- Research Paper
- Published:
Metformin and soybean-derived bioactive molecules attenuate the expansion of stem cell-like epithelial subpopulation and confer apoptotic sensitivity in human colon cancer cells
Genes & Nutrition volume 10, Article number: 49 (2015)
Abstract
Colorectal cancer (CRC) is a disease whose genesis may include metabolic dysregulation. Cancer stem cells are attractive targets for therapeutic interventions since their aberrant expansion may underlie tumor initiation, progression, and recurrence. To investigate the actions of metabolic regulators on cancer stem cell-like cells (CSC) in CRC, we determined the effects of soybean-derived bioactive molecules and the anti-diabetes drug metformin (MET), alone and together, on the growth, survival, and frequency of CSC in human HCT116 cells. Effects of MET (60 μM) and soybean components genistein (Gen, 2 μM), lunasin (Lun, 2 μM), β-conglycinin (β-con, 3 μM), and glycinin (Gly, 3 μM) on HCT116 cell proliferation, apoptosis, and mRNA/protein expression and on the frequency of the CSC CD133+CD44+ subpopulation by colonosphere assay and fluorescence-activated cell sorting/flow cytometry were evaluated. MET, Gen, and Lun, individually and together, inhibited HCT116 viability and colonosphere formation and, conversely, enhanced HCT116 apoptosis. Reductions in frequency of the CSC CD133+CD44+ subpopulation with MET, Gen, and Lun were found to be associated with increased PTEN and reduced FASN expression. In cells under a hyperinsulinemic state mimicking metabolic dysregulation and without and with added PTEN-specific inhibitor SF1670, colonosphere formation and frequency of the CD133+CD44+ subpopulation were decreased by MET, Lun and Gen, alone and when combined. Moreover, MET + Lun + Gen co-treatment increased the pro-apoptotic and CD133+CD44+-inhibitory efficacy of 5-fluorouracil under hyperinsulinemic conditions. Results identify molecular networks shared by MET and bioavailable soy food components, which potentially may be harnessed to increase drug efficacy in diabetic and non-diabetic patients with CRC.
Introduction
Colorectal cancer (CRC) remains a major contributor to cancer morbidity and mortality worldwide and is the third leading cause of cancer deaths in the USA (Siegel et al. 2015). While the molecular mechanisms by which CRC begins and then progresses to metastatic disease are better understood relative to some other cancer types (Tomasetti et al. 2014; Linnekamp et al. 2015), current treatments such as with the first-line chemotherapeutic drug 5-fluorouracil (5-FU) are largely ineffective once the tumor becomes invasive, metastatic and/or chemo-resistant (Paldino et al. 2014). Hence, preventing CRC initiation, progression and recurrence and defining bona fide targets for effective therapies are paramount for reducing CRC-related mortality.
Cancer stem cells (CSC) have emerged as attractive therapeutic targets to inhibit tumor initiation, chemo-resistance, and relapse in many cancer types (Paldino et al. 2014; Zeuner et al. 2014; White and Lowry 2015). According to the CSC hypothesis, a small population of normal stem cells requisite for maintaining tissue homeostasis/renewal can initiate a tumor upon acquiring one or more driver mutations (Chandler 2010). Moreover, a recent study (Tomasetti and Vogelstein 2015) has suggested that the lifetime risk of many different types of cancer is strongly correlated with the total number of divisions of these normal self-renewing cells, and that random mutations arising during DNA replication are likely causative for the emergence of CSC. Interestingly, among the different epithelial stem cells analyzed in that study, those derived from the colon were shown to undergo one of the highest numbers of cell divisions. Inhibiting the expansion of normal SC as well as CSC may thus constitute a useful approach for CRC prevention and treatment.
Diet is a well-accepted risk factor for CRC, the latter potentially mediated by bioactive and bioavailable components in foods that can influence colon epithelial cell growth and survival (Yan et al. 2010; Spector et al. 2013). Previous work in our laboratory demonstrated that dietary intake of soy (Xiao et al. 2008) and whey (Xiao et al. 2006) protein-based diets favorably influenced insulin-signaling components and reduced intestinal tumor incidence in rats exposed to the intestinal carcinogen azoxymethane. Further, we showed in the context of a high-fat diet that a soy protein-based dietary regimen suppressed intestinal epithelial cell proliferation and systemic insulin levels in mice (Al-Dwairi et al. 2012, 2014). The association between colonic intestinal proliferation and insulin is noteworthy, given that the anti-diabetic drug metformin (1,1-dimethylbiguanide hydrochloride; MET) has recently attracted much interest as a potential anticancer medication. Several epidemiological studies reported an inverse correlation between MET intake (at standard doses of 1500–2250 mg/day) and CRC incidence and related mortality in diabetic patients (Zhang et al. 2013; Sehdev et al. 2015). Further, MET at a low dose (250 mg/day) was found to reduce colon epithelial cell proliferation and numbers of aberrant crypt foci in non-diabetic patients (Dowling et al. 2011). Moreover, combinations of MET with conventional chemotherapeutic agents 5-fluorouracil and oxaliplatin were reported to decrease recurrence of colon tumors in a mouse xenograft model (Nangia-Makker et al. 2014). Nevertheless, the potential interactions of MET and bioactive dietary components with known anti-tumor activities for CRC prevention and treatment, and specifically in the context of targeting CSC, have not been delineated. The importance of the latter is underscored by recent reports that dietary factors including the soy isoflavone genistein (Gen) (Montales et al. 2012; Rahal et al. 2013), polyphenolic compounds present in blueberries (Montales et al. 2012), curcumin (Kakarala et al. 2010), and resveratrol (Kao et al. 2009), all of which have been reported to possess anti-metabolic/anti-diabetic properties (Gilbert and Liu 2013; Valsecchi 2013; Yeh et al. 2014; Rouse et al. 2014), can inhibit expansion of basal stem-like mammary cells with tumorigenic potential.
Here, we evaluated the effects of MET and several bioavailable and bioactive molecules present in soy protein isolate, namely the isoflavone Gen, peptide lunasin (Lun), and proteins β-conglycinin (β-con) and glycinin (Gly), alone and in combination, on the survival of CSCs present in the human colon cancer cell line HCT116 (Yeung et al. 2010). We examined potential chemo-preventive mechanisms of these components in CSCs and determined whether the combination of MET, Gen, and Lun with 5-FU could increase CRC cell death relative to 5-FU alone. Our results show that MET, Gen, and Lun together confer enhanced inhibitory effects on CSC expansion and suggest that the targeting of CSC with these molecules should be further explored for the clinical management of CRC.
Materials and methods
Cell culture and treatments
The human colon cancer cell lines HT29 and HCT116 (American Type Culture Collection; ATCC, Manassas, VA) were propagated in McCoy’s medium (ATCC) supplemented with 10 % fetal bovine serum (FBS; GIBCO, Carlsbad, CA) and 5 % antibiotic–antimycotic solution (ABAM; GIBCO) in a humidified incubator (5 % CO2:95 % air) at 37 °C. Cells were seeded in six-well plates at an initial density of 2 × 105 per well, and treated (in culture medium) with metformin (MET 60 µM; Sigma-Aldrich, St. Louis, MO), lunasin (Lun 2 µM; American Peptide Co., Sunnyside, CA), β-conglycinin (β-con 3 µM; provided by Dr. Ferreira, Federal University of Bahia, Brazil), glycinin (Gly 3 µM; provided by Dr. Ferreira), and genistein (Gen 2 µM; Sigma-Aldrich), alone or in combination. β-con and Gly were isolated and purified as described below. Metformin, Lun, β-con, and Gly were dissolved in phosphate-buffered saline (PBS; GIBCO), whereas Gen was dissolved in dimethyl sulfoxide (DMSO; Sigma-Aldrich). In other experiments, cells were treated with insulin (2 µM; Sigma), PTEN inhibitor SF1670 (2 µM; Echelon Biosciences, Salt Lake, UT), and 5-fluorouracil (5-FU 50 µM; Teva Parenteral Medicines, Irvine, CA). Treated cells were collected at select time points for subsequent analyses.
Extraction and isolation of β-conglycinin and glycinin
Soybean flour (60 mesh) was defatted with hexane [1:8 (wt/vol) flour-to-solvent ratio] by rocking at room temperature for 12 h and then re-extracted in the same solvent [1:6 (wt/vol)]. The extract was evaporated to dryness at room temperature and used for isolation of β-conglycinin and glycinin as previously described (Ferreira et al. 2010). The β-conglycinin and glycinin proteins were further purified by size exclusion chromatography on a Sepharose CL-6B column (1.0 × 100 cm) equilibrated with 0.05 M potassium phosphate, pH 7.5 containing 0.5 M NaCl and 0.01 % NaN3 at a column flow rate of 5.8 ml/tube. Column eluates were monitored at 280 nm, and protein-containing fractions were dialyzed and lyophilized. Column fractionation produced one major peak for each of the isolated proteins. SDS-PAGE confirmed the specific molecular weight and indicated over 95 % purity for β-con and Gly proteins (data not shown).
Cell viability, cell cycle distribution, and apoptosis
Cells were seeded in six-well plates at an initial density of 2 × 105 cells per well and incubated overnight prior to treatment. Cells were collected 48 h post-treatment, and the quantity of viable cells was determined by the trypan blue exclusion method using the Vi-Cell cell viability analyzer (Beckman Coulter Inc., Atlanta, GA). For cell cycle analysis, cells were serum-starved for 24 h after plating in 0.5 % charcoal-stripped FBS (CS-FBS; GIBCO), and 24 h later were treated with medium supplemented with 2.5 % CS-FBS and the described treatments. The analysis of cell cycle parameters followed previously described protocols (Rahal and Simmen 2010). At least 1 × 105 cells were stained with propidium iodide (PI; Sigma-Aldrich), and the proportion of cells in Sub G1, G1, S, and G2 phases was analyzed with the Becton–Dickinson LSRFortessa Flow Cytometer and associated software (BD Biosciences, San Jose, CA). The percent of apoptotic cells 48 h post-treatment was evaluated by Annexin V staining (Trevigen, Gaithersburg, MD), followed by analysis in the Becton–Dickinson LSRFortessa Flow Cytometer.
Flow cytometry
Single-cell suspensions were evaluated for relative frequency of CD133+CD44+-stained cells by flow cytometry and the use of PE-conjugated anti-CD133 (Miltenyi Biotec Incorporated, San Diego, CA) and FITC-conjugated anti-CD44 (BD Pharmingen, San Jose, CA), as previously described (Chen et al. 2011). Cells were sorted on a FACS Aria Cell Sorting Flow Cytometer (BD Biosciences); Supplementary Figure S1 shows the gating strategy used for FACS analysis. Dead cells were excluded using 4′,6-diamidino-2-phenylindole (DAPI; 1 µg/ml; Sigma-Aldrich). CD133+CD44+-positive cells were collected for colonosphere formation assay and gene expression analysis (below).
Colonosphere formation
To examine treatment effects on colonosphere formation, unsorted HCT116 cells and FACS-sorted CD133+CD44+ cell fractions were seeded at a density of 2 × 103 cells per well in ultra-low attachment plates (Corning Inc., Corning, NY) in plating medium [phenol red-free MEM supplemented with B27 media (Invitrogen); 20 ng/ml human basic fibroblast growth factor (Invitrogen, Carlsbad, CA), 20 ng/ml epidermal growth factor (Invitrogen), 10 μg/ml heparin (Sigma-Aldrich), 1 % ABAM, and 100 μg/ml gentamicin (Sigma-Aldrich)] (Montales et al. 2012). Treatments were MET (60 µM), soy bioactive factors (3 µM each for β-con and Gly; 2 µM each for Lun and Gen), and insulin (2 µM), singly and with the indicated combinations. To investigate whether MET limited colonosphere-forming ability, HCT116 cells were treated after initial plating with MET or vehicle, collected 24 h later and then grown as single-cell suspensions under low attachment-plating conditions. For PTEN inhibitor studies, unsorted cells (2 × 105 per well) were plated in six-well plates, allowed to attach overnight, and then treated with the PTEN inhibitor SF1670 (2 µM) for 24 h, prior to collection and re-plating in ultra-low attachment plates. Colonosphere number was counted 5 days after the initial (one-time) treatment using an inverted microscope. To evaluate self-renewal, primary colonospheres (P1) were collected after counting, dissociated into single cells, filtered using a 40 μM sieve, and re-plated in ultra-low attachment plates. Colonospheres formed at secondary passage (P2) were counted at 7 days post-plating and expressed as a percentage of the total number of initially plated cells. Treatment effects were determined from at least three independent experiments, with 3–4 replicates per experiment.
RNA isolation and quantification
Total RNA was extracted from unsorted and FACS-sorted cells and from colonospheres using TRIzol reagent (Invitrogen). cDNA synthesis and real-time quantitative PCR (QPCR) were performed following published methods (Montales et al. 2012). Primers (Supplemental Table 1) were designed using Primer Express (Applied Biosystems, Foster City, CA) and synthesized by Integrated DNA Technologies, Inc. (Coralville, IA). Gene expression was determined by QPCR using Bio-Rad iTaq SYBR Green Supermix (Bio-Rad; Hercules, CA). Transcript levels for target genes were calibrated to a standard curve generated using pooled cDNAs and normalized to a factor that was derived from the geometric mean of expression for β-actin (ACTB) and cyclophilin A (CYPA) genes, using GeNorm excel file software as previously described (Vandesompele et al. 2002).
Western blot analysis
Whole extracts from treated cells (60 µM MET or vehicle; 24 h) were harvested in RIPA buffer containing protease inhibitor cocktail (Santa Cruz Biotechnology; Santa Cruz, CA). Protein concentrations were determined using the bicinchoninic acid protein assay kit (Pierce Biotechnology; Rockford, IL). Equal amounts of lysate protein (50 μg) were subjected to immunoblotting as described previously (Al-Dwairi et al. 2012). Primary antibodies used were anti-PTEN (1:1000; Cell Signaling, Danvers, MA), anti-FASN (1:1000; Abcam, Cambridge, MA), and anti-β-actin (1:10,000; Sigma-Aldrich). Blots were stripped with Restore Western blot stripping buffer (Pierce Biotechnology) prior to incubation with another primary antibody. Immunoreactive proteins were visualized with Amersham ECL Plus kit (GE Health Care Life Sciences, Piscataway, NJ). Digital images were captured with a FlourChem HD2 Imager system (Alpha Innotech, San Leandro, CA), and densitometry was performed using Alpha View software (Cell Biosciences; Santa Clara, CA). Levels of immunoreactive PTEN and FASN proteins were normalized to that for β-actin.
Data analysis
All experiments were performed at least twice, with each experiment conducted in triplicate or quadruplicate. Data are presented as the mean ± SEM; except for that in Fig. 5b which are LS mean ± SEM. Statistical significance between diet groups was evaluated by Student’s t test or one-way analysis of variance using Sigma Stat version 3.5 for Windows. Data in Fig. 5b were subjected to a two-way ANOVA with ‘experiment’ being considered as a ‘random’ factor, and pairwise multiple comparison procedures (Holm–Sidak method) were used to ascribe statistically significant differences between treatment groups. A P value <0.05 was considered to be statistically significant, with tendency for significance at 0.05 < P < 0.1.
Results
HT29 and HCT116 colonospheres
The human colon cancer cell lines HT29 and HCT116 contain a sub-population of cancer stem cell-like cells (CSC), which are distinguished by their expression of cell surface antigens CD133 and CD44 (Sahlberg et al. 2014) and their ability to initiate tumors in nude mice (Yeung et al. 2010; Chen et al. 2011). The relative abundance of the CD133+CD44+ subpopulation between these cell lines was determined by FACS. HCT116 cells showed a higher percentage of CD133+CD44+ subpopulation (~80 %) when compared to HT29 cells (~60 %) (Fig. 1a, b). Further, colonospheres formed from CD133+CD44+ fractions of HCT116 were larger in diameter (Fig. 1c) and were expressed at higher frequencies in both initial (P1; 1 %) and secondary (P2; 3 %) passages (Fig. 1d) than those of HT29 (P1; 0.5 %; P2; 1 %) cells. The increased frequencies in the CD133+CD44+ epithelial subpopulation in P2 relative to P1 for each cell line indicated self-renewal competence. Transcript levels for the CSC marker genes CD166, CD44, and NOTCH1 and for the key lipogenic enzyme, fatty acid synthase (FASN), were higher in the CD133+CD44+ subpopulation from HCT116 than from HT29 cells (Fig. 1e). By contrast, PTEN mRNA abundance was comparable between the two cell subpopulations (Fig. 1e). These collective results provided a rationale for our selection of the HCT116 cell line as in vitro model to test the effects of MET and soy-derived bioactive molecules on human colon cancer.
MET attenuated cell viability and colonosphere formation, induced cell apoptosis and altered PTEN and FASN expression of HCT116 cells
Clinical studies suggest that MET treatment of diabetic patients reduces their risk of and mortality from CRC (Zhang et al. 2013; Sehdev et al. 2015). The effects of MET on colon cancer proliferation and survival were investigated in HCT116 cells using MET at a near-physiological concentration of 60 µM (readily achievable in humans with daily doses of oral MET) (Ochs 2014). MET decreased cell viability (Fig. 2a) in the absence of changes in cell cycle distribution (Fig. 2b). MET increased the percentage of cells in early and late phases of apoptosis (Fig. 2c), and induced the expression of the pro-apoptotic gene BAX (Fig. 2c, inset) relative to control (non-treated) cells. Cells treated with MET had lower percentage of colonospheres formed at both P1 and P2 (a ~ 50 % decrease) when compared to control cells; however, both control and MET-treated cells retained the property of self-renewal (P2 > P1) (Fig. 2d).
We next quantified the temporal expression of the tumor suppressor PTEN and the lipogenic enzyme FASN in control and MET-treated cells. MET increased PTEN mRNA levels (Fig. 2e) and decreased FASN mRNA abundance (Fig. 2f) in a time-dependent manner. Interestingly, MET effects on PTEN and FASN gene expression were maximal at 6 h and persisted to 24 h (Fig. 2e, f) and 48 h (data not shown). The increase in PTEN and decrease in FASN mRNA abundance at 24 h with MET were confirmed at the level of their respective proteins (Fig. 2g, h), demonstrating concordance of mRNA and protein expression.
Soy bioactive components alter HCT116 cell viability, apoptosis, and colonosphere formation
To examine effects of soy bioactive components with potential anti-carcinogenic activities (Yan et al. 2010; Dia and Gonzales de Mejia 2011) on the cell cycle, HCT116 cells were treated with Gen, Lun, β-con, and Gly at concentrations (2–3 μM) that, in the case of Gen, approximated that found for sera of regular soy food consumers (Iwasaki et al. 2008). Treatment with the soy-derived components decreased cell viability although to differing extents, with the efficacy of Gen > Gly > β-con = Lun (Fig. 3a). There was an associated, although not a proportionate, decrease in cyclin D1 (CCND1) gene expression with loss of cell viability (Lun > Gen = Gly = β-con) (Fig. 3b). Gen, β-con, and Gly, but not Lun inhibited cell cycle progression relative to control cells (Fig. 3c). Specifically, Gen induced G1-S arrest, whereas β-con and Gly induced G2-M arrest (Fig. 3c). With apoptosis as the measured endpoint, Gen and Lun demonstrated enhancement (Gen > Lun), with β-con and Gly showing no comparable activities over control cells (Fig. 3d).
We next examined whether the above soy factors affected colonosphere formation in HCT116 cells. Gen, Lun, and Gly each suppressed colonosphere formation by ~50 %, while β-con was ineffective at the concentration tested (Fig. 3e). Co-treatment with MET and Lun reduced colonosphere formation to a slightly lesser extent (by ~60 %) than did co-treatment with MET and Gen (by ~76 %) (Fig. 3f). Interestingly, co-treatment with all three components failed to reduce colonosphere formation further than that elicited with MET in the presence of either Lun or Gen (Fig. 3g).
MET, Lun, and Gen limit the cancer stem cell-like cell subpopulation, in part, through PTEN
The changes in growth and survival elicited by Gen and by Lun (Fig. 3) were accompanied by decreased FASN and increased PTEN mRNA levels in P1 colonospheres (Fig. 4a). By contrast, P1 colonospheres from β-con- and Gly-treated cells failed to manifest changes in PTEN and FASN gene expression (Fig. 4a). The tumor suppressor PTEN is a common target for inactivation in cancer, including CRC (Rahal and Simmen 2010; Sawai et al. 2008; Molinari and Frattini 2013). We next evaluated whether induction of PTEN is responsible, in part, for the inhibitory effects of MET, Gen, and Lun on colonosphere formation. HCT116 cells were pre-treated with the PTEN inhibitor SF1670 (2 μM) for 24 h (untreated HCT116 cells served as control); treated cells were subsequently plated under non-adherent conditions with added MET (60 µM), Lun (2 µm), or Gen (2 µM). SF1670 binds to the PTEN active site, resulting in elevated phosphatidylinositol (3,4,5) triphosphate signaling (Rosivatz et al. 2006). We found that inhibition of PTEN activity increased colonosphere formation relative to control cells (Fig. 4b). MET, Lun, or Gen alone reduced the colonosphere-promoting effect of SF1670, with inhibition for Gen = Lun > MET (Fig. 4b).
Epidemiological studies suggest that hyperinsulinemia is associated with increased CRC risk (Giovannucci 2007). Moreover, insulin signaling was found to inhibit PTEN expression (Liu et al. 2014), and insulin resistance has been linked to increased FASN expression (Menendez et al. 2009), indicating that aberrant insulin signaling may underlie altered PTEN and FASN expression levels and activity in CRC. To evaluate whether Lun, Gen, and/or MET attenuate CSC frequency by opposing insulin action, we exposed HCT116 cells to an elevated level of insulin (2 µM, modeling hyperinsulinemia) and determined effects of added factors on colonosphere formation and on the CD133+CD44+ subpopulation. Insulin increased colonosphere formation (Fig. 4c) and the percentage of CD133+CD44+ cells (Fig. 4d). Lun, Gen, and MET reduced CSC formation under in vitro hyperinsulinemic condition, with Gen demonstrating greater inhibitory activity than either MET and Lun (MET = Lun) (Fig. 4c, d).
HCT116 cell killing by 5-FU plus MET, Gen, and Lun
5-Fluorouracil (5-FU) is a first-line chemotherapeutic agent for CRC treatment. To examine whether combination treatment with Lun + Gen + MET enhances the sensitivity of HCT116 cells to the cytotoxic effects of 5-FU under a hyperinsulinemic condition, cells were treated with insulin (2 µM) and 5-FU in the presence of MET alone or in combination with Lun + Gen (Fig. 5a, b). Amount of apoptosis induced by 5-FU in the presence of insulin was similar to that elicited by MET + Gen + Lun alone (in the absence of 5-FU) but was not affected by co-addition of only MET (Fig. 5a, b). However, co-addition of MET + Gen + Lun under hyperinsulinemic conditions modestly but significantly increased 5-FU effects on apoptosis (Fig. 5a, b) and cell viability (Fig. 5c) relative to the effects seen with MET alone. Since we only tested combinations of factors at a single dose for each, it is not possible to discern whether effects were additive or synergistic. In addition, exposure to MET + Gen + Lun for 72 h significantly attenuated (by ~20 %) the percentage of CD133+CD44+ cells, when compared to 5-FU alone-treated cells (Fig. 5d).
Discussion
CRC is increasingly considered to have metabolic underpinnings; hence, dietary factors that maintain metabolic homeostasis or normalize metabolic dysfunction may be beneficial to inhibiting CRC progression, metastasis, and relapse. Clinical and epidemiological studies have implicated the anti-diabetic drug metformin (MET) in reducing the risk of CRC among diabetic patients as well as improving CRC outcome in patients with diabetes (Dowling et al. 2011; Zhang et al. 2013; Nangia-Makker et al. 2014; Sehdev et al. 2015). Additionally, in diabetic patients with CRC, the decrease in mortality with MET was inversely associated with the frequency of CD133+ subpopulation with CSC properties (Zhang et al. 2013). Nevertheless, MET effects on colon cancer remain controversial (Sui et al. 2014). Moreover, MET elicits unfavorable side effects in some individuals, precluding its use at typically prescribed doses for diabetic patients. Thus, factors that could increase the efficacy of MET may be advantageous for its inclusion as a therapy to address CRC prevention and/or treatment.
Previous studies in several laboratories including our own (Xiao et al. 2008; Al-Dwairi et al. 2012; Yeh et al. 2014; Isanga and Zhang 2011) have shown that soybean components when consumed can favorably affect metabolic pathways and associated genes in animal models and in humans. Moreover, in a mouse model of breast cancer and in breast cancer cells in vitro, dietary intake of soy protein isolate and treatment with Gen, respectively, decreased the frequency of the mammary basal stem cell-like subpopulation, which was associated with decreased mammary tumor incidence and oncogenic molecular profile (Montales et al. 2012; Rahal et al. 2013). Based on this previous body of work, we hypothesized that combination(s) of MET and bioactive soy factors would increase the therapeutic efficacy of the CRC drug 5-FU by targeting cancer stem cells. Our findings present a novel and potentially important role for combination MET, Lun, and Gen in not only reducing cancer cell viability and survival, but in limiting the frequency of the cancer cell subpopulation (CD133+CD44+) with self-renewal properties. We also provide evidence, using the PTEN-specific inhibitor SF1760 and insulin, to indicate that MET, Gen, and Lun actions are comparably mediated by their concurrent induction of tumor suppressor PTEN and inhibition of pro-oncogenic FASN expression. The functional significance of these results is supported by the increased efficacy of 5-FU in eliminating CSCs under high-insulin conditions, the latter mimicking aspects of type 2 diabetes and/or obesity, when co-exposed with MET, Gen, and Lun.
In these studies, we primarily utilized the human colon cancer cell line HCT116 since its highly aggressive nature and increased proportion of non-differentiating CSCs are clinically correlated with lymph node metastases and reduced survival (Yeung et al. 2010). We show that relative to the more differentiated, less aggressive colon cancer cell line HT29, HCT116 cells exhibit a greater proportion of the CSC subpopulation (designated CD133+CD44+), increased ability to form colonospheres (a measure of tumor-forming ability and self-renewal in vitro), and enhanced expression of CSC genes. Using these cells to model aggressive CRC in vitro, we observed that MET, Gen, and Lun, individually and together, limited the expansion of the CSC subpopulation, suggesting their potential value in inhibiting disease progression and conceivably, metastasis and recurrence. The higher efficacy of Gen relative to MET, at the doses used here, in reducing colonosphere formation under high-insulin conditions is a novel and an unexpected finding. Previous studies have shown a dose-related inverse association between soy consumption and CRC risk (Yan et al. 2010). Nevertheless, molecular mechanisms to explain the preventative effects of soy on colon cancer remain relatively unknown. The demonstration of the efficacy of Gen, Lun, and Gly in targeting CSCs at relatively low doses (2–3 uM) suggests their potential to reduce currently prescribed doses of CRC conventional therapies with adverse side effects. While our data point to potential combination therapies of soybean components and 5FU, the current study has not attempted the complex task of defining optimal doses for these single agents and combinations (future studies). Our findings also provide an impetus to pursue preclinical studies of these bioactive molecules in animal models of colon cancer metastasis and under conditions of hyperinsulinemia.
The present data identify PTEN and FASN as target genes for MET, Lun, and Gen in the CSC subpopulation (Fig. 6). The bioactive molecules did not demonstrate additive or synergistic effects on PTEN and FASN expression, possibly indicating that once the threshold induction (for PTEN) or reduction (for FASN) in these signaling pathways is achieved, further blocking of the CSC self-renewal/expansion by these molecules becomes limited. The latter idea is consistent with the notion that additional pathways independent of MET, Lun, and Gen signaling regulate CSC expansion. Our observations that MET alone was less effective than MET + Gen in inhibiting CSC expansion, however, suggest that Gen affects other pathways to further limit CSC expansion and hence, CRC (Fig. 6). The identities of these potential contributing pathways are unknown, but may involve inflammatory, cytokine/chemokine, and epithelial–mesenchymal transition signaling, each of which has been separately linked to the promotion of CSCs when aberrantly activated (Bruttel and Wischhusen 2014) and to the favorable health effects of soy when inhibited (Hsu et al. 2010).
Conclusions
The present study utilizing an in vitro model of aggressive colon cancer in a hyperinsulinemic environment, provides insight into molecular networks shared by the well-prescribed, anti-diabetic drug MET and bioactive/bioavailable soy food components, which potentially may be harnessed to increase drug efficacy in patients with CRC. Additionally, given that PTEN loss of function/expression is a hallmark of many primary and metastatic CRCs, our results suggest further exploration of the potential of combination therapy with MET and the soy factors to improve CRC outcome. Induction of tumor suppressor PTEN expression in cancer cells by dietary bioactive factors is an emerging theme (Liu et al. 2013; Pabona et al. 2013) that warrants further confirmation in vivo. In addition, our results for β-conglycinin and glycinin suggest further examination of these molecules for efficacy in CRC, in light of their high abundance and bioavailability either as intact or smaller proteins/peptides in consumed soy products. The mode(s) of action of these two latter proteins in cancer cell growth regulation remain virtually unexplored, with the exception of a previous report on β-conglycinin as being inhibitory to leukemia cell growth in vitro (Wang et al. 2008). Finally, the identification of food constituents that target CSCs underscores the potential importance of diet/nutrition in the genesis (and treatment) of primary and metastatic CRC and of other cancer types with metabolic underpinnings.
Abbreviations
- β-con:
-
β-Conglycinin
- CSC:
-
Cancer stem cell
- CRC:
-
Colorectal cancer
- FASN:
-
Fatty acid synthase
- 5-FU:
-
5-Fluorouracil
- Gen:
-
Genistein
- Gly:
-
Glycinin
- Met:
-
Metformin
- PTEN:
-
Phosphatase and tensin homologue deleted on chromosome ten
- QPCR:
-
Real-time quantitative polymerase chain reaction
References
Al-Dwairi A, Pabona JM, Simmen RC, Simmen FA (2012) Cystolic malic enzyme 1 (ME1) mediates high fat diet-induced adiposity, endocrine profile, and gastrointestinal tract proliferation-associated biomarkers in male mice. PLoS ONE 7(10):e46716
Al-Dwairi A, Brown AR, Pabona JM, Van TH, Hamdam H, Mercado CP, Quick CM, Wight PA, Simmen RC, Simmen FA (2014) Enhanced gastrointestinal expression of cytosolic malic enzyme (ME1) induces intestinal and liver lipogenic gene expression and intestinal cell proliferation in mice. PLoS ONE 9(11):e113058
Bruttel VS, Wischhusen J (2014) Cancer stem cell immunology: key to understanding tumorigenesis and tumor immune escape? Front Immunol 5:360
Chandler JM Cancerous (2010) Cancerous stem cells. Deviant stem cells with cancer causing misbehavior. Stem Cell Res Ther 1:13
Chen KL, Pan F, Jiang H, Chen JF, Pei L, Xie FW, Liang HJ (2011) Highly enriched CD133+CD44+ stem-like cells with CD133+CD44hi metastatic subset in HCT 116 colon cancer cells. Clinical Exp. Metastasis 28:751–763
Dia VP, Gonzales de Mejia E (2011) Lunasin potentiates the effect of oxaliplatin preventing outgrowth of colon cancer metastasis, binds to α5β1 integrin and suppresses FAK/ERK/NF-κB signaling. Cancer Lett 313:168–180
Dowling RJ, Goodwin P, Stambolic V (2011) Understanding the benefit of metformin use in cancer treatment. BMC Med 9:33
Ferreira E, Silva M, Demonte A, Neves V (2010) β-Conglycinin (7S) and glycinin (11S) exert a hypocholesterolemic effect comparable to that of fenofibrate in rats fed a high-cholesterol diet. J Funct Foods 2:275–283
Gilbert ER, Liu D (2013) Anti-diabetic functions of soy isoflavone genistein: mechanisms underlying its effects on pancreatic β-cell function. Food Funct 4:200–212
Giovannucci E (2007) Metabolic syndrome, hyperinsulinemia, and colon cancer: a review. Am J Clin Nutr 86:s836–s842
Hsu A, Bray TM, Ho E (2010) Anti-inflammatory activity of soy and tea in prostate cancer prevention. Exp Biol Med 235(6):659–667
Isanga J, Zhang G-N (2011) Soybean bioactive components and their implications to health—a review. Food Rev Int 24:252–276
Iwasaki M, Inoue M, Otani T, Sasazuki S, Kurahashi N, Miura T, Yamamoto S et al (2008) Plasma isoflavone levels and subsequent risk of breast cancer among Japanese women: a nested case-control study from the Japan Public Health Center-based prospective study group. J Clin Oncol 26:1677–1683
Kakarala M, Brenner D, Korkaya H, Cheng C, Tazi K, Ginestier C, Liu S, Dontu G, Wicha MS (2010) Targeting breast stem cells with the cancer preventive compounds curcumin and piperine. Breast Cancer Res Treat 122:777–785
Kao CL, Huang PI, Tsai PH, Lo JF, Lee YY, Chen YJ, Chen YW, Chiou SH (2009) Resveratrol-induced apoptosis and increased radiosensitivity in CD133-positive cells derived from atypical teratoid/rhabdoid tumor. Int J Radiat Oncol Biol Phys 74:219–228
Linnekamp JF, Wang X, Medema JP, Vermeulen L (2015) Colorectal cancer heterogeneity and targeted therapy: a case for molecular disease subtypes. Cancer Res 75:245–249
Liu YL, Zhang GQ, Yang Y, Zhang CY, Fu RX, Yang YM (2013) Genistein induces G2/M arrest in gastric cancer cells by increasing the tumor suppressor PTEN expression. Nutr Cancer 65:1034–1041
Liu J, Visser-Grieve S, Boudreau J, Yeung B, Lo S et al (2014) Insulin activates the insulin receptor to downregulate the PTEN tumour suppressor. Oncogene 33:3878–3885
Menendez JA, Vazquez-Martin A, Ortega FJ, Fernandez-Real JM (2009) Fatty acid synthase: association with insulin resistance, type 2 diabetes, and cancer. Clin Chem 55:425–438
Molinari F, Frattini M (2013) Functions and regulation of the PTEN gene in colorectal cancer. Front Oncol 3:326
Montales MT, Rahal O, Kang J, Rogers TJ, Prior R, Wu X, Simmen RC (2012) Repression of mammosphere formation of human breast cancer cells by soy isoflavone genistein and blueberry polyphenolic acids suggests diet-mediated targeting of cancer stem-like/progenitor cells. Carcinogenesis 33:652–660
Nangia-Makker P, Yu Y, Vasudevan A, Farhana L, Rajendara SG, Levi E, Majumdar AP (2014) Metformin: a potential therapeutic agent for recurrent colon cancer. PLoS ONE 9(1):e84369
Ochs R (2014) Metformin: a case study in the meaning of serum concentrations. J Mol Pharm Org Process Res 2:e111
Pabona JM, Dave B, Su Y, Montales MT, de Lumen BO et al (2013) The soybean peptide lunasin promotes apoptosis of mammary epithelial cells via induction of tumor suppressor PTEN: similarities and distinct actions from soy isoflavone genistein. Genes Nutr 8:79–90
Paldino E, Tesori V, Casalbore P, Gasbarrini A, Puglisi MA (2014) Tumor initiating cells and chemoresistance: which is the best strategy to target colon cancer stem cells? Biomed Res Int 2014:859871
Rahal O, Simmen RC (2010) PTEN and p53 cross-regulation induced by soy isoflavone genistein promotes mammary epithelial cell cycle arrest and lobuloalveolar differentiation. Carcinogenesis 31:1491–1500
Rahal O, Machado H, Montales MT, Pabona JM, Heard M, Nagarajan S, Simmen RC (2013) Dietary suppression of the mammary CD29(hi)CD24(+) epithelial subpopulation and its cytokine/chemokine transcriptional signatures modifies mammary tumor risk in MMTV-Wnt1 transgenic mice. Stem Cell Res 11:1149–1162
Rosivatz E, Matthews J, McDonald N, Mulet X, Ho KK, Lossi N et al (2006) A small-molecule inhibitor for phosphatase and tensin homologue deleted on chromosome 10 (PTEN). ACS Chem Biol 1:780–790
Rouse M, Younes A, Egan JM (2014) Resveratrol and curcumin enhance pancreatic β-cell function by inhibiting phosphodiesterase activity. J Endocrinol 223:107–117
Sahlberg SH, Spiegelberg D, Glimelius B, Stenerlow B, Nestor M (2014) Evaluation of cancer stem cell markers CD133, CD44, CD24: association with AKT isoforms and radiation resistance in colon cancer cells. PLoS ONE 9(4):e94621
Sawai H, Yasuda A, Ochi N, Ma J, Matsuo Y et al (2008) Loss of PTEN expression is associated with colorectal cancer liver metastasis and poor patient survival. BMC Gastroenterol 8:56
Sehdev A, Shih YC, Vekhter B, Bissonnette MB, Olopade O, Polite BN (2015) Metformin for primary colorectal cancer prevention in patients with diabetes: a case-control study in a US population. Cancer 121(7):1071–1078
Siegel RL, Miller K, Jemal A (2015) Cancer statistics, 2015. CA Cancer J Clin 65:5–29
Spector D, Anthony M, Alexander D, Arab L (2013) Soy consumption and colorectal cancer. Nutr Cancer 47:1–12
Sui X, Xu Y, Yang J, Fang Y, Lou H, Han W et al (2014) Use of metformin alone is not associated with survival outcomes of colorectal cancer cell but AMPK activator AICAR sensitizes anti-cancer effect of 5-fluorouracil through AMPK activation. PLoS ONE 9(5):e97781
Tomasetti C, Vogelstein B (2015) Cancer etiology. Variation in cancer risk among tissues can be explained by the number of stem cell divisions. Science 347:78–81
Tomasetti C, Marchionni L, Nowak M, Parmigiani G, Vogelstein B (2014) Only three driver gene mutations are required for the development of lung and colorectal cancers. Proc Natl Acad Sci USA 112:118–123
Valsecchi AE (2013) The soy isoflavone genistein reverses oxidative and inflammatory state, neuropathic pain, neurotrophic and vasculature deficits in diabetes mouse model. Eur J Pharmacol 650:694–702
Vandesompele J, De Preter K, Pattyn F, Poppe B, Van Roy N, De Paepe A, Speleman F (2002) Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes. Genome Biol 3(7):RESEARCH0034
Wang W, Bringe NA, Berhow MA, de Meija Gonzalez (2008) Beta-conglycinins among sources of bioactives in hydrolysates of different soybean varieties that inhibit leukemia cells in vitro. J Agric Food Chem 56:4012–4020
White AC, Lowry W (2015) Refining the role for adult stem cells as cancer cells of origin. Trends Cell Biol 25:11–20
Xiao R, Carter JA, Linz AL, Ferguson M, Badger TM, Simmen FA (2006) Dietary whey protein lowers serum C-peptide concentration and duodenal SREBP-1c mRNA abundance, and reduces occurrence of duodenal tumors and colon aberrant crypt foci in azoxymethane-treated male rats. J Nutr Biochem 17:626–634
Xiao R, Su Y, Simmen RC, Simmen FA (2008) Dietary soy protein inhibits DNA damage and cell survival of colon epithelial cells through attenuated expression of fatty acid synthase. Am J Physiol Gastrointest Liver Physiol 294:G868–G876
Yan L, Spitznagel EL, Bosland MC (2010) Soy consumption and colorectal cancer risk in humans: a meta-analysis. Cancer Epidemiol Biomarkers Prev 19:148–158
Yeh WJ, Yang HY, Chen JR (2014) Soy β-conglycinin retards progression of diabetic nephropathy via modulating the insulin sensitivity and angiotensin-converting enzyme activity in rats fed with high salt diet. Food Funct 5:2898–2904
Yeung TM, Gandhi S, Wilding J, Muschel R, Bodmer W (2010) Cancer stem cells from colorectal cancer-derived cell lines. Proc Natl Acad Sci USA 107:3722–3727
Zeuner A, Todaro M, Stassi G, De Maria R (2014) Colorectal cancer stem cells: from the crypt to the clinic. Cell Stem Cell 15:692–705
Zhang Y, Guan M, Zheng Z, Zhang Q, Gao F, Xue Y (2013) Effects of metformin on CD133+ colorectal cancer cells in diabetic patients. PLoS ONE 8(11):e81264
Acknowledgments
We thank other members of our laboratories for helpful discussions during the course of this work. We are also grateful to Lorenzo Fernandes and Eric Siegel for their assistance with statistical analysis and data presentation.
Authors’ contributions
MTM carried out the experiments and performed statistical analysis. EF and VN performed the extraction and purification of β-conglycinin and glycinin. MTM, RCMS, and FAS interpreted the data and drafted the manuscript. RCMS and FAS supervised the entire project. All authors read and approved the final manuscript.
Funding
This work was supported by NIH R01CA136493, NIH UL1TR000039, and the Huie Family Trust.
Author information
Authors and Affiliations
Corresponding author
Electronic supplementary material
Below is the link to the electronic supplementary material.
Rights and permissions
About this article
Cite this article
Montales, M.T.E., Simmen, R.C.M., Ferreira, E.S. et al. Metformin and soybean-derived bioactive molecules attenuate the expansion of stem cell-like epithelial subpopulation and confer apoptotic sensitivity in human colon cancer cells. Genes Nutr 10, 49 (2015). https://doi.org/10.1007/s12263-015-0499-6
Received:
Accepted:
Published:
DOI: https://doi.org/10.1007/s12263-015-0499-6