Lippia origanoides extract induces cell cycle arrest and apoptosis and suppresses NF-κB signaling in triple-negative breast cancer cells

  • Authors:
    • Vishak Raman
    • Jorge Luis Fuentes Lorenzo
    • Elena E. Stashenko
    • Morris Levy
    • Maria M. Levy
    • Ignacio G. Camarillo
  • View Affiliations

  • Published online on: October 19, 2017     https://doi.org/10.3892/ijo.2017.4169
  • Pages: 1801-1808
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Treatments targeting hormone receptors typically fail to provide a positive clinical outcome against triple-negative breast cancers (TNBC), which lack expression of estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor 2 (Her2/neu). Towards identifying viable treatments for aggressive breast cancer, we have tested an extract of the tropical plant Lippia origanoides (LOE) on TNBC and normal cells lines to uncover its potential anticancer effects. Treatment with LOE reduced TNBC cell viability in a dose-dependent manner to a greater extent than in normal mammary epithelial MCF10A cells. In MDA-MB‑231 cells, LOE was found to halt the cell cycle in the G0/G1 phase via cyclin D1 and cIAP2 regulation, and induce apoptosis without promoting necrosis via caspase-8/-3 and PARP cleavage. Constitutive nuclear factor-κB (NF-κB) signaling has been shown to contribute to the heightened inflammatory state and survival in TNBC cells. Herein, we also provide evidence that LOE inhibits NF-κB signaling by reducing RIP1 protein levels in MDA-MB-231 cells. These studies reveal that LOE suppresses key features of the progression of aggressive breast cancer cells and provides a basis for further definition of its underlying mechanisms of action and anticancer potential.

Introduction

Breast cancer is a major health concern worldwide, and a leading cause of cancer-related deaths among women in the United States where over 245,000 new cases are diagnosed and 45,000 deaths occur each year (1). In the majority of diagnoses, this disease presents as a hormone-sensitive breast cancer that overexpressed estrogen receptor (ER), progesterone receptor (PR) and/or human epidermal growth factor receptor 2 (HER2/neu). These cancers typically depend on hormone-receptor signaling for tumor growth and progression. Consequently, conventional therapy against the majority of breast cancers targets these receptors (e.g., tamoxifen, a selective estrogen receptor modulator and herceptin, an antibody that inhibits HER) or attempts to inhibit enzymes involved in estrogen synthesis (e.g., aromatase inhibitors such as letrozole) (24). In contrast, the most deadly subtype of this disease is known clinically as triple-negative breast cancer (TNBC), and constitutes approximately 15% of invasive breast cancers. TNBCs lack expression of genes coding for ER, PR and HER2/neu, are not dependent on hormonal signaling for progression, and do not respond to conventional therapy (5).

TNBCs are frequently of the highly invasive and recurrent basal-like subtype (6). They are also associated with a variety of aberrations (e.g., higher incidence of TP53 mutations and the constitutive activation of pro-survival and inflammatory pathways such as MAPK and NF-κB), all of which may contribute to their growth, survival and resistance to chemotherapy (710). NF-κB signaling maintains an autocrine loop of pro-inflammatory cytokines such as IL-6 and TNF-α, promotes expression of pro-survival factors such as Bcl-xL, cIAP1 and cIAP2 and proteins involved in cell cycle progression (e.g., cyclin D1), as well as invasion and metastasis markers such as MMP-9 and vimentin (1117). Constitutive activation of NF-κB in triple-negative breast cancer may contribute to its aggressiveness and ability to maintain hormone-independent growth. Inhibition of NF-κB signaling is thus a viable target for novel therapeutics against TNBC.

Plant-derived molecules have been a critical source of cancer therapeutic agents. Generally, there are four main classes of plant-derived anticancer drugs currently in clinical use: Vinca alkaloids, taxanes, epipodophyllotoxins and camptothecins; the former two have entered clinical use against breast cancer following successful trials (18,19). Studies have also identified other natural plant compounds as capable of exerting pleiotropic anticancer effects. Plumbagin, for example, was shown to inhibit the DNA-binding activity of NF-κB and induce apoptosis in triple-negative MDA-MB-231 breast cancer cells (20). Further, halofuginone inhibited the nuclear localization of NF-κB and AP1, which are critical transcriptional activators of matrix metalloproteinase-9 (MMP-9), thereby reducing migration and invasiveness of these cells (21). Curcumin, honokiol, resveratrol and pinocembrin, among others, have also been shown to possess potent anti-cancer effects (2225). These positive outcomes highlight the importance of fully uncovering the untapped clinical potential of natural compounds.

Herein, we report on an extract from Lippia origanoides, a medicinal tropical plant native to South America for its potential anticancer effects on TNBC. Also known as 'oregano de monte' or mountain oregano, infusions of the plant have been used traditionally to alleviate gastrointestinal ailments and as topical analgesics (26). Towards a better understanding of L. origanoides actions and health benefits, the novel L. origanoides extract (LOE) used in this study was originally isolated by supercritical fluid extraction and characterized using HPLC-MS (27,28). The LOE was found to have numerous major components including pinocembrin, carvacrol, thymol and trans-β-caryophyllene. Studies using commercially available reagents of these compounds have shown they have pro-apoptotic and anti-proliferative effects on various cancer cell lines. For example, pinocembrin, a flavanone which comprises nearly 55% of the total LOE, has been shown to decrease viability and prevent epithelial-mesenchymal transition induced by TGF-β in Y-79 retinoblastoma cells (25). Further, trans-β-caryophyllene and α-humulene, two sesquiterpenes present in the extract, were shown to synergize and inhibit cell growth and proliferation in MCF-7 breast cancer cells (29). Strikingly, several components of LOE have shown inhibitory effects on NF-κB signaling, a key survival and proliferative pathway in TNBC (3034). These reports support the idea that L. origanoides may be a source of novel components that can effectively target aggressive breast cancer.

Our results show that treatment with LOE leads to a G0/G1 phase halt and apoptosis in MDA-MB-231 triple-negative breast cancer cells without promoting necrotic cell death. Further, we reveal that cell cycle proteins and apoptotic markers, as well as key NF-κB regulatory molecules, are modulated by treatment with LOE, thereby shedding light on a mechanism of action behind the anticancer effects of LOE. These data provide an important first step towards defining the potential utility of LOE in the identification and development of novel therapeutic strategies for TNBC.

Materials and methods

Plant material and extract

Lippia origanoides plants were collected from the Chicamocha River Canyon (Los Santos, Santander, Colombia). Taxonomic identification of L. origanoides was performed by Dr José Luis Fernández Alonso (National University, Bogotá, Colombia). The L. origanoides specimen (COL560259) was placed in the Colombian National Herbarium (Bogotá). Fresh leaves and flowers from L. origanoides were used for extraction as previously described (28). The extract was dissolved in methanol at a concentration of 50 mg/ml (stock solution), and then different extract concentrations were prepared in methanol for in vitro bioassays.

Cell culture

Triple-negative breast cancer (MDA-MB-231 and CRL-2321) and normal mammary epithelial (MCF10A) cell lines were obtained from the American Type Culture Collection. MCF10A-H cells, derived via H-ras transformation of MCF10A cells, were a kind gift from Dr Barbara Stefanska, Purdue University. MDA-MB-231 cells were cultured in Dublecco's modified essential medium (DMEM; Life Technologies, Grand Island, NY, USA) supplemented with 10% fetal bovine serum (FBS; Atlanta Biologicals, Norcross, GA, USA), 100 IU/ml penicillin and 100 µg/ml streptomycin (Life Technologies). MCF10A and MCF10A-H cells were cultured in a 1:1 ratio of DMEM:Ham's F12 supplemented with 5% horse serum (HS; Atlanta Biologicals), 20 ng/ml human epidermal growth factor (Sigma-Aldrich, St. Louis, MO, USA), 0.5 mg/ml hydrocortisone (Sigma-Aldrich), 100 ng/ml cholera toxin (Sigma-Aldrich), 10 µg/ml bovine insulin (Sigma-Aldrich), 100 IU/ml penicillin and 100 µg/ml streptomycin. CRL2321 cells were cultured in Roswell Park Memorial Institute 1640 medium (RPMI-1640, Mediatech Inc., Herndon, VA, USA) supplemented with 10% FBS, 100 IU/ml penicillin and 100 µg/ml streptomycin.

Assessment of metabolic activity via MTT assay

MCF10A, MCF10A-H, MDA-MB-231 and CRL-2321 cells were seeded at 105 cells/well in 96-well plates and allowed to attach overnight. Spent media was then replaced with fresh media containing treatment dosages between 0.15–0.2 mg/ml LOE. Media in control wells was replaced with fresh media without additives (No treatment, NT) or media containing the vehicle, methanol (Veh). Following 24 h treatment, 12 mM of MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (Life Technologies), was added to the cells followed by incubation for 4 h at 37°C. Formazan crystals formed at the end of incubation period were dissolved using dimethyl sulfoxide (DMSO) and plates were read at 570 nm with a reference wavelength of 630 nm.

Assessment of cell cycle arrest via flow cytometry

For cell cycle assay, MDA-MB-231 were seeded in 6-well plates at a concentration of 6×105 cells/well. Cells were allowed to attach overnight followed by replacement of spent media with serum-free media after 24 h to allow for synchronization of cell cycle. Cells were then treated in fresh media containing LOE at 0.06 and 0.15 mg/ml. Controls were treated in triplicate with either serum-free media as a positive control or media containing the vehicle, methanol. At the end of 36 h of treatment, media was aspirated, cells were washed once with 1X Phosphate Buffered Saline (PBS) and collected via trypsinization. Samples were centrifuged at 3,000 rpm for 5 min and supernatant was drained. Cell pellets were washed and resuspended in PBS containing 2.5 mM EDTA (1X PBS-EDTA buffer). Cells were then fixed by adding cell suspension drop-wise into microcentrifuge tubes containing 1 ml ice-cold 70% ethanol. Fixed cells were stored at -20°C until staining. For staining, fixed samples were centrifuged at 4000 rpm for 5 min and supernatant was drained following which cells were washed and resuspended in 200 µl 1X PBS-EDTA buffer. Samples were centrifuged at 4000 rpm for 5 min and supernatant was drained. Next, 200 µl Muse® Cell Cycle Staining Reagent (EMD Millipore, Billerica, MA, USA) containing propidium iodide and RNase A was added to each sample. Samples were stored at room temperature for at least 30 min to allow for adequate staining before being run through an FC500 flow cytometer (Beckman Coulter, Brea, CA, USA) at the Bindley Bioscience Center, Purdue University.

Assessment of apoptosis via flow cytometry

MDA-MB-231 were seeded in 12-well plates at a concentration of 2×105 cells/well. Cells were allowed to attach overnight followed by synchronization in serum-free media. Cells were then treated with fresh media containing LOE at 0.15 and 0.30 mg/ml for 24 h in triplicate while controls were treated with media containing methanol. Cells collected via trypsinization and centrifuged and supernatant was removed. Next, 100 µl Muse™ Annexin V Dead Cell Assay stain (EMD Millipore) was added to the cell pellet and samples were resuspended by gentle pipetting. Samples were incubated in the dark for 20 min, following which they were run through a Muse™ Cell Analyzer (EMD Millipore) using manufacturer's protocol.

Caspase-3/7 activation assay

MDA-MB-231 cells were seeded at 105 cells/well in a 96-well plate and allowed to attach overnight. LOE and control treatments were prepared in fresh media containing 5 µM IncuCyte™ Caspase-3/7 apoptosis assay reagent and added to the cells, following which the plate was placed in an IncuCyte® ZOOM live-cell analyzer. Cells were imaged recurrently for 24 h and quantified for changes in caspase-3/7 activation via measurement of fluorescence intensity at a wavelength of 400 nm.

Western blotting

MDA-MB-231 cells were seeded at 106 cells/well in 6-well plates and allowed to attach overnight. Cells were then treated with fresh media containing LOE at 0.06 or 0.15 mg/ml for 3, 6, 12 and 24 h intervals. Treated cells were collected in RIPA buffer and sonicated to obtain lysates. Cell lysates were cleared of debris by centrifugation and protein concentration of lysates was estimated via the bicinchoninic acid (BCA) assay. Next, 20 µg of protein from each sample was subjected to SDS-polyacrylamide gel electrophoresis, transferred onto a polyvinyl difluoride (PVDF) membrane and immunoblotted. Primary antibodies for cyclin D1, cellular Inhibitor of Apoptosis Protein 2 (cIAP2), RIP1, cleaved caspase-8 and cleaved Poly-ADP Ribose Polymerase (PARP) were purchased from Cell Signaling Technologies, Danvers, MA, USA. Cleaved caspase-3 antibody was purchased from Epigenomics, WA. β-actin and β-tubulin antibodies were obtained from Sigma-Aldrich and Abcam, Cambridge, MA, USA, respectively. Quantification was carried out using ImageJ software (35).

Statistical analysis

Data are presented as mean values with corresponding standard errors from various experiments. Analysis for statistical significance for MTT assay was carried out using one-way analysis of variance (ANOVA). Analysis for statistical significance for western blotting, flow cytometry and caspase-3/-7 assay were carried out using Student's unpaired two-tailed t-test.

Results

LOE decreases viability of triple-negative breast cancer cells

In order to study the differential response of normal, invasive, and malignant triple-negative cells to LOE treatment, MCF10A, MCF10A-H, MDA-MB-231 and CRL-2321 cells were treated with increasing dosages of LOE for 24 h and changes in cellular metabolic activity were quantified using the MTT assay and utilized as a reflection of cell viability (Fig. 1).

Relative to control treatment, there was a dose-dependent decrease in cell viability of MDA-MB-231 cells. LOE at a concentration of 0.15 mg/ml resulted in 50% reduction in MDA-MB-231 cell viability while a higher dosage of 0.2 mg/ml resulted in a 95% reduction in MDA-MB-231 cell viability. Importantly, LOE impacted cell viability of MDA-MB-231, as well as the other triple-negative invasive and malignant cell lines (MCF10A-H and CRL-2321, respectively) to a much greater degree than in normal MCF10A mammary epithelial cells. In contrast with MDA-MB-231 cells, there was only a 20% decrease in cell viability of MCF10A cells with 0.15 mg/ml LOE treatment and a 40% decrease with 0.2 mg/ml LOE. Curiously, CRL-2321 cells demonstrated greater sensitivity than MDA-MB-231 cells in their initial response to low doses of LOE, but viability in this cell line did not decrease further with increasing dosage.

LOE induces cell cycle arrest in triple-negative breast cancer cells

To quantify changes in cell cycle progression induced by LOE treatment, MDA-MB-231 cells were treated with LOE for 36 h. Control cells were treated with either serum-free media or growth media containing methanol. Cells were then collected and stained with propidium iodide followed by analysis using an FC500 flow cytometer. Representative raw data showing shifts in cell cycle phases under various treatments are shown in Fig. 2A.

Treatment with LOE at 0.15 mg/ml induced a significant shift away from the replication stage (S-phase) of the cell cycle as compared to vehicle-treated cells (Fig. 2B). Of the total cells counted, almost 20% shifted away from the S-phase and toward the G0/G1 phase upon LOE treatment at 0.15 mg/ml. Further, there was a striking resemblance in the S-phase distribution between 0.15 mg/ml LOE-treated cells and serum-starved cells (0.15 mg/ml LOE: 79.78% G0/G1; SF: 79.27% G0/G1), corroborating that LOE induces a halt in cell cycle progression in MDA-MB-231 cells.

LOE induces apoptosis in triple-negative breast cancer cells

Cell death may take the form of apoptosis, a programmed and controlled process involving the degradation and clearance of cellular constituents; or necrosis, a traumatic and inflammatory process which leads to the expulsion of cellular material into the extracellular environment (36). In order to analyze the impact of LOE on cell death in triple-negative breast cancer cells, MDA-MB-231 cells were treated with LOE for 24 h. Cells were stained with Annexin V/7-Aminoactinomycin D and evaluated in a Muse Cell Analyzer (Merck Millipore, Billerica. MA, USA). Representative raw data from various treatments indicating shifts from live to early (EA) and late (LA) apoptosis as well as necrosis (Dead) is shown in Fig. 3.

Treatment with LOE induced a significant decrease in live cells as well a significant increase in apoptosis but not necrosis in MDA-MB-231 cells. Furthermore, 40% of cells were apoptotic after 24 h of 0.15 mg/ml LOE treatment while ~80% of cells underwent apoptosis after 0.3 mg/ml LOE treatment over the same time period.

LOE impacts critical regulators of cell cycle progression

As seen from Fig. 2B, LOE treatment induced a shift toward the G0/G1 phase and away from the S phase of the cell cycle in MDA-MB-231 cells. The progression from G0/G1 is mediated by cyclin D1, with cellular inhibitor of apoptosis (cIAP) proteins playing a supportive role by suppressing apoptosis (37,38). As observed in Fig. 4B, there was a significant (5-fold) decrease in cyclin D1 protein at 24 h post-treatment. cIAP2 levels were significantly reduced by 40% after 12 h and by ~65% after 24 h.

Taken together, these results suggest that the halt in cell cycle progression observed upon treatment of MDA-MB-231 cells with LOE is brought about by its impact on cyclin D1 and cIAP2 expression.

LOE induces apoptosis via caspase-8/-3 activation

Based on the observation that LOE induces apoptosis and not necrosis in MDA-MB-231 cells (Fig. 3B), our goal was to determine if LOE induces the extrinsic pathway of apoptosis via caspase-8 activation. As shown in Fig. 5B, LOE induced the rapid cleavage of caspase-8 within 3 h post-treatment with PARP cleavage peaking at 6 h post-treatment.

To further understand apoptotic signaling, MDA-MB-231 cells were analyzed for executioner caspase-3/-7 activity via live cell imaging using the IncuCyte ZOOM system. These studies showed a marked visible increase in caspase-3/-7 activation upon LOE treatment. As quantified in Fig. 6, there was a significant increase in caspase-3/-7 activity 3 h post-treatment with 0.15 and 0.3 mg/ml LOE, with peak activity at approximately 10 h post-treatment. This was supported by western blotting time-course data, which indicated a peak in caspase-3 activity at 12 h post-treatment with 0.15 mg/ml LOE (data not shown).

LOE reduces RIP1 protein levels

TNBC is often characterized by constitutive activation of NF-κB signaling via recruitment of key effector proteins by RIP1 (39,40). Immunoblotting revealed a significant decrease of ~40% in RIP1 protein levels in lysates from MDA-MB-231 cells treated with LOE for 9 h as compared to controls (Fig. 7B).

Discussion

TNBC continues to be a monumental concern in women's health issues. As our understanding of the mechanisms that govern the initiation and progression of breast cancer grows, it has become apparent that conventional chemotherapeutics are limited by their inability to target subtypes that are inherently more aggressive while being drug resistant. One such subtype is TNBC, which lacks expression of ER, PR and HER2/neu, thereby rendering TNBC resistant to conventional endocrine-targeted therapy. In this study, we provide support that an extract from the tropical South American plant L. origanoides (LOE) may be a possible source for therapeutic agents against TNBCs. In brief, we utilized the MTT metabolic activity assay to show LOE induces a dose-dependent decrease in the viability of MDA-MB-231 mammary adenocarcinoma cells, a universal standard in vitro model of highly invasive and drug resistant TNBC. This effect was much greater compared to normal-like MCF10A mammary epithelial cells (Fig. 1). Our observation that LOE also significantly reduced cell viability in triple-negative Hras-transformed MCF10A invasive cells as well as the CRL2321 cell-line, recently shown to be the best in vitro phenotypic representative of grade 3 TNBC tumors (41), further confirms its anti-TNBC effects. This observation suggests the LOE induces molecular actions that target pathways specific to cancer cells. We also note that while CRL-2321 cells have a greater sensitivity to low doses of LOE than MDA-MB-231 cells, higher dosages of LOE do not induce decreases in CRL-2321 viability beyond 80%. Previous studies have shown that cancer cell line subpopulations and sub-lines can exhibit a high degree of transcriptome and phenotypic variability leading to observable differences in metastatic activity (42) and drug sensitivity (43). This suggests the possibility of heterogeneity within subpopulations of the CRL-2321 cell line allowing for compensatory survival pathways providing modest resistance to LOE treatment. The identification of novel mechanisms targeted by new small molecular inhibitors is of major interest in our continued studies of the actions of LOE.

Our study also quantified the molecular influences of the extract within cancer cells. Cyclin D1 is a protein which associates with cyclin-dependent kinase-4 and -6 (CDK-4 and -6) to form heterodimers that progress the cell cycle from G0/G1 to S phase (37). In addition, cellular inhibitor of apoptosis proteins (cIAPs) prevent apoptosis and permit cell cycle progression via binding and possibly inactivating executioner caspases, i.e., caspase-3 and -7 (38). cIAP2, is known to be constitutively expressed in MDA-MB-231 cells, and has been shown to protect against death-inducing ligands such as TNF-α by possibly enhancing NF-κB signaling through RIP1 activation as well as by binding and inhibiting caspase-3 (39). Herein, we observed that LOE induced G0/G1 phase halt in MDA-MB-231 cells (Fig. 2). Western blot analysis revealed the effects of LOE on cell cycle progression could be explained by a reduction in cyclin D1 and cIAP2 levels upon treatment of the cells with the extract (Fig. 4). These experiments begin to build an important framework for the actions of LOE on cell cycle regulation.

We next studied LOE's impact on MDA-MB-231 cell death. Upon triggering of the extrinsic pathway of apoptosis, procas-pase-8 is activated via dimerization-induced auto-cleavage and subsequently cleaves and activates the master 'executioner' caspase, caspase-3 (44,45). We revealed a potential mechanism behind the actions of the extract by showing LOE-induced apoptosis involved activation of caspase-8/-3 and cleavage of PARP (Figs. 5 and 6), hallmarks of extrinsic apoptotic pathway activation (36). Additionally, LOE induced apoptosis in these cells unaccompanied by necrotic cell death (Fig. 3). As cancer therapy is focused on preventing damage to non-targeted tissue areas, this result provides additional support for LOE as a source for potential therapeutics with desirable characteristics.

In previous work, Stashenko et al have characterized the major components of LOE (28). However, it remains to be defined which component, or combination of components, is responsible for the extract's apoptotic effects. Furthermore, it is imperative that major cellular pathways involved in LOE signal transduction be delineated. Based on previous studies involving components present in the LOE, one possible candidate pathway central to LOE-induced apoptosis is NF-κB signaling. Constitutive activation of this pathway has been previously linked to triple-negative breast cancer and blocking NF-κB signaling can spontaneously induce cell death via a caspase-8/-3-dependent mechanism or render cells susceptible to pro-apoptotic signals (40,46,47). In normal cells, ligand binding to TNFR1 is followed by rapid activation of the extrinsic pathway of apoptosis. However, in contrast to normal cells, ligand binding to TNFR1 in TNBC cells instead activates NF-κB signaling via membrane-recruitment of the scaffold protein RIP1, a Ser/Thr kinase (39). In brief, cIAP2 exerts E3 ubiquitin ligase activity to polyubiquitinate and activate RIP1, which goes on to phosphorylate the IKKα/β-NEMO complex. Active IKKs then phosphorylate IκB proteins which sequester p50/RelA (NF-κB) dimers to the cytoplasm, targeting the IκBs for polyubiquitination and subsequent degradation. This frees p50/RelA to translocate to the nucleus and act as a transcription factor for pro-survival genes such as cyclin D1 and cIAPs (48,49).

We hypothesized that components of LOE could interfere with NF-κB signaling, leading to a halt in the cell cycle and induction of apoptosis. Indeed, we showed LOE treatment resulted in a 40% decrease in protein levels of RIP1 in MDA-MB-231 cells (Fig. 7B). This may, at least in part, be explained by our previous observation that LOE treatment induced activation of caspase-8. Active caspase-8 has previously been shown to cleave RIP-1, thereby inhibiting pro-survival NF-κB signaling and inducing apoptosis (50). This result, coupled with our findings that LOE inhibits cyclin D1 and cIAP2, proteins activated by NF-κB signaling, while simultaneously stimulating caspase-8 dependent cell death, a characteristic of NF-κB inhibition, sheds critical light on the mechanistic foreground of LOE action. All together, these data highlight the potential of L. origanoides extract and its major constituents as cancer therapeutics.

In summary, we have shown that LOE promotes apoptosis in TNBC cells, while having a much reduced impact on cell death in normal mammary cells. We also found that LOE reduces cell viability, alters the cell cycle and primarily leads to tumor cell apoptosis and not necrosis. Further, we established some of the mechanisms of LOE actions, showing that it suppresses markers of cell cycle progression and cell survival while inducing the extrinsic pathway of apoptosis via caspase-8/-3 activation. Finally, we revealed that levels of RIP1, an upstream effector of pro-survival NF-κB signaling, are reduced upon LOE treatment. These data collectively support that LOE can be a valuable source for development of novel anticancer agents. Moving forward, beyond fully describing the cellular mechanisms of LOE action, a top priority will be identifying the major component or combination of components responsible for its bioactivity. Plant-derived compounds offer a vast source of small molecule inhibitors that could be used both in their native state as well as chemically modified to optimize their pharmacological activity. The bio-reactivity of the LOE-treatment in TNBC cells in vitro supports the idea that future treatments for individual cancer subtypes lie in naturally-derived chemicals.

Acknowledgments

The authors would like to thank Ms. Gabriela Sincich and Ms. Shelli Taylor from the Department of Biological Sciences, Purdue University, for their help in editing this manuscript. The authors gratefully acknowledge the Flow Cytometry and Cell Separation Facility and support from the Purdue University Center for Cancer Research, NIH grant P30 CA023168. The LOE used in this work was obtained within contract no. RC-0572-2012 supported by 'Patrimonio Autónomo Fondo Nacional de Financiamiento para la Ciencia, la Tecnología y la Innovación, Francisco José de Caldas'. The Ministerio de Ambiente y Desarrollo Sostenible (MADS) of Colombia supported the project through permits to access genetic resources and derivatives products during scientific research (contract no. 101, resolution no. 0812).

Glossary

Abbreviations

Abbreviations:

cIAP

cellular inhibitor of apoptosis

ER

estrogen receptor

Her2/neu

human epidermal growth factor receptor 2

LOE

Lippia origanoides extract

NF-κB

nuclear factor κB

PARP

poly-ADP ribose polymerase

PR

progesterone receptor

TNBC

triple-negative breast cancer

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2016. CA Cancer J Clin. 66:7–30. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Jordan VC: The role of tamoxifen in the treatment and prevention of breast cancer. Curr Probl Cancer. 16:129–176. 1992.PubMed/NCBI

3 

Buzdar A and Howell A: Advances in aromatase inhibition: Clinical efficacy and tolerability in the treatment of breast cancer. Clin Cancer Res. 7:2620–2635. 2001.PubMed/NCBI

4 

Romond EH, Perez EA, Bryant J, Suman VJ, Geyer CE Jr, Davidson NE, Tan-Chiu E, Martino S, Paik S, Kaufman PA, et al: Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N Engl J Med. 353:1673–1684. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Dent R, Hanna WM, Trudeau M, Rawlinson E, Sun P and Narod SA: Pattern of metastatic spread in triple-negative breast cancer. Breast Cancer Res Treat. 115:423–428. 2009. View Article : Google Scholar

6 

Brenton JD, Carey LA, Ahmed AA and Caldas C: Molecular classification and molecular forecasting of breast cancer: Ready for clinical application? J Clin Oncol. 23:7350–7360. 2005. View Article : Google Scholar : PubMed/NCBI

7 

Cancer Genome Atlas Network: Comprehensive molecular portraits of human breast tumours. Nature. 490:61–70. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Qi X, Yin N, Ma S, Lepp A, Tang J, Jing W, Johnson B, Dwinell MB, Chitambar CR and Chen G: p38γ MAPK is a therapeutic target for triple-negative breast cancer by stimulation of cancer stem-like cell expansion. Stem Cells. 33:2738–2747. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Nakshatri H, Bhat-Nakshatri P, Martin DA, Goulet RJ Jr and Sledge GW Jr: Constitutive activation of NF-kappaB during progression of breast cancer to hormone-independent growth. Mol Cell Biol. 17:3629–3639. 1997. View Article : Google Scholar : PubMed/NCBI

10 

Sero JE, Sailem HZ, Ardy RC, Almuttaqi H, Zhang T and Bakal C: Cell shape and the microenvironment regulate nuclear translocation of NF-κB in breast epithelial and tumor cells. Mol Syst Biol. 11:7902015. View Article : Google Scholar

11 

Khoshnan A, Tindell C, Laux I, Bae D, Bennett B and Nel AE: The NF-kappa B cascade is important in Bcl-xL expression and for the anti-apoptotic effects of the CD28 receptor in primary human CD4+ lymphocytes. J Immunol. 165:1743–1754. 2000. View Article : Google Scholar : PubMed/NCBI

12 

Wang CY, Mayo MW, Korneluk RG, Goeddel DV and Baldwin AS Jr: NF-kappaB antiapoptosis: Induction of TRAF1 and TRAF2 and c-IAP1 and c-IAP2 to suppress caspase-8 activation. Science. 281:1680–1683. 1998. View Article : Google Scholar : PubMed/NCBI

13 

Huber MA, Azoitei N, Baumann B, Grünert S, Sommer A, Pehamberger H, Kraut N, Beug H and Wirth T: NF-kappaB is essential for epithelial-mesenchymal transition and metastasis in a model of breast cancer progression. J Clin Invest. 114:569–581. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Hartman ZC, Poage GM, den Hollander P, Tsimelzon A, Hill J, Panupinthu N, Zhang Y, Mazumdar A, Hilsenbeck SG, Mills GB, et al: Growth of triple-negative breast cancer cells relies upon coordinate autocrine expression of the proinflammatory cytokines IL-6 and IL-8. Cancer Res. 73:3470–3480. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Hinz M, Krappmann D, Eichten A, Heder A, Scheidereit C and Strauss M: NF-kappaB function in growth control: Regulation of cyclin D1 expression and G0/G1-to-S-phase transition. Mol Cell Biol. 19:2690–2698. 1999. View Article : Google Scholar : PubMed/NCBI

16 

Yousef EM, Tahir MR, St-Pierre Y and Gaboury LA: MMP-9 expression varies according to molecular subtypes of breast cancer. BMC Cancer. 14:6092014. View Article : Google Scholar : PubMed/NCBI

17 

Kagoya Y, Yoshimi A, Kataoka K, Nakagawa M, Kumano K, Arai S, Kobayashi H, Saito T, Iwakura Y and Kurokawa M: Positive feedback between NF-κB and TNF-α promotes leukemia-initiating cell capacity. J Clin Invest. 124:528–542. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Gill PS, Rarick M, McCutchan JA, Slater L, Parker B, Muchmore E, Bernstein-Singer M, Akil B, Espina BM, Krailo M, et al: Systemic treatment of AIDS-related Kaposi's sarcoma: Results of a randomized trial. Am J Med. 90:427–433. 1991. View Article : Google Scholar : PubMed/NCBI

19 

Saville MW, Lietzau J, Pluda JM, Feuerstein I, Odom J, Wilson WH, Humphrey RW, Feigal E, Steinberg SM, Broder S, et al: Treatment of HIV-associated Kaposi's sarcoma with paclitaxel. Lancet. 346:26–28. 1995. View Article : Google Scholar : PubMed/NCBI

20 

Ahmad A, Banerjee S, Wang Z, Kong D and Sarkar FH: Plumbagin-induced apoptosis of human breast cancer cells is mediated by inactivation of NF-kappaB and Bcl-2. J Cell Biochem. 105:1461–1471. 2008. View Article : Google Scholar : PubMed/NCBI

21 

Jin ML, Park SY, Kim YH, Park G and Lee SJ: Halofuginone induces the apoptosis of breast cancer cells and inhibits migration via downregulation of matrix metalloproteinase-9. Int J Oncol. 44:309–318. 2014. View Article : Google Scholar

22 

Pozo-Guisado E, Merino JM, Mulero-Navar ro S, Lorenzo-Benayas MJ, Centeno F, Alvarez-Barrientos A and Fernandez-Salguero PM: Resveratrol-induced apoptosis in MCF-7 human breast cancer cells involves a caspase-independent mechanism with downregulation of Bcl-2 and NF-kappaB. Int J Cancer. 115:74–84. 2005. View Article : Google Scholar : PubMed/NCBI

23 

Singh S and Aggarwal BB: Activation of transcription factor NF-kappa B is suppressed by curcumin (diferuloylmethane) [corrected]. J Biol Chem. 270:24995–25000. 1995. View Article : Google Scholar : PubMed/NCBI

24 

Tse AK, Wan CK, Shen XL, Yang M and Fong WF: Honokiol inhibits TNF-alpha-stimulated NF-kappaB activation and NF-kappaB-regulated gene expression through suppression of IKK activation. Biochem Pharmacol. 70:1443–1457. 2005. View Article : Google Scholar : PubMed/NCBI

25 

Chen KS, Shi MD, Chien CS and Shih YW: Pinocembrin suppresses TGF-β1-induced epithelial-mesenchymal transition and metastasis of human Y-79 retinoblastoma cells through inactivating αvβ3 integrin/FAK/p38α signaling pathway. Cell Biosci. 4:412014. View Article : Google Scholar

26 

Pascual ME, Slowing K, Carretero E, Sánchez Mata D and Villar A: Lippia: traditional uses, chemistry and pharmacology: a review. J Ethnopharmacol. 76:201–214. 2001. View Article : Google Scholar : PubMed/NCBI

27 

Stashenko EE, Martínez JR, Ruíz CA, Arias G, Durán C, Salgar W and Cala M: Lippia origanoides chemotype differentiation based on essential oil GC-MS and principal component analysis. J Sep Sci. 33:93–103. 2010. View Article : Google Scholar

28 

Stashenko EE, Martínez JR, Cala MP, Durán DC and Caballero D: Chromatographic and mass spectrometric characterization of essential oils and extracts from Lippia (Verbenaceae) aromatic plants. J Sep Sci. 36:192–202. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Legault J and Pichette A: Potentiating effect of beta-caryophyllene on anticancer activity of alpha-humulene, isocaryophyllene and paclitaxel. J Pharm Pharmacol. 59:1643–1647. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Aristatile B, Al-Assaf AH and Pugalendi KV: Carvacrol suppresses the expression of inflammatory marker genes in D-galactosamine-hepatotoxic rats. Asian Pac J Trop Med. 6:205–211. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Greiner JF, Müller J, Zeuner MT, Hauser S, Seidel T, Klenke C, Grunwald LM, Schomann T, Widera D, Sudhoff H, et al: 1,8-Cineol inhibits nuclear translocation of NF-κB p65 and NF-κB-dependent transcriptional activity. Biochim Biophys Acta. 1833:2866–2878. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Lee JC, Kundu JK, Hwang DM, Na HK and Surh YJ: Humulone inhibits phorbol ester-induced COX-2 expression in mouse skin by blocking activation of NF-kappaB and AP-1: IkappaB kinase and c-Jun-N-terminal kinase as respective potential upstream targets. Carcinogenesis. 28:1491–1498. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Liang D, Li F, Fu Y, Cao Y, Song X, Wang T, Wang W, Guo M, Zhou E, Li D, et al: Thymol inhibits LPS-stimulated inflammatory response via down-regulation of NF-κB and MAPK signaling pathways in mouse mammary epithelial cells. Inflammation. 37:214–222. 2014. View Article : Google Scholar

34 

Kim C, Cho SK, Kim KD, Nam D, Chung WS, Jang HJ, Lee SG, Shim BS, Sethi G and Ahn KS: β-Caryophyllene oxide potentiates TNFα-induced apoptosis and inhibits invasion through down-modulation of NF-κB-regulated gene products. Apoptosis. 19:708–718. 2014. View Article : Google Scholar

35 

Schneider CA, Rasband WS and Eliceiri KW: NIH Image to ImageJ: 25 years of image analysis. Nat Methods. 9:671–675. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Portt L, Norman G, Clapp C, Greenwood M and Greenwood MT: Anti-apoptosis and cell survival: A review. Biochim Biophys Acta. 1813:238–259. 2011. View Article : Google Scholar

37 

Baldin V, Lukas J, Marcote MJ, Pagano M and Draetta G: Cyclin D1 is a nuclear protein required for cell cycle progression in G1. Genes Dev. 7:812–821. 1993. View Article : Google Scholar : PubMed/NCBI

38 

Salvesen GS and Duckett CS: IAP proteins: Blocking the road to death's door. Nat Rev Mol Cell Biol. 3:401–410. 2002. View Article : Google Scholar : PubMed/NCBI

39 

Bertrand MJ, Milutinovic S, Dickson KM, Ho WC, Boudreault A, Durkin J, Gillard JW, Jaquith JB, Morris SJ and Barker PA: cIAP1 and cIAP2 facilitate cancer cell survival by functioning as E3 ligases that promote RIP1 ubiquitination. Mol Cell. 30:689–700. 2008. View Article : Google Scholar : PubMed/NCBI

40 

Yamaguchi N, Ito T, Azuma S, Ito E, Honma R, Yanagisawa Y, Nishikawa A, Kawamura M, Imai J, Watanabe S, et al: Constitutive activation of nuclear factor-kappaB is preferentially involved in the proliferation of basal-like subtype breast cancer cell lines. Cancer Sci. 100:1668–1674. 2009. View Article : Google Scholar : PubMed/NCBI

41 

Grigoriadis A, Mackay A, Noel E, Wu PJ, Natrajan R, Frankum J, Reis-Filho JS and Tutt A: Molecular characterisation of cell line models for triple-negative breast cancers. BMC Genomics. 13:6192012. View Article : Google Scholar : PubMed/NCBI

42 

Nguyen A, Yoshida M, Goodarzi H and Tavazoie SF: Highly variable cancer subpopulations that exhibit enhanced transcriptome variability and metastatic fitness. Nat Commun. 7:112462016. View Article : Google Scholar : PubMed/NCBI

43 

Leung E, Kannan N, Krissansen GW, Findlay MP and Baguley BC: MCF-7 breast cancer cells selected for tamoxifen resistance acquire new phenotypes differing in DNA content, phospho-HER2 and PAX2 expression, and rapamycin sensitivity. Cancer Biol Ther. 9:717–724. 2010. View Article : Google Scholar : PubMed/NCBI

44 

Peter ME and Krammer PH: The CD95(APO-1/Fas) DISC and beyond. Cell Death Differ. 10:26–35. 2003. View Article : Google Scholar : PubMed/NCBI

45 

Beaudouin J, Liesche C, Aschenbrenner S, Hörner M and Eils R: Caspase-8 cleaves its substrates from the plasma membrane upon CD95-induced apoptosis. Cell Death Differ. 20:599–610. 2013. View Article : Google Scholar : PubMed/NCBI

46 

Yamamoto M, Taguchi Y, Ito-Kureha T, Semba K, Yamaguchi N and Inoue J: NF-κB non-cell-autonomously regulates cancer stem cell populations in the basal-like breast cancer subtype. Nat Commun. 4:22992013. View Article : Google Scholar

47 

Micheau O and Tschopp J: Induction of TNF receptor I-mediated apoptosis via two sequential signaling complexes. Cell. 114:181–190. 2003. View Article : Google Scholar : PubMed/NCBI

48 

Guttridge DC, Albanese C, Reuther JY, Pestell RG and Baldwin AS Jr: NF-kappaB controls cell growth and differentiation through transcriptional regulation of cyclin D1. Mol Cell Biol. 19:5785–5799. 1999. View Article : Google Scholar : PubMed/NCBI

49 

You M, Ku PT, Hrdlicková R and Bose HR Jr: ch-IAP1, a member of the inhibitor-of-apoptosis protein family, is a mediator of the antiapoptotic activity of the v-Rel oncoprotein. Mol Cell Biol. 17:7328–7341. 1997. View Article : Google Scholar : PubMed/NCBI

50 

Lin Y, Devin A, Rodriguez Y and Liu ZG: Cleavage of the death domain kinase RIP by caspase-8 prompts TNF-induced apoptosis. Genes Dev. 13:2514–2526. 1999. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2017
Volume 51 Issue 6

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Raman V, Fuentes Lorenzo JL, Stashenko EE, Levy M, Levy MM and Camarillo IG: Lippia origanoides extract induces cell cycle arrest and apoptosis and suppresses NF-κB signaling in triple-negative breast cancer cells. Int J Oncol 51: 1801-1808, 2017
APA
Raman, V., Fuentes Lorenzo, J.L., Stashenko, E.E., Levy, M., Levy, M.M., & Camarillo, I.G. (2017). Lippia origanoides extract induces cell cycle arrest and apoptosis and suppresses NF-κB signaling in triple-negative breast cancer cells. International Journal of Oncology, 51, 1801-1808. https://doi.org/10.3892/ijo.2017.4169
MLA
Raman, V., Fuentes Lorenzo, J. L., Stashenko, E. E., Levy, M., Levy, M. M., Camarillo, I. G."Lippia origanoides extract induces cell cycle arrest and apoptosis and suppresses NF-κB signaling in triple-negative breast cancer cells". International Journal of Oncology 51.6 (2017): 1801-1808.
Chicago
Raman, V., Fuentes Lorenzo, J. L., Stashenko, E. E., Levy, M., Levy, M. M., Camarillo, I. G."Lippia origanoides extract induces cell cycle arrest and apoptosis and suppresses NF-κB signaling in triple-negative breast cancer cells". International Journal of Oncology 51, no. 6 (2017): 1801-1808. https://doi.org/10.3892/ijo.2017.4169