating miR-663, a microRNA targeting JunB and JunD. Carcinogenesis. 31:1561–1566. 2011. View Article : Google Scholar : PubMed/NCBI

58 

Pan J, Hu H, Zhou Z, et al: Tumor-suppressive mir-663 gene induces mitotic catastrophe growth arrest in human gastric cancer cells. Oncol Rep. 24:105–112. 2011.PubMed/NCBI

59 

Zhang X, Yu H, Lou JR, et al: MicroRNA-19 (miR-19) regulates tissue factor expression in breast cancer cells. J Biol Chem. 286:1429–1435. 2011. View Article : Google Scholar : PubMed/NCBI

60 

Ory B, Ramsey MR, Wilson C, et al: A microRNA-dependent program controls p53-independent survival and chemosensitivity in human and murine squamous cell carcinoma. J Clin Invest. 121:809–820. 2011. View Article : Google Scholar : PubMed/NCBI

61 

Du L, Schageman JJ, Subauste MC, et al: miR-93, miR-98, and miR-197 regulate expression of tumor suppressor gene FUS1. Mol Cancer Res. 7:1234–1243. 2009. View Article : Google Scholar : PubMed/NCBI

62 

Arndt GM, Dossey L, Cullen LM, et al: Characterization of global microRNA expression reveals oncogenic potential of miR-145 in metastatic colorectal cancer. BMC Cancer. 9:3742009. View Article : Google Scholar : PubMed/NCBI

63 

Li Q, Wang G, Shan JL, et al: MicroRNA-224 is upregulated in HepG2 cells and involved in cellular migration and invasion. J Gastroenterol Hepatol. 25:164–171. 2011. View Article : Google Scholar : PubMed/NCBI

64 

Mees ST, Mardin WA, Sielker S, et al: Involvement of CD40 targeting miR-224 and miR-486 on the progression of pancreatic ductal adenocarcinomas. Ann Surg Oncol. 16:2339–2350. 2009. View Article : Google Scholar : PubMed/NCBI

65 

Wingett DG, Vestal RE, Forcier K, Hadjokas N and Nielson CP: CD40 is functionally expressed on human breast carcinomas: variable inducibility by cytokines and enhancement of Fas-mediated apoptosis. Breast Cancer Res Treat. 50:27–36. 1998. View Article : Google Scholar : PubMed/NCBI

66 

Castilla MA, Moreno-Bueno G, Romero-Perez L, et al: Micro- RNA signature of the epithelial-mesenchymal transition in endometrial carcinosarcoma. J Pathol. 223:72–80. 2011. View Article : Google Scholar : PubMed/NCBI

67 

Bonci D: MicroRNA-21 as therapeutic target in cancer and cardiovascular disease. Recent Pat Cardiovasc Drug Discov. 5:156–161. 2011. View Article : Google Scholar : PubMed/NCBI

68 

Radojicic J, Zaravinos A, Vrekoussis T, Kafousi M, Spandidos DA and Stathopoulos EN: MicroRNA expression analysis in triple-negative (ER, PR and Her2/neu) breast cancer. Cell Cycle. 10:507–517. 2011. View Article : Google Scholar : PubMed/NCBI

69 

Mei M, Ren Y, Zhou X, et al: Downregulation of miR-21 enhances chemotherapeutic effect of taxol in breast carcinoma cells. Technol Cancer Res Treat. 9:77–86. 2011. View Article : Google Scholar : PubMed/NCBI

70 

Gong C, Yao Y, Wang Y, et al: Up-regulation of miR-21 mediates resistance to trastuzumab therapy for breast cancer. J Biol Chem. 286:19127–19137. 2011. View Article : Google Scholar : PubMed/NCBI

71 

Meng F, Henson R, Wehbe-Janek H, Ghoshal K, Jacob ST and Patel T: MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology. 133:647–658. 2007. View Article : Google Scholar : PubMed/NCBI

72 

Qi L, Bart J, Tan LP, et al: Expression of miR-21 and its targets (PTEN, PDCD4, TM1) in flat epithelial atypia of the breast in relation to ductal carcinoma in situ and invasive carcinoma. BMC Cancer. 9:1632009. View Article : Google Scholar : PubMed/NCBI

73 

Frankel LB, Christoffersen NR, Jacobsen A, Lindow M, Krogh A and Lund AH: Programmed cell death 4 (PDCD4) is an important functional target of the microRNA miR-21 in breast cancer cells. J Biol Chem. 283:1026–1033. 2008. View Article : Google Scholar : PubMed/NCBI

74 

Song B, Wang C, Liu J, et al: MicroRNA-21 regulates breast cancer invasion partly by targeting tissue inhibitor of metalloproteinase 3 expression. J Exp Clin Cancer Res. 29:292010. View Article : Google Scholar : PubMed/NCBI

75 

Zhu S, Si ML, Wu H and Mo YY: MicroRNA-21 targets the tumor suppressor gene tropomyosin 1 (TPM1). J Biol Chem. 282:14328–14336. 2007. View Article : Google Scholar : PubMed/NCBI

76 

Zhu S, Wu H, Wu F, Nie D, Sheng S and Mo YY: MicroRNA-21 targets tumor suppressor genes in invasion and metastasis. Cell Res. 18:350–359. 2008. View Article : Google Scholar : PubMed/NCBI

77 

Russell RL, Pedersen AN, Kantor J, et al: Relationship of nm23 to proteolytic factors, proliferation and motility in breast cancer tissues and cell lines. Br J Cancer. 78:710–717. 1998. View Article : Google Scholar : PubMed/NCBI

78 

Kokkinos MI, Wafai R, Wong MK, Newgreen DF, Thompson EW and Waltham M: Vimentin and epithelial-mesenchymal transition in human breast cancer – observations in vitro and in vivo. Cells Tissues Organs. 185:191–203. 2007.

79 

Joglekar MV, Patil D, Joglekar VM, et al: The miR-30 family microRNAs confer epithelial phenotype to human pancreatic cells. Islets. 1:137–147. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January 2012
Volume 3 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Rhodes LV, Tilghman SL, Boue SM, Wang S, Khalili H, Muir SE, Bratton MR, Zhang Q, Wang G, Burow ME, Burow ME, et al: Glyceollins as novel targeted therapeutic for the treatment of triple-negative breast cancer. Oncol Lett 3: 163-171, 2012
APA
Rhodes, L.V., Tilghman, S.L., Boue, S.M., Wang, S., Khalili, H., Muir, S.E. ... Collins-Burow, B.M. (2012). Glyceollins as novel targeted therapeutic for the treatment of triple-negative breast cancer. Oncology Letters, 3, 163-171. https://doi.org/10.3892/ol.2011.460
MLA
Rhodes, L. V., Tilghman, S. L., Boue, S. M., Wang, S., Khalili, H., Muir, S. E., Bratton, M. R., Zhang, Q., Wang, G., Burow, M. E., Collins-Burow, B. M."Glyceollins as novel targeted therapeutic for the treatment of triple-negative breast cancer". Oncology Letters 3.1 (2012): 163-171.
Chicago
Rhodes, L. V., Tilghman, S. L., Boue, S. M., Wang, S., Khalili, H., Muir, S. E., Bratton, M. R., Zhang, Q., Wang, G., Burow, M. E., Collins-Burow, B. M."Glyceollins as novel targeted therapeutic for the treatment of triple-negative breast cancer". Oncology Letters 3, no. 1 (2012): 163-171. https://doi.org/10.3892/ol.2011.460