Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Sep 1.
Published in final edited form as: Cytokine. 2015 Jun 20;75(1):117–126. doi: 10.1016/j.cyto.2014.12.007

Diallyl disulfide inhibits TNFα induced CCL2 release through MAPK/ERK and NF-Kappa-B Signaling

D Bauer 1, N Redmon 1, E Mazzio 1, E Taka 1, JS Reuben 1, A Day 1, S Sadrud-Din 1, H Flores-Rozas 1, KFA Soliman 1, S Darling-Reed 1,*
PMCID: PMC4532635  NIHMSID: NIHMS662693  PMID: 26100848

Abstract

TNFα receptors are constitutively overexpressed in tumor cells, correlating to sustain elevated NFκB and monocyte chemotactic protein-1 (MCP-1/CCL2) expression. The elevation of CCL2 evokes aggressive forms of malignant tumors marked by tumor associated macrophage (TAM) recruitment, cell proliferation, invasion and angiogenesis. Previously, we have shown that the organo-sulfur compound diallyl disulfide (DADS) found in garlic (Allium sativum) attenuates TNFα induced CCL2 production in MDA-MB-231 cells. In the current study, we explored the signaling pathways responsible for DADS suppressive effect on TNFα mediated CCL2 release using PCR Arrays, RT-PCR and western blots. The data in this study show that TNFα initiates a rise in NFκB mRNA, which is not reversed by DADS. However, TNFα induced heightened expression of IKKε and phosphorylated ERK. The expression of these proteins corresponds to increased CCL2 release that can be attenuated by DADS. CCL2 induction by TNFα was also lessened by inhibitors of p38 (SB202190) and MEK (U0126) but not JNK (SP 600125), all of which were suppressed by DADS. In conclusion, the obtained results indicate that DADS down regulates TNFα invoked CCL2 production primarily through reduction of IKKε and phosphorylated-ERK, thereby impairing MAPK/ERK, and NFκB pathway signaling. Future research will be required to evaluate the effects of DADS on the function and expression of TNFα surface receptors.

Keywords: diallyl disulfide, tumor necrosis factor alpha, monocyte chemoattractant protein 1, nuclear factor kappa b, map kinase b

1. INTRODUCTION

Metastatic breast cancer brings together rapid tumor proliferation, detachment and development of secondary tumors with acquired characteristics of the primary tumor. Chemokines such as monocyte chemotactic protein-1 (MCP-1), known as CC chemokine-2 (CCL2) play a critical role in this process. These chemokines recruit monocytes that differentiate into tumor-associated macrophages (TAMs) which subsequently release substances needed for tissue remodeling, angiogenesis and metastasis [13]. TAMs can also directly release more TNFα, an inflammatory cytokine [4], furthering the processes of production/release of CCL2 in diverse tumor tissue.[5].

Previously we have shown that diallyl disulfide (DADS), found in garlic (Allium sativum), attenuates TNFα induced CCL2 production in human breast cancer cells. It is likely that TNFα induced CCL2 production occurs through up regulation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). These three components are concurrently expressed to a greater degree in aggressively advanced tumors marked by TAM recruitment [68] and elevated TNF-receptors (TNFRs) on diverse human cancers [9]. Drugs or compounds such as DADS that antagonize these effects are becoming significant therapeutic vehicles. These include infliximab (Remicade), adalimumab (Humira), anti-TNF antibodies [1011], all of which prevent tumor infiltrating leucocytes [12]. Therefore, the purpose of this study was to determine if the garlic constituent diallyl disulfide, could impact the MDA-MB-231 cells since they are the most studied TNBC to date. In addition, many researchers have used this cell model because it mimics aggressive nature of clinical isolates and have displayed great ability to metastasize in xenograft models. Nakagawa et al. [13] demonstrated the ability of DADS to inhibit tumor growth through its modulation of the apoptotic genes in this cell line. Other studies have used this cell line to study the mechanisms involved in the immune response [14,15]. Moreover, in this study we further explore the mechanism behind the inhibitory effect by DADS on TNFα induced CCL2 release in human breast carcinoma cells, with focus on MAPK/ERK NFκB signaling.

2. MATERIALS AND METHODS

Cell lines, chemicals and reagents: Triple negative human breast tumor (MDA-MB-231) cells were obtained from American Type Culture Collection (Rockville, MD). Dulbecco’s Modified Eagle Medium (DMEM) media, fetal bovine serum (FBS) and penicillin/streptomycin were all obtained from Invitrogen (Carlsbad, CA). Recombinant human TNFα was purchased from RayBiotech (RayBiotech Inc., Norcross, GA, USA). Diallyl disulfide (>80%) was purchased from Sigma-Aldrich (St. Louis, MO, USA).

2.1. Cell culture and Treatment

MDA-MB-231 cells were cultured in 75cm2 or 175cm2 flasks containing DMEM media supplemented with 10% FBS and 1% pen/strep (10,000u/ml penicillin G sodium, 10,000μg/ml streptomycin sulfate). Cells were grown in an environment of 37°C with humidified 95% air and 5% CO2. Control cells received vehicle only (0.01% ethanol). DADS treated cells received 100 μM DADS in vehicle and 40ng of TNFα was given to TNFα-treated and co-treated cells. Cells were incubated for 24 hrs. after treatment. In this study, 100μM was used as the dose for DADS concentration. Although some studies have shown that different concentrations of garlic components exert different responses, such as increasing proliferation and tumor growth, in our laboratory cell viability studies were conducted to determine a working concentration and we recently reported that the 100μM dose was the optimum dose for this cell line [16]. The present study is a continuation of the study performed by Bauer et al. [16]. Moreover, initial studies were done using both 100μM and 400μM with approximately the same effect on CCL2 release. Additionally in our previous studies, we demonstrated an optimum overall response using lower doses of DADS in MCF10A human epithelial cells [17]. Therefore, we used 100μM since it would be less likely to have toxic effects on the normal cells at this dose level.

2.2. Inhibition study

Cultured MDA-MB-231 cells were treated for 24 hours with DADS with and without TNFα treatment at above conditions. Additionally, cells were co-treated with inhibitors of JNK, MEK and p38 at concentrations of 10 μM, 2 μM and 2 μM, respectively. The inhibitors for JNK (SP600125), MEK (U0126) and p38 (SB202190) were purchased from Sigma Aldrich (St. Louis, MO). Cells were detached and the lysate collected.

2.3. ELISA: CCL2 detection

Supernatants from resting and stimulated (24 hrs) MDA-MB-231 cells were collected and centrifuged at 1000× g for 5 min at 4°C. Specific ELISA was performed using MCP-1/CCL2 ELISA kit (Raybiotech, Norcross, GA, USA) following manufacturer’s instructions. Briefly, 100 μl of supernatants from samples and standards were added to 96 well plates pre-coated with capture antibody. After incubation 100μl of prepared biotinylated antibody mixture was added to each well. After 1 hour, mixture was decanted and 100 μl streptavidin solution was placed in each well and incubated. Substrate reagent (100 μl) was then added to each well for 30 min followed by the addition of 50 μl stop solution. Plates were read at 450nm using a UV microplate reader.

2.4. Western Blot: MAPK/ERK pathway and IKKE

Total cell protein concentrations from MDA-MB-231 cells treated with DADS, with and without TNFα co-treatment for 24 hrs, was determined using a modified Bio-Rad “DC” protein assay (Bio-Rad Laboratories, Hercules, CA, USA). A series of concentration standards ranging from 0–20 μg/ml were prepared using IgG. Test samples were prepared by adding 5 μl of a 1/10 dilution of the cell lysate to 795 μl H2O. The standards and samples were mixed with 200 μl Bio-Rad “DC” protein assay dye concentrate and thoroughly mixed by vortexing. Following incubation for 5 minutes at room temperature, the samples were vortexed again and 200 μl of each loaded into a 96-well plate. Protein concentrations were quantified at a wavelength of 595 nm with the Power Wave X 340 microplate reader equipped with KC4 v3.0 PowerReports software (Bio-Tek Instruments, Winooski, VT, USA).

Cell lysates were separated by electrophoresis on 10% SDS-polyacrylamide gels and then transferred to Immobilon-P PVDF membranes. Equal loading was verified by staining with Ponceau S (Sigma-Aldrich Chemical Co, St. Louis, MO). Blots were blocked at 4°C overnight in 5% Carnation Instant Milk in Tris-buffered saline with 0.05% Tween 20 in PBS (PBST) and then incubated overnight at 4°C with mouse anti-human p38 MAPK, ERK and JNK affinity purified antibody (IMGENEX, San Diego, CA). Membranes were washed with PBST and incubated overnight with anti-goat IgG-horseradish peroxidase (Santa Cruz Biotechnology, CA) in PBST overnight at 4°C. Protein loading was monitored in each gel lane by probing the membranes with anti-GAPDH antibodies (R & D Systems, Minneapolis, MN). Immunoblot images were obtained using a Flour-S Max Multimager (Bio-Rad Laboratories, Hercules, CA). Lane density data was acquired with Quantity One Software (Bio-Rad Laboratories, Hercules, CA).

2.5. RT-PCR

MDA-MB-231 cells treated with or without DADS, were subcultured in 6-well plates until confluent. Cells were lysed with 1 ml Trizol reagent. Chloroform (0.2 ml) was added to the lysed samples, tubes were shaken, incubated at 15–30°C for 2–3 min and centrifuged at 10,000 × g for 15 min. at 2–8°C. The aqueous phase was transferred to a fresh tube and the RNA precipitated by mixing 0.5 ml isopropyl alcohol. After incubation the samples were centrifuged, the supernatant was removed and the RNA pellets were washed with 75% ethanol. The samples were mixed before being centrifuged at 7,500 × g for 5 min. at 2–8°C. The RNA pellet was dried and dissolved in RNase-free water and incubated for 10 min. at 55–60°C.

RT reaction

RNA (5 μg/10 μl) was heated for 10 min. then quenched on ice before use. The following components were added to the reaction: 10 μl heat denatured RNA, 3.0 μl 10 x PCR buffer, 2.5 μl 10 mM dNTPs, 6.0 μl 25 mM MgCl2, 1.0 μl random primers, 0.5 μl SuperScript II reverse transcriptase and 17.0 μl water. Samples were allowed to sit for 10 min. at 25°C then incubated for 1 hr. at 42°C. The cDNA was denatured at 95°C and placed on ice. PCR reaction: The following components were mixed in a 0.5 ml PCR tube: 6.0 μl cDNA product, 1.5 μl 10 x PCR buffer, 0.2 μl Taq polymerase, 0.5 μl primer and 10.3 μl water. PCR will be performed with 30 cycles of denaturation: 30 sec. at 95°C; annealing: 45 sec. at 60°C; and extension 60 sec. at 72°C using BioDoc-it System (UVP, Upland CA, USA). cDNA synthesis and Real-Time PCR was performed using First Strand cDNA synthesis kit/SABiosciences RT2 qPCR Master Mix from Qiagen (Gaithersburg, Md., USA) according to manufacturers instructions.

2.6. Statistical Analysis

Statistical analysis on the data was determined by Graph Pad Prism 5.0. All data was expressed as mean ± standard error from at least 3 independent experiments. Differences between mean values were analyzed by a one-way analysis of variance (ANOVA) with Dunnett’s Multiple Comparison test, *p<0.05; **p<0.01.

3. RESULTS

3.1 Effect of DADS on the TNF-mediated release of CCL2 in MDA-MB-231 cells

MDA-MB-231 cells were stimulated with TNFα at sub-lethal concentrations (40 ng/ml) and the time-dependent release of CCL2 was monitored. As indicated in Figure 1, the levels of CCL2 remained unchanged for the first 1.25 hours, and began to accumulate after 3 hours with peak levels, approximately 3-fold higher than basal levels, achieved at 24 hours post induction. This time point provides a level of accumulation that is adequate for further analysis (Figure 1). As reported in the literature the induction of TNFα-induced CCL2 is mediated via MAPK signaling pathways (Figure 2A). To evaluate its effect on CCL2 release, cells were treated with DADS in the absence and presence of MAP kinase inhibitors. As shown in Figure 2, neither DADS nor the MAPK inhibitors in combination with DADS, affected CCL2 basal levels in the absence of TNFα-induction. Upon stimulation with TNFα, a significant increase in CCL2 was not prevented by inhibition of JNK (Figure 2). DADS alone reduced the accumulation of CCL2 in TNFα-induced cells, and further enhanced the reduction exerted by p38 and MEK1 inhibitors when co-treated. These data suggest a controlling role for TNFα-induced CCL2 via MAPK involving MEK and P38 signaling, but not JNK.

Figure 1. Time course of the TNFα-dependent accumulation of CCL2 in MDA-MB-231 cells.

Figure 1

MDA-MB-231 cells were exposed to sublethal levels of TNFα (40ng/ml) and CCL2 release was monitored as described in Materials and Methods. The data is presented as % Ctrl at Timezero, displayed as the Mean ± S.E.M., n=4. Differences from Control were determined using a determined by a one-way ANOVA, with a Tukey post hoc test. *p<0.05.

Figure 2.

Figure 2

Figure 2A. Kegg Diagram interconnecting MAPK signaling with CCL2 release. CCL2 release is initiated by TNFα acting on TNFR1 and contributes to leukocyte recruitment. The factors analyzed in this study are highlighted.

Figure 2B. Effect of DADS and MAPK signaling inhibitors on CCL2 release in MDA-MB-231 cells. Additive or synergistic effects of MAPK inhibitors on DADS-treated (100μM), TNFα-treated (40ng/ml) and co-treated MDA-MB-231 cells after 24 hrs. The data are presented as CCL2 release (OD 450nm) and represent the Mean ± S.E.M. n=4. Significance of differences from the control in both groups was determined by T-test. *p<0.05.

3.2 DADS does not affect the protein levels of MAPK factors, but attenuates the TNFα-dependent phosphorylation of ERK and the induction of IKKε in MDA-MB-231 cells

Based on the ability of DADS to enhance the effect of MAPK inhibitors we evaluated the protein expression levels of the signaling proteins involved in the pathway. Neither DADS, or TNFα, nor their combination considerably altered the protein levels of JNK1/2/3, p38 or ERK (Figure 3). Evaluation of the phosphorylation status of ERK reveals that DADS does not affect its phosphorylation status but significantly attenuates the TNFα-dependent phosphorylation. In addition, DADS also reduced TNFα-dependent induction of IKKε (Figure 3).

Figure 3. Evaluation total or phosphorylated proteins involved with NFκB Signaling Pathway.

Figure 3

DADS-treated (100μM), TNFα-treated (40ng/ml) and co-treated MDA-MB-231 cell lysates were evaluated for the protein levels of JNK1/2/3, p38, ERK and IKKε. Phosphorylation status of ERK was also determined as described in the Materials and methods.

3.3 Evaluation of the effect of DADS on the mRNA profile of NFκB signaling

To investigate if DADS inhibition of CCL2 release in the presence of TNFα is mitigated by inhibition of NFκB signaling, we analyzed mRNA profiles of the pathway components using the NFκB Signaling Pathway RT2 Profiler PCR Array PAMM-025Z (Qiagen, Gaithersburg, Md., USA) (Table 1). Briefly, the array includes genes involved with Rel, NFκB, and IκB families, NFκB-responsive genes, ligand, receptors, kinases and transcription factors that propagate the signal. The data showed no statistical differences for baseline values between controls vs DADS (Figure 4A) for any gene in the array. The effects of TNFα on mRNA expression showed significant elevation in Bcl10, IL1b, Csf1, Crebbp, Fas, Tnfrsf1a, IL-10, Ikbkγ and Tnfrsf1b (Figure 4B). Genes were entered into bio-informatic analysis (Database for Annotation, Visualization and Integrated Discovery (DAVID) v6.7) to identify major systems affected (Table 2), where classifications provided by DAVID enrichment scores are presented for those averaging less than p<0.01 [18, 19]. There were significant differences found between Control vs TNFα-treated groups in the upward direction showing statistical probabilities for: regulation of cytokine biosynthesis, positive regulator of NFκB signaling, cell death and Nod-Like receptor signaling. Effect of TNFα vs TNFα/DADS shows DADS downregulation of Ccl2, Casp8 and Tradd at equal to or greater than 2-fold with p<0.05 (Figure 4C). We further investigated the effect of DADS on the TNFα NFκB expression pattern using RT-PCR. As shown in Figure 5, there were no changes in the mRNA expression for NFκB1 corroborating the results of the mRNA profile analysis.

Table 1.

Gene Table.

NFκB Signaling Pathway RT2 Profiler PCR Array PAMM – 025Z plate layout. Well position, gene identifiers, official gene symbol and gene description.

Position Unigene GeneBank Symbol Description
A01 Mm.301626 NM_007428 Agt Angiotensinogen (serpin peptidase inhibitor, clade A, member 8)
A02 Mm.6645 NM_009652 Akt1 Thymoma viral proto-oncogene 1
A03 Mm.676 NM_007497 Atf1 Activating transcription factor 1
A04 Mm.209903 NM_009715 Atf2 Activating transcription factor 2
A05 Mm.239141 NM_009740 Bcl10 B-cell leukemia/lymphoma 10
A06 Mm.479217 NM_009742 Bcl2a1a B-cell leukemia/lymphoma 2 related protein A1a
A07 Mm.238213 NM_009743 Bcl2l1 Bcl2-like 1
A08 Mm.439658 NM_033601 Bcl3 B-cell leukemia/lymphoma 3
A09 Mm.2026 NM_007464 Birc3 Baculoviral IAP repeat-containing 3
A10 Mm.17629 NM_130859 Card10 Caspase recruitment domain family, member 10
A11 Mm.46187 NM_175362 Card11 Caspase recruitment domain family, member 11
A12 Mm.1051 NM_009807 Casp1 Caspase 1
B01 Mm.336851 NM_009812 Casp8 Caspase 8
B02 Mm.290320 NM_011333 Ccl2 Chemokine (C-C motif) ligand 2
B03 Mm.284248 NM_013653 Ccl5 Chemokine (C-C motif) ligand 5
B04 Mm.367714 NM_001033126 Cd27 CD27 antigen
B05 Mm.271833 NM_011611 Cd40 CD40 antigen
B06 Mm.486313 NM_009805 Cflar CASP8 and FADD-like apoptosis regulator
B07 Mm.3996 NM_007700 Chuk Conserved helix-loop-helix ubiquitous kinase
B08 Mm.392384 NM_001025432 Crebbp CREB binding protein
B09 Mm.795 NM_007778 Csf1 Colony stimulating factor 1 (macrophage)
B10 Mm.4922 NM_009969 Csf2 Colony stimulating factor 2 (granulocyte-macrophage)
B11 Mm.1238 NM_009971 Csf3 Colony stimulating factor 3 (granulocyte)
B12 Mm.420648 NM_007912 Egfr Epidermal growth factor receptor
C01 Mm.181959 NM_007913 Egr1 Early growth response 1
C02 Mm.378990 NM_011163 Eif2ak2 Eukaryotic translation initiation factor 2-alpha kinase 2
C03 Mm.490895 NM_007922 Elk1 ELK1, member of ETS oncogene family
C04 Mm.24816 NM_010169 F2r Coagulation factor II (thrombin) receptor
C05 Mm.5126 NM_010175 Fadd Fas (TNFRSF6)-associated via death domain
C06 Mm.3355 NM_010177 Fasl Fas ligand (TNF superfamily, member 6)
C07 Mm.246513 NM_010234 Fos FBJ osteosarcoma oncogene
C08 Mm.276389 NM_010442 Hmox1 Heme oxygenase (decycling) 1
C09 Mm.435508 NM_010493 Icam1 Intercellular adhesion molecule 1
C10 Mm.240327 NM_008337 Ifng Interferon gamma
C11 Mm.277886 NM_010546 Ikbkb Inhibitor of kappaB kinase beta
C12 Mm.386783 NM_019777 Ikbke Inhibitor of kappaB kinase epsilon
D01 Mm.12967 NM_010547 Ikbkg Inhibitor of kappaB kinase gamma
D02 Mm.874 NM_010548 Il10 Interleukin 10
D03 Mm.15534 NM_010554 Il1a Interleukin 1 alpha
D04 Mm.222830 NM_008361 Il1b Interleukin 1 beta
D05 Mm.896 NM_008362 Il1r1 Interleukin 1 receptor, type I
D06 Mm.38241 NM_008363 Irak1 Interleukin-1 receptor-associated kinase 1
D07 Mm.152142 NM_172161 Irak2 Interleukin-1 receptor-associated kinase 2
D08 Mm.105218 NM_008390 Irf1 Interferon regulatory factor 1
D09 Mm.275071 NM_010591 Jun Jun oncogene
D10 Mm.87787 NM_010735 Lta Lymphotoxin A
D11 Mm.3122 NM_010736 Ltbr Lymphotoxin B receptor
D12 Mm.15918 NM_011945 Map3k1 Mitogen-activated protein kinase kinase kinase 1
E01 Mm.8385 NM_011952 Mapk3 Mitogen-activated protein kinase 3
E02 Mm.213003 NM_010851 Myd88 Myeloid differentiation primary response gene 88
E03 Mm.256765 NM_008689 Nfkb1 Nuclear factor of kappa light polypeptide gene enhancer in B-cells 1, p105
E04 Mm.102365 NM_019408 Nfkb2 Nuclear factor of kappa light polypeptide gene enhancer in B-cells 2, p49/p100
E05 Mm.170515 NM_010907 Nfkbia Nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha
E06 Mm.28498 NM_172729 Nod1 Nucleotide-binding oligomerization domain containing 1
E07 Mm.184163 NM_029780 Raf1 V-raf-leukemia viral oncogene 1
E08 Mm.4869 NM_009044 Rel Reticuloendotheliosis oncogene
E09 Mm.249966 NM_009045 Rela V-rel reticuloendotheliosis viral oncogene homolog A (avian)
E10 Mm.1741 NM_009046 Relb Avian reticuloendotheliosis viral (v-rel) oncogene related B
E11 Mm.374799 NM_009068 Ripk1 Receptor (TNFRSF)-interacting serine-threonine kinase 1
E12 Mm.112765 NM_138952 Ripk2 Receptor (TNFRSF)-interacting serine-threonine kinase 2
F01 Mm.272675 NM_015747 Slc20a1 Solute carrier family 20, member 1
F02 Mm.7320 NM_016769 Smad3 MAD homolog 3 (Drosophila)
F03 Mm.487336 NM_009283 Stat1 Signal transducer and activator of transcription 1
F04 Mm.34580 NM_019786 Tbk1 TANK-binding kinase 1
F05 Mm.273024 NM_030682 Tlr1 Toll-like receptor 1
F06 Mm.87596 NM_011905 Tlr2 Toll-like receptor 2
F07 Mm.33874 NM_126166 Tlr3 Toll-like receptor 3
F08 Mm.38049 NM_021297 Tlr4 Toll-like receptor 4
F09 Mm.42146 NM_011604 Tlr6 Toll-like receptor 6
F10 Mm.44889 NM_031178 Tlr9 Toll-like receptor 9
F11 Mm.1293 NM_013693 Tnf Tumor necrosis factor
F12 Mm.116683 NM_009397 Tnfaip3 Tumor necrosis factor, alpha-induced protein 3
G01 Mm.193430 NM_020275 Tnfrsf10b Tumor necrosis factor receptor superfamily, member 10b
G02 Mm.474976 NM_011609 Tnfrsf1a Tumor necrosis factor receptor superfamily, member 1a
G03 Mm.235328 NM_011610 Tnfrsf1b Tumor necrosis factor receptor superfamily, member 1b
G04 Mm.1062 NM_009425 Tnfsf10 Tumor necrosis factor (ligand) superfamily, member 10
G05 Mm.483369 NM_019418 Tnfsf14 Tumor necrosis factor (ligand) superfamily, member 14
G06 Mm.103551 NM_023764 Tollip Toll interacting protein
G07 Mm.264255 NM_001033161 Tradd TNFRSF1A-associated via death domain
G08 Mm.3399 NM_009422 Traf2 Tnf receptor-associated factor 2
G09 Mm.27431 NM_011632 Traf3 Tnf receptor-associated factor 3
G10 Mm.389227 NM_011633 Traf5 Tnf receptor-associated factor 5
G11 Mm.292729 NM_009424 Traf6 Tnf receptor-associated factor 6
G12 Mm.8038 NM_009539 Zap70 Zeta-chain (TCR) associated protein kinase
H01 Mm.391967 NM_007393 Actb Actin, beta
H02 Mm.163 NM_009735 B2m Beta-2 microglobulin
H03 Mm.304088 NM_008084 Gapdh Glyceraldehyde-3-phosphate dehydrogenase
H04 Mm.3317 NM_010368 Gusb Glucuronidase, beta
H05 Mm.2180 NM_008302 Hsp90ab1 Heat shock protein 90 alpha (cytosolic), class B member 1
H06 N/A SA_00106 MGDC Mouse Genomic DNA Contamination
H07 N/A SA_00104 RTC Reverse Transcription Control
H08 N/A SA_00104 RTC Reverse Transcription Control
H09 N/A SA_00104 RTC Reverse Transcription Control
H10 N/A SA_00103 PPC Positive PCR Control
H11 N/A SA_00103 PPC Positive PCR Control
H12 N/A SA_00103 PPC Positive PCR Control

Figure 4.

Figure 4

Figure 4

Figure 4

Figure 4A. NFκB Signaling Pathway RT2 Profiler PCR Array of DADS vs Control. Effect of DADS vs control in MDA-MB-231 cells, displayed on a volcano plot showing significance, fold change and direction. There were no significant differences found between these groups at p<0.05.

Figure 4B. NFκB Signaling Pathway RT2 Profiler PCR Array of TNFα vs control. Analysis of TNFα vs control displayed on a volcano plot showing significance, fold change and direction. Transcriptome upward directional shifts (A), with significance and Log2 (Fold Change) listed along official gene symbols (B).

Figure 4C. NFκB Signaling Pathway RT2 Profiler PCR Array of TNFα vs TNFα/DADS. Effect of TNFα vs TNFα/DADS on gene expression: displayed on a volcano plot showing significance, fold change and direction. There were significant differences found between these groups using the PCR Array PAMM – 025Z in the downward direction (A), with significance and Log2 (Fold Change) listed along official gene symbols (B).

Table 2.

Function Categories Affected by TNF-alpha vs Control

Effect of TNFα vs Controls on targeted pathways using DAVID functional annotation statistical program. Gene-annotation enrichment analysis, functional annotation clustering on statistically differentially expressed genes. Statistical DAVID genes were analyzed for annotation cluster enrichment scores. The data represent the score, number of genes, shift direction and p-values (Fisher Exact/EASE Score).

Annotation Cluster 1 Enrichment Score: 7.62 Count P_Value Benjamini
GOTERM_BP_FAT regulation of cytokine biosynthetic process RT 6 2.50E-10 1.40E-08
Annotation Cluster 2 Enrichment Score: 7.02 Count P_Value Benjamini
GOTERM_BP_FAT positive regulation of protein kinase cascade RT 7 5.10E-12 8.5E-10
GOTERM_BP_FAT regulation of interleukin-6 production RT 6 6.5E-12 8.10E-10
GOTERM_BP_FAT positive regulation of I-kappaB kinase/NF-kappaB cascade RT 6 7.7E-12 7.70E-10
GOTERM_BP_FAT regulation of stress-activated protein kinase signaling pathway RT 5 1.40E-08 4.20E-07
Annotation Cluster 3 Enrichment Score: 4.08 Count P_Value Benjamini
GOTERM_BP_FAT regulation of chemokine product RT 4 3.30E-08 8.60E-07

Figure 5. Effect of DADs on TNFα NF-Kappa B expression pattern using RT-PCR.

Figure 5

The data represent the Mean ± S.E.M. n=3. There were no statistical differences found between the Control and TNFα controls ± DADS for NF-Kappa B1 (A) or NF-KappaB2 (B), also corroborating PCR expression arrays for both subtypes (C).

DISCUSSION

CCL2 is a cancer promoting chemokine with capacity to enhance malignant cell migration, proliferation and invasive properties [20]. This enhancement occurs by mobilization of monocytes, macrophages and other inflammatory components to infiltrate the tumor area [21] enabling differentiation into TAMs [22]. The potent effects of CCL2 on metastatic invasion are likely due to it association with the elevation of matrix metalloproteinases (MMPs) (e.g. MMP-1, MMP-9) [23], prooncogenic substances such as TNF-α, vascular endothelial growth factor (VEGF)-A, TGFβ1 and IL-8 which collectively assist differentiation of human monocytes into TAMS. In a self-perpetuating cycle, CCL2 release is indirectly controlled by TNFα via TNF-receptors (TNFRs), with both CCL2 and TNFα being highly co-expressed in many human cancers [9,24]. Overactive TNFα receptor signaling is associated with coordination of tumor angiogenesis and metastasis [5] necessitating the development of therapeutic anti-cancer drugs. These drugs were designed to sequester TNFα [25] or block TNFRs [26 27] such as infliximab (Remicade), adalimumab (Humira), all of which downregulate TAMS [10] or other tumor infiltrating leucocytes in the tumor microenvironment [12]. Likewise, drugs or agents that can suppress CCL2-CCR2 signaling, can block monocyte recruitment and inhibit metastasis in vivo [28].

DADS, one of the major organo-sulfur compounds in garlic, is becoming recognized as a potential cancer chemopreventive compound. DADS is effective against growth of diverse cancer cell types such as HT-29 [24] HL-60 [30] cultured human colon tumor cells (HCT-15) skin (SK MEL-2) and lung (A549) [31]. Preliminary studies in our lab have indicated that DADS can attenuate CCL2 release in TNFα stimulated human breast carcinoma cells. DADS has recently been shown to reduce migration and invasion of human colon cancer in part mediated by NF-κB, ERK1/2, JNK1/2 and p38 signaling. [32] In this study, we explore signaling involved with DADS ability to down-regulate CCL2 release in TNFα-stimulated MDA-MB-231 cells.

In tumor cells, elevated NFκB signaling is triggered by TNFα, corresponding to a rise in CCL2 and TAM recruitment, cell proliferation, invasiveness and angiogenesis.[68]. TNF-α activation of NFκB requires its translocation from the cytoplasm to the nucleus to function. The location of NFκB is controlled by IκBs, which binds NFκB and prevents nuclear uptake. Further downstream, IκBs are themselves regulated by phosphorylation which can trigger ubiquitin-dependent degradation. The phosphorylation of IκB by IκB kinase (IKK) occurs on IKKbeta, itself a component of IKK complexes housing regulatory subunits IKKα, IKKγ and NEMO. [33]. Phosphorylation enables the recognition by E3RS (IκB/β-TrCP) to E3 ubiquitin ligase, leading to degradation, and thereby breaking controlling elements for IκB, enabling rapid NFκB translocation to the nucleus to turn on proinflammatory molecules [34]. The data in this study suggest that TNFα initiated a rise in NFκB1/2 gene expression (confirmed by PCR Array PAMM – 025Z and RT-PCR), both sustained in the presence or absence of DADS. However, DADs reduced protein expression of IKKε, which could negatively control NFκB activation signaling, and account for loss of CCL2 protein expression.

IKKi/IKKε plays an important role in carrying out TNFα signaling, via acting as a serine-threonine kinase [33]. It is capable of phosphorylating NFκB subunit RelA (also known as p65) correlating to NFκB activation [35], a rise in CCAAA/enhancer-binding protein (C/EBPδ) [36] and phosphorylation/rapid degradation of inhibitors of NFκB. Subsequent dissociation of the inhibitor/NFκB complex allows free NFκB translocation to the nucleus and initiates gene transcription. The ability of DADS to downregulate IKKε could in effect hamper TNFα induced IKKε-mediated NFκB activation [37]. This is an otherwise strong correlate to many human cancers, including, breast, ovarian, prostate, glial, [38, 39], esophageal, [40] and aggressive metastasis, tumor survival, [41] and poor clinical prognosis in diverse cancers [42]. Further, the correlation of IKKε with cell proliferation and transformation, has given rise to its being classified as oncogene [43]. Silencing or inhibition of IKKε results in inhibition of cell growth, proliferation, invasion, [44] clonogenicity, migration [45] and overcoming its contributory resistance to tamoxifen [46] in breast cancer, as well as cisplatin in ovarian tumors.[42] The identification of novel molecules that can inhibit IKKε is currently underway as a means to inflammatory processes associated with cancer progression.[47] Moreover, if DADS can reduce IKKε, this could also prevent events downstream to IKKε over expression such as activation of p52 NF-κB dimers [48], [49] estrogen receptor ERα activation, upregulation of cyclin D1 and chemotherapy resistance in breast cancer cells in particular to tamoxifen [50]. In the current study we were focused on IKKi/IKKε because it plays an important role in TNFα signaling. The presented data show a correlation between IKKε and cell proliferation and transformation as well as many different cancers. The data also show the involvement of IKKi/IKKε in tumor survival and aggressive metastasis. We are reporting that IKKε expression is reduced in this model, which is not isolated to this model but is important since this model has been known to be highly aggressive and has fewer treatment options. In the study we have not examined signaling molecules in other TNBC cell lines but we are planning to do so in future studies.

The data presented in this study suggest DADS can down-regulate IKK and CCL2 but the mechanism for this is unclear. It is possible that DADS could be down regulating the TNFα receptor complex, which would correlate to subcellular localization of NFκB, and its influence on induction of CCL2. The effect of DADS in this study, did not appear to involve mRNA of NFκB, but possibly reduced TNFRSF1A gene and adaptor protein tumor necrosis factor receptor (TNFR-associated death domain, TRADD), which are well known to “activate” via altering subcellular localization of NFκB. [5153] Future research will be required to evaluate if the effects of DADS on CCL2 occur due to upstream events including TNFR down-regulation or potential involvement of AKT, which directly leads to up regulation of IKKε protein expression in MDA-MB-231 cells [54].

Down stream TNFα-triggered multiple signaling pathways that lead to expressed and secreted RANTES and CCL2 [55]. These pathways are believed to involve TNFR1 association with Jak2, c-Src, which could lead to CCL2 release through one or more of activating p38 MAPK, JNK, and Akt or activation of NFκB [56]. The data from this study show no change in the total proteins for ERK, P38, MEK and JNK, in the presence or absence of TNFα and DADS, however TNFα induction of CCL2 is mediated through signaling which involves MAPK phosphorylation signaling, which were further reduced in the presence of DADS. These findings suggest that level of control of DADS in CCL2 reduction is either or both occurring at the TNFα receptor or phosphorylation of ERK and P38, the former being confirmed by the data.

In summary, the findings of this study contribute to the body of work describing garlic as a chemopreventive agent, carrying diverse properties which range from carcinogen detoxification to cell-cycle arrest/apoptosis and a reduced expression of monocyte-chemoattractant protein [57]. DADS or any compound that can suppress TAM recruitment is considered an effective therapeutic approach in treatment of human cancers [5859].

Highlights.

  • DADS/TNFα decreased CCL2, Casp8, and Tradd gene expression in a TNBC cell line

  • DADS/TNFα reduced IKKε protein expression in a TNBC cell line.

  • DADS/TNFα reduced phosphorylated ERK protein expression in a TNBC cell line.

Acknowledgments

This project was supported by the National Center for Research Resources NIH NCRR RCMI program (G12RR 03020) and the National Institute on Minority Health and Health Disparities, NIH (8G12MD007582-28 and 1P20 MD006738-01)

Footnotes

This manuscript version is made available under the CC BY-NC-ND 4.0 license.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Zlotnik A. Chemokines in neoplastic progression. Seminars in cancer biology. 2004;14:181–5. doi: 10.1016/j.semcancer.2003.10.004. [DOI] [PubMed] [Google Scholar]
  • 2.Tanaka T, Bai Z, Srinoulprasert Y, Yang BG, Hayasaka H, Miyasaka M. Chemokines in tumor progression and metastasis. Cancer science. 2005;96:317–22. doi: 10.1111/j.1349-7006.2005.00059.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Rossi D, Zlotnik A. The biology of chemokines and their receptors. Annual review of immunology. 2000;18:217–42. doi: 10.1146/annurev.immunol.18.1.217. [DOI] [PubMed] [Google Scholar]
  • 4.Jaattela M. Biologic activities and mechanisms of action of tumor necrosis factor-alpha/cachectin. Laboratory investigation; a journal of technical methods and pathology. 1991;64:724–42. [PubMed] [Google Scholar]
  • 5.Balkwill F. TNF-alpha in promotion and progression of cancer. Cancer metastasis reviews. 2006;25:409–16. doi: 10.1007/s10555-006-9005-3. [DOI] [PubMed] [Google Scholar]
  • 6.Kehlen A, Haegele M, Menge K, Gans K, Immel UD, Hoang-Vu C, et al. Role of glutaminyl cyclases in thyroid carcinomas. Endocrine-related cancer. 2013;20:79–90. doi: 10.1530/ERC-12-0053. [DOI] [PubMed] [Google Scholar]
  • 7.Hopewell EL, Zhao W, Fulp WJ, Bronk CC, Lopez AS, Massengill M, et al. Lung tumor NF-kappaB signaling promotes T cell-mediated immune surveillance. The Journal of clinical investigation. 2013;123:2509–22. doi: 10.1172/JCI67250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Lerebours F, Vacher S, Andrieu C, Espie M, Marty M, Lidereau R, et al. NF-kappa B genes have a major role in inflammatory breast cancer. BMC cancer. 2008;8:41. doi: 10.1186/1471-2407-8-41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Dobrzycka B, Terlikowski SJ, Garbowicz M, Niklinska W, Bernaczyk PS, Niklinski J, et al. Tumor necrosis factor-alpha and its receptors in epithelial ovarian cancer. Folia histochemica et cytobiologica/Polish Academy of Sciences, Polish Histochemical and Cytochemical Society. 2009;47:609–13. doi: 10.2478/v10042-008-0117-1. [DOI] [PubMed] [Google Scholar]
  • 10.Sethi G, Sung B, Kunnumakkara AB, Aggarwal BB. Targeting TNF for Treatment of Cancer and Autoimmunity. Advances in experimental medicine and biology. 2009;647:37–51. doi: 10.1007/978-0-387-89520-8_3. [DOI] [PubMed] [Google Scholar]
  • 11.Zhou P, Qiu J, L’Italien L, Gu D, Hodges D, Chao CC, et al. Mature B cells are critical to T-cell-mediated tumor immunity induced by an agonist anti-GITR monoclonal antibody. J Immunother. 2010;33:789–97. doi: 10.1097/CJI.0b013e3181ee6ba9. [DOI] [PubMed] [Google Scholar]
  • 12.Schaer DA, Cohen AD, Wolchok JD. Anti-GITR antibodies--potential clinical applications for tumor immunotherapy. Curr Opin Investig Drugs. 2010;11:1378–86. [PubMed] [Google Scholar]
  • 13.Nakagawa H, Tsuta K, Kiuchi K, Senzaki H, Tanaka K, Hioki K, Tsubura A. Growth inhibitory effects of diallyl disulfide on human breast cancer cell lines. Carcinogenesis. 2001 Jun;22(6):891–7. doi: 10.1093/carcin/22.6.891. [DOI] [PubMed] [Google Scholar]
  • 14.Fang WB, Jokar I, Zou A, Lambert D, Dendukuri P, Cheng N. CCL2/CCR2 Chemokine signaling coordinates survival and motility of breast cancer cells through smad3 protein and p42/44 mitogen activated protein kinase (MAPK)-dependent mechanisms. The Journal of Biological Chemistry. 2012 Oct;287(43):36593–36608. doi: 10.1074/jbc.M112.365999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Gordon AH, O’Keefe RJ, Schwartz EM, Rosier RN, Puzas JE. Nuclear Factor κB-dependent mechanisms in breast cancer cells regulate tumor burden and osteolysis in bone. Cancer Res. 2005;65:3209–3217. doi: 10.1158/0008-5472.CAN-04-4017. [DOI] [PubMed] [Google Scholar]
  • 16.Bauer D, Mazzio E, Soliman KF, Taka E, Oriaku E, Womble T. Darling-Reed S. Diallyl disulfide inhibits TNFα-induced CCL2 release by MDA-MB-231 cells. Anticancer Res. 2014 Jun;34(6):2763–70. [PMC free article] [PubMed] [Google Scholar]
  • 17.Nkrumah-Elie YM1, Reuben JS, Hudson AM, Taka E, Badisa R, Ardley T, Israel B, Sadrud-Din SY, Oriaku ET, Darling-Reed SF. The attenuation of early benzo(a)pyrene-induced carcinogenic insults by diallyl disulfide (DADS) in MCF-10A cells. Nutr Cancer. 2012;64(7):1112–21. doi: 10.1080/01635581.2012.712738. Epub 2012 Sep 24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Huang da W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nature protocols. 2009;4:44–57. doi: 10.1038/nprot.2008.211. [DOI] [PubMed] [Google Scholar]
  • 19.Huang da W, Sherman BT, Tan Q, Kir J, Liu D, Bryant D, et al. DAVID Bioinformatics Resources: expanded annotation database and novel algorithms to better extract biology from large gene lists. Nucleic Acids Res. 2007;35:W169–75. doi: 10.1093/nar/gkm415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Craddock JA, Lu A, Bear A, Pule M, Brenner MK, Rooney CM, et al. Enhanced tumor trafficking of GD2 chimeric antigen receptor T cells by expression of the chemokine receptor CCR2b. J Immunother. 2010;33:780–8. doi: 10.1097/CJI.0b013e3181ee6675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Perry JA, Thamm DH, Eickhoff J, Avery AC, Dow SW. Increased monocyte chemotactic protein-1 concentration and monocyte count independently associate with a poor prognosis in dogs with lymphoma. Veterinary and comparative oncology. 2011;9:55–64. doi: 10.1111/j.1476-5829.2010.00235.x. [DOI] [PubMed] [Google Scholar]
  • 22.Mishra P, Banerjee D, Ben-Baruch A. Chemokines at the crossroads of tumor-fibroblast interactions that promote malignancy. Journal of leukocyte biology. 2011;89:31–9. doi: 10.1189/jlb.0310182. [DOI] [PubMed] [Google Scholar]
  • 23.Hatfield KJ, Reikvam H, Bruserud O. The crosstalk between the matrix metalloprotease system and the chemokine network in acute myeloid leukemia. Current medicinal chemistry. 2010;17:4448–61. doi: 10.2174/092986710794183033. [DOI] [PubMed] [Google Scholar]
  • 24.Cendan JC, Topping DL, Pruitt J, Snowdy S, Copeland EM, 3rd, Lind DS. Inflammatory mediators stimulate arginine transport and arginine-derived nitric oxide production in a murine breast cancer cell line. The Journal of surgical research. 1996;60:284–8. doi: 10.1006/jsre.1996.0044. [DOI] [PubMed] [Google Scholar]
  • 25.Moran AE, Kovacsovics-Bankowski M, Weinberg AD. The TNFRs OX40, 4-1BB, and CD40 as targets for cancer immunotherapy. Current opinion in immunology. 2013;25:230–7. doi: 10.1016/j.coi.2013.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Daniel D, Wilson NS. Tumor necrosis factor: renaissance as a cancer therapeutic? Current cancer drug targets. 2008;8:124–31. doi: 10.2174/156800908783769346. [DOI] [PubMed] [Google Scholar]
  • 27.Mahmood Z, Shukla Y. Death receptors: targets for cancer therapy. Experimental cell research. 2010;316:887–99. doi: 10.1016/j.yexcr.2009.12.011. [DOI] [PubMed] [Google Scholar]
  • 28.Qian BZ, Li J, Zhang H, Kitamura T, Zhang J, Campion LR, et al. CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis. Nature. 2011;475:222–5. doi: 10.1038/nature10138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Huang YS, Xie N, Su Q, Su J, Huang C, Liao QJ. Diallyl disulfide inhibits the proliferation of HT-29 human colon cancer cells by inducing differentially expressed genes. Molecular medicine reports. 2011;4:553–9. doi: 10.3892/mmr.2011.453. [DOI] [PubMed] [Google Scholar]
  • 30.Yi L, Ji XX, Lin M, Tan H, Tang Y, Wen L, et al. Diallyl disulfide induces apoptosis in human leukemia HL-60 cells through activation of JNK mediated by reactive oxygen. Die Pharmazie. 2010;65:693–8. [PubMed] [Google Scholar]
  • 31.Sundaram SG, Milner JA. Diallyl disulfide induces apoptosis of human colon tumor cells. Carcinogenesis. 1996;17:669–73. doi: 10.1093/carcin/17.4.669. [DOI] [PubMed] [Google Scholar]
  • 32.Lai KC, Hsu SC, Kuo CL, Yang JS, Ma CY, Lu HF, et al. Diallyl sulfide, diallyl disulfide, and diallyl trisulfide inhibit migration and invasion in human colon cancer colo 205 cells through the inhibition of matrix metalloproteinase-2, -7, and -9 expressions. Environmental toxicology. 2011 doi: 10.1002/tox.20737. [DOI] [PubMed] [Google Scholar]
  • 33.Hacker H, Karin M. Regulation and function of IKK and IKK-related kinases. Science’s STKE: signal transduction knowledge environment. 2006;2006:re13. doi: 10.1126/stke.3572006re13. [DOI] [PubMed] [Google Scholar]
  • 34.Karin M, Ben-Neriah Y. Phosphorylation meets ubiquitination: the control of NF-[kappa]B activity. Annual review of immunology. 2000;18:621–63. doi: 10.1146/annurev.immunol.18.1.621. [DOI] [PubMed] [Google Scholar]
  • 35.Wang Y, Lu X, Zhu L, Shen Y, Chengedza S, Feng H, et al. IKK epsilon kinase is crucial for viral G protein-coupled receptor tumorigenesis. Proceedings of the National Academy of Sciences of the United States of America. 2013;110:11139–44. doi: 10.1073/pnas.1219829110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Wang N, Ahmed S, Haqqi TM. Genomic structure and functional characterization of the promoter region of human IkappaB kinase-related kinase IKKi/IKKvarepsilon gene. Gene. 2005;353:118–33. doi: 10.1016/j.gene.2005.04.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Zhou AY, Shen RR, Kim E, Lock YJ, Xu M, Chen ZJ, et al. IKKepsilon-mediated tumorigenesis requires K63-linked polyubiquitination by a cIAP1/cIAP2/TRAF2 E3 ubiquitin ligase complex. Cell reports. 2013;3:724–33. doi: 10.1016/j.celrep.2013.01.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Peant B, Forest V, Trudeau V, Latour M, Mes-Masson AM, Saad F. IkappaB-Kinase-epsilon (IKKepsilon/IKKi/IkappaBKepsilon) expression and localization in prostate cancer tissues. The Prostate. 2011;71:1131–8. doi: 10.1002/pros.21329. [DOI] [PubMed] [Google Scholar]
  • 39.Cheng A, Guo J, Henderson-Jackson E, Kim D, Malafa M, Coppola D. IkappaB Kinase epsilon expression in pancreatic ductal adenocarcinoma. American journal of clinical pathology. 2011;136:60–6. doi: 10.1309/AJCP2JJGYNIUAS2V. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Kang MR, Kim MS, Kim SS, Ahn CH, Yoo NJ, Lee SH. NF-kappaB signalling proteins p50/p105, p52/p100, RelA, and IKKepsilon are over-expressed in oesophageal squamous cell carcinomas. Pathology. 2009;41:622–5. doi: 10.3109/00313020903257756. [DOI] [PubMed] [Google Scholar]
  • 41.Baldwin AS. Regulation of cell death and autophagy by IKK and NF-kappaB: critical mechanisms in immune function and cancer. Immunological reviews. 2012;246:327–45. doi: 10.1111/j.1600-065X.2012.01095.x. [DOI] [PubMed] [Google Scholar]
  • 42.Guo JP, Shu SK, He L, Lee YC, Kruk PA, Grenman S, et al. Deregulation of IKBKE is associated with tumor progression, poor prognosis, and cisplatin resistance in ovarian cancer. The American journal of pathology. 2009;175:324–33. doi: 10.2353/ajpath.2009.080767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Verhelst K, Verstrepen L, Carpentier I, Beyaert R. IkappaB kinase epsilon (IKKepsilon): a therapeutic target in inflammation and cancer. Biochemical pharmacology. 2013;85:873–80. doi: 10.1016/j.bcp.2013.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Li H, Chen L, Zhang A, Wang G, Han L, Yu K, et al. Silencing of IKKepsilon using siRNA inhibits proliferation and invasion of glioma cells in vitro and in vivo. International journal of oncology. 2012;41:169–78. doi: 10.3892/ijo.2012.1452. [DOI] [PubMed] [Google Scholar]
  • 45.Qin B, Cheng K. Silencing of the IKKepsilon gene by siRNA inhibits invasiveness and growth of breast cancer cells. Breast cancer research: BCR. 2010;12:R74. doi: 10.1186/bcr2644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Grandvaux N. The IKKepsilon kinase in breast cancer: from oncogenesis to treatment resistance. Medecine sciences: M/S. 2011;27:619–25. doi: 10.1051/medsci/2011276014. [DOI] [PubMed] [Google Scholar]
  • 47.Hutti JE, Porter MA, Cheely AW, Cantley LC, Wang X, Kireev D, et al. Development of a high-throughput assay for identifying inhibitors of TBK1 and IKKepsilon. PloS one. 2012;7:e41494. doi: 10.1371/journal.pone.0041494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Wietek C, Cleaver CS, Ludbrook V, Wilde J, White J, Bell DJ, et al. IkappaB kinase epsilon interacts with p52 and promotes transactivation via p65. The Journal of biological chemistry. 2006;281:34973–81. doi: 10.1074/jbc.M607018200. [DOI] [PubMed] [Google Scholar]
  • 49.Li Q, Sun H, Zou J, Ge C, Yu K, Cao Y, et al. Increased Expression of Estrogen Receptor alpha-36 by Breast Cancer Oncogene IKKepsilon Promotes Growth of ER-Negative Breast Cancer Cells. Cellular physiology and biochemistry: international journal of experimental cellular physiology, biochemistry, and pharmacology. 2013;31:833–41. doi: 10.1159/000350101. [DOI] [PubMed] [Google Scholar]
  • 50.Guo JP, Shu SK, Esposito NN, Coppola D, Koomen JM, Cheng JQ. IKKepsilon phosphorylation of estrogen receptor alpha Ser-167 and contribution to tamoxifen resistance in breast cancer. The Journal of biological chemistry. 2010;285:3676–84. doi: 10.1074/jbc.M109.078212. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 51.Guan YJ, Zhang Z, Yu C, Ma L, Hu W, Xu L, et al. Phospho-SXXE/D motif mediated TNF receptor 1-TRADD death domain complex formation for T cell activation and migration. J Immunol. 2011;187:1289–97. doi: 10.4049/jimmunol.1003399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Zheng L, Bidere N, Staudt D, Cubre A, Orenstein J, Chan FK, et al. Competitive control of independent programs of tumor necrosis factor receptor-induced cell death by TRADD and RIP1. Molecular and cellular biology. 2006;26:3505–13. doi: 10.1128/MCB.26.9.3505-3513.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Pobezinskaya YL, Liu Z. The role of TRADD in death receptor signaling. Cell Cycle. 2012;11:871–6. doi: 10.4161/cc.11.5.19300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Krishnamurthy S, Basu A. Regulation of IKKepsilon Expression by Akt2 Isoform. Genes & cancer. 2011;2:1044–50. doi: 10.1177/1947601912444604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Westlund KN, Zhang L, Ma F, Oz HS. Chronic inflammation and pain in a tumor necrosis factor receptor (TNFR) (p55/p75−/−) dual deficient murine model. Translational research: the journal of laboratory and clinical medicine. 2012;160:84–94. doi: 10.1016/j.trsl.2011.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Pincheira R, Castro AF, Ozes ON, Idumalla PS, Donner DB. Type 1 TNF receptor forms a complex with and uses Jak2 and c-Src to selectively engage signaling pathways that regulate transcription factor activity. J Immunol. 2008;181:1288–98. doi: 10.4049/jimmunol.181.2.1288. [DOI] [PubMed] [Google Scholar]
  • 57.Tsubura A, Lai YC, Kuwata M, Uehara N, Yoshizawa K. Anticancer effects of garlic and garlic-derived compounds for breast cancer control. Anti-cancer agents in medicinal chemistry. 2011;11:249–53. doi: 10.2174/187152011795347441. [DOI] [PubMed] [Google Scholar]
  • 58.Zhang J, Patel L, Pienta KJ. Targeting chemokine (C-C motif) ligand 2 (CCL2) as an example of translation of cancer molecular biology to the clinic. Progress in molecular biology and translational science. 2010;95:31–53. doi: 10.1016/B978-0-12-385071-3.00003-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Rafei M, Galipeau J. A CCL2-based fusokine as a novel biopharmaceutical for the treatment of CCR2-driven autoimmune diseases. Critical reviews in immunology. 2010;30:449–61. doi: 10.1615/critrevimmunol.v30.i5.40. [DOI] [PubMed] [Google Scholar]

RESOURCES