Skip to main content

Main menu

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Subscribers
    • Advertisers
    • Editorial Board
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics

User menu

  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Anticancer Research
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics
  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart
Anticancer Research

Advanced Search

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Subscribers
    • Advertisers
    • Editorial Board
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Visit us on Facebook
  • Follow us on Linkedin
Research ArticleExperimental Studies

Diallyl Disulfide Inhibits TNFα-induced CCL2 Release by MDA-MB-231 Cells

DAVID BAUER, ELIZABETH MAZZIO, KARAM FA SOLIMAN, EQUAR TAKA, EBENEZER ORIAKU, TRACEY WOMBLE and SELINA DARLING-REED
Anticancer Research June 2014, 34 (6) 2763-2770;
DAVID BAUER
College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
ELIZABETH MAZZIO
College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
KARAM FA SOLIMAN
College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
EQUAR TAKA
College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
EBENEZER ORIAKU
College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
TRACEY WOMBLE
College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
SELINA DARLING-REED
College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: selina.darling@famu.edu
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Monocyte chemotactic protein-1 (MCP-1/CCL2) is released by tumor tissues, serving as a potent chemokine enabling directional homing of mononuclear cells to tumor tissue, which subsequently differentiate into tumor-associated macrophages (TAMs) via TGFβ1 signaling. TAMs readily invade tumor tissue and continue to synthesize pro-oncogenic proteins including tumor growth factors, matrix proteases (metastasis), angiogenic factors (neovascularization) and CCL2. Substances, which can attenuate or block the initial release of CCL2 have been shown to prevent cancer-associated inflammative pro-oncogenic processes. In the current study, we investigated the effects of the organosulfur compound diallyl disulfide (DADS), a natural constituent of Allium sativum (garlic) on suppression of TNFα-induced release of CCL2 from triple-negative human breast tumor (MDA-MB-231) cells. Using an initial adipokine/chemokine protein panel microarray, the data show a predominant expression profile in resting/untreated MDA-MB-231 cells for sustained release of IL6, IL8, plasminogen Activator Inhibitor 1 and TIMP1/2. Treatment with TNFα (40 ng/ml) had no effect on many of these molecules, with a single major elevation in release of CCL2 (~1,300-fold up-regulation). TNFα-induced CCL2 release was reversed by a sub-lethal concentration of DADS (100 μM), evident in antibody based assays. These findings provide evidence to support another avenue of anticancer/chemopreventative properties attributable to garlic constituents through immunomodulation.

  • Tumor-associated macrophages
  • monocyte chemotactic protein-1
  • garlic constituents
  • diallyl disulfide

Cancer-associated inflammation involves complex collective events within the immune system which enable growth and metastasis of diverse cancers l. In brief, human tumors of almost every type including gliomas, melanoma (1) lung (2) renal (3) prostate (4) and breast (5) produce and release high concentrations of CCL2, a most prolific tumor-promoting chemokine which attracts monocytes to the tumor area (3, 6) via monocyte G-coupled CCL2 receptors such as CCR2A/2B (7). Once monocytes arrive at the tumor site, transforming growth factor beta-1 (TGFβ1) and interleukin-8 assist with advanced differentiation whereby these cells acquire traits beneficial to tumor cells, with a phenotypic change leading them to be recognized as tumor-associated macrophages (TAMs) (8, 9). TAMs then embed within the tumor, and increase tumor growth by fostering production and release of tumor growth factors (tumor growth), matrix proteases (invasion), angiogenic factors (neovascularization) and mechanistic blocking of tumor reactive T-cells/reducing (immune evasion) (10-12).

Therapeutic targeting of either the monocyte CCR2 receptor or release of CCL2 constitutes a dynamic means of blocking recruitment and mobilization of infiltrating monocytes to the tumor site (13, 14). A number of studies have demonstrated efficacy of monoclonal antibody to CCL2 IgG1κ (carlumab) or broccoli-derived compounds (i.e. indole-3-carbinol and 3,3’-diindolylmethane) against deplete monocyte infiltration and thereby also reduce tumor growth and metastasis (14-16). In the current study, we investigated the effects of a primary organosulfur compound diallyl disulfide (DADS) constituent of Allium sativum (garlic) on suppression of TNFα-induced release of CCL2 from triple-negative human breast tumor (MDA-MB-231) cells.

Materials and Methods

Cell line, chemicals and reagents. Triple-negative human breast tumor (MDA-MB-231) cells were obtained from the American Type Culture Collection (Rockville, MD, USA). Dulbecco's modified Eagle's medium (DMEM), fetal bovine serum (FBS) and penicillin/streptomycin were all obtained from Invitrogen (Carlsbad, CA, USA). Recombinant human TNFα was purchased from RayBiotech (RayBiotech Inc., Norcross, GA, USA). DADS (>80% purity) was purchased from Sigma-Aldrich (St. Louis, MO, USA).

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

The effect of DADS on cell viability of MDA-MB-231 cells at 5% CO2/Atm for 24 hr. The data are presented as mean±S.E.M. (n=4). Significance of differences from the control were determined by a one-way ANOVA, with a Tukey post hoc test. *p<0.05 compared to control.

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

The effect of TNFα on cell viability of MDA-MB-231 cells at 5% CO2/Atm for 24 h. The data are presented as the mean±S.E.M. (n=4). Significance of differences from the control were determined by a one-way ANOVA, with a Tukey post-hoc test. *p<0.05.

Cell culture. MDA-MB-231 cells were cultured in 75 cm2 or 175 cm2 flasks containing DMEM supplemented with 10% FBS and 1% 10,000 U/ml penicillin G sodium/10,000 μg/ml streptomycin sulfate. Cells were grown at 37°C with humidified 95% air and 5% CO2.

Cell viability assay. Alamar Blue cell viability assay was used to determine cytotoxicity. Viable cells are capable of reducing resazurin to resorufin, resulting in fluorescence changes. Briefly, 96-well plates were seeded with MDA-MB-231 cells at a density of 5×104cells/100 μl/well. Cells were treated without or with either DADS (50 μM, 100 μM, 400 μM, 800 μM or 1.2 mM) or TNFα (0.1, 1, 10, 20, 40, 80, 100 ng/ml) for 24 h at 37°C, 5% CO2. Alamar blue (0.1mg/ml in HBSS) was added at 15% v/v to each well, and incubated for 6-8 hrs. Quantitative analysis of dye conversion was measured on a microplate fluorometer–Model 7620-version 5.02 (Cambridge Technologies Inc, Watertown, MA, USA) set at 550/580 (excitation/emission). The data were expressed as a percentage of live untreated controls.

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table I.

Array listing of 62 adipokines evaluated by protein microarray.

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

A: Microarray layout. OSM, Oncostatin M; TPO, thrombopoietin; POS, positive control; NEG, negative control. Each protein is in duplicate. Positive controls are located in the upper left (n=4) and lower right (n=2) corners to insure equal distribution of supernatant (top). B: Microarray chemiluminescent spot intensity analysis of supernatant derived from resting MDA-MB-231 cells. POS controls are located in the upper left and lower right corners with dominant cytokines demarcated. C: Baseline cytokine release in untreated MDA-MB-231 cells corresponding to image. The data are presented as spot intensity and are the mean±S.E.M. (n=6). See Table I for cytokine abbreviations.

Human adipokine obesity array. Sandwich-based obesity arrays purchased from RayBiotech (Norcross, GA, USA) consist of array membranes with 62 different proteins in duplicate. Each experiment was carried out in accordance with manufacturer's instructions. Briefly, antibody-coated array membranes were treated with 1 ml of medium from resting, DADS-treated (100 μM), TNFα-treated (40 ng) and co-treated cells and incubated overnight at 4°C on a rocker/shaker. The medium was decanted, the membranes were washed with wash buffer and then incubated with 1 ml biotin-conjugated antibodies (overnight 4°C). The mixture of biotin-conjugated antibodies were removed and membranes were incubated with horse radish peroxidase -conjugated streptavidin (2 h). After a final wash, membrane intensity was acquired using chemiluminescence and analyzed using Quantity One software (Biorad Laboratories, Hercules. CA, US). Densities were measured as a percentage of the positive controls included on each membrane.

CCL2 detection by ELISA. Supernatants from resting and stimulated (24 h) MDA-MB-231 cells were collected and centrifuged at 100× g for 5 min at 4°C. Specific ELISAs were performed using MCP-1/CCL2 ELISA kit (Raybiotech) following the 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, 100 μl of prepared biotinylated antibody mixture was added to each well. After 1 h, the 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 followed by the addition of 50 μl stop solution 30 min later. The plate was read at 450 nm using UV microplate reader.

Statistical analysis. Statistical analysis was performed using GraphPad Prism (version 3.0; GraphPad Software Inc. San Diego, CA, USA) with significance of difference between the groups assessed using a one-way ANOVA, followed by Tukey post hoc means comparison test, two way ANOVA or Student's t-test.

Results

Both DADS and TNFα initiated a mild loss of cell viability in MDA-MB-231 cells (Figures 1 and 2), respectively. Based on observations from cell viability assays, we elected to use 100 μM of DADS and 40 ng/ml of TNFα as our working concentrations for subsequent evaluation. In order to elucidate cytokines affected by DADS, TNFα or a co-treatment of TNFα with DADS vs. controls, a global assessment was carried out using sandwich-based obesity adipokine arrays from RayBiotech for detection of 62 proteins (Table I). A baseline profile was established for untreated resting MDA-MB-231 cells and presented as probe array layout in Figure 3a and the corresponding array blot in Figure 3b. These show a sustained elevated release of IL6, IL8, TIMP1/2 and PAII in untreated cells. The intensity analysis profile is presented in Figure 3c.

In cells treated with TNFα, there were no differential effects on abundantly-released proteins, however, a major elevation in CCL2 was observed (Figure 4) and this was significantly attenuated by DADS, as shown by dot blot intensity analysis (Figure 5a and b). In order to corroborate these findings, CCL2 was determined using an ELISA protocol (Figure 5c). The findings from this study demonstrate that TNFα, induces up-regulation of CCL2 in human breast cancer cells which is blocked by DADS. Preliminary data from the antibody arrays also suggest a consistent elevation of sTNF receptor I by DADS, in both the control group (1.7 fold p<0.05) and TNFα-treated group (1.52-fold p<0.001), which could lead to attenuated TNF signaling at the receptor site (Figure 6). Future studies are required to further investigate the influence of DADS on TNF receptor signaling pathways.

Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4.

TNFα induced cytokine expression by a dominant fold change in MDA-MB-231 cells. The data show a large differential up-regulation of CCL2 protein release amongst the 62 proteins evaluated. The data are presented as fold change and are the mean±S.E.M. (n=6).

Discussion

The data from this study show that CCL2 induced by TNFα is down-regulated by DADS in human breast carcinoma cells. It is well-known that tumor tissue can release promoting chemokines such as CCL2, amongst chemo-attractants and growth factors, which collectively enhance malignant cell migration, proliferation and invasive properties (6). CCL2 is responsible for triggering the recruitment and mobilization of monocytes, macrophages and other inflammatory components in order to infiltrate the tumor area (13). As in the case with breast cancer, CCL2 mobilizes CD14+ CD16+ monocytes (17) where it can bind to monocyte CCL2 receptors CCR2A/2B (6, 7) enabling differentiation into TAMs, which promote metastasis largely by matrix remodeling (18). TAMS surrounding the perimeter of tumor tissue will also exacerbate the rise in CCL2 by locally-positioned reactive macrophages, astrocytes, microglia immunocompetant/host cells (8, 19). The presence of TAM infiltrates are associated with many types of human cancer, as is an elevated expression of CCL2 as a correlate to poor treatment outcome (20, 21). CCL2 is involved in a number of additional processes including up-regulation of the β-catenin/T-cell factor lymphoid-enhancing factor 1 transcriptional activation complex in breast tumor cells (22) and plays a role in almost every aspect of tumor progression from cell migration, cancer progression to epithelial-to-mesenchymal transition (23). Agents which can suppress CCL2–CCR2 signaling can block monocyte recruitment, inhibit metastasis in vivo (24) and are considered to be an effective therapeutic approach in treatment of human cancer (25, 26).

While future research is required to specifically determine the pathways involved in these effects, it is believed that CCL2 release by human breast cancer cells can be initiated by pro-inflammatory cytokines which occur through nuclear factor κ-B (NF-κB), extracellular signal-regulated kinase signaling (ERK) (27) or poly(ADP-ribose) polymerase-1 (PARP-1)/NF-κB signaling (28, 29). Moreover, elevated NF-κB signaling coincides with elevation of CCL2 and matrix remodeling processes (e.g. elevated expression of matrix metalloproteinases (MMPs) e.g. MMP1 and MMP9 (30). A reciprocal relationship may also exist where CCL2 stimulates MMP9 and MMP2, both induced by elevated circulating levels of TNFα (31). TNFα itself is released by cancer-associated fibroblasts and in some cases, tumor cells (32, 33), in particular breast cancer (34). Elevated levels of CCL2/TNFα/MMP9 also coincide with expression patterns of vascular endothelial growth factor A, TGFβ1 and IL8 which collectively assist with differentiation of human monocytes into TAMS (9), and down-regulation of caspase-3 in cancer cells (35). This dynamic synergy can be potentiated by the direct role of CCL2 in epithelial-to-mesenchymal transition via its ability to up-regulate the transcription factor twist basic helix-loop-helix transcription factor 1, needed for extracellular matrix degradation and metastasis (36). These elements drive many pathological events associated with aggressive tumor pathology.

Figure 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 5.

Spot intensity analysis (A) of antibody-coated array membranes with quantitative analysis of chemiluminescent signal (B) and ELISA (C) for controls, DADS-treated (100 μM), TNFα-treated (40 ng/ml) and co-treated cells. The data are presented as the mean±S.E.M. (n=6) and significance of differences were determined by t-test. *p<0.05.

Figure 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 6.

Soluble TNFRI release in MDA-MB-231 cells for groups: control, DADS-treated (100 μM), TNFα-treated (40 ng/ml) and co-treated cells. The data are presented the mean±S.E.M. as percentage of control (n=6). Significance of differences from the controls in both groups were determined by t-test. *p<0.05.

Garlic contains DADS, which was recently shown to reduce migration and invasion of human colon cancer, in part, mediated by attenuation of signaling pathways involving NF-κB, phosphatidylinositide 3-kinases, mitogen-activated protein kinases and p38 (37). These effects are consistent throughout the literature, where DADS has shown ability to inhibit growth of diverse cancer cell types such as HT-29 (38), HL-60 (39), HCT-15 (human colon tumor cells), SK MEL-2 (skin) and A549 (lung) (40). DADS can also act to suppress CCL2–CCR2 signaling, impede monocyte recruitment, and inhibit metastasis in vivo (24), a very effective therapeutic approach for treatment of human cancer (25, 26). The findings from this study contribute to this body of literature, demonstrating yet another potential avenue for DADS in mitigating tumor progression, which could possibly abrogate infiltration of TAMs that promote metastasis through down-regulating CCL2 expression.

Acknowledgements

This research was supported by the National Center for Research Resources and the National Institute of Minority Health and Health Disparities of the National Institutes of Health through Grant Number 8 G12MD007582-28, and the National Institute on Minority Health and Health Disparities, NIH (1P20 MD006738-01).

  • Received March 14, 2014.
  • Revision received April 21, 2014.
  • Accepted April 22, 2014.
  • Copyright© 2014 International Institute of Anticancer Research (Dr. John G. Delinassios), All rights reserved

References

  1. ↵
    1. Varney ML,
    2. Johansson SL,
    3. Singh RK
    : Tumour-associated macrophage infiltration, neovascularization and aggressiveness in malignant melanoma: role of monocyte chemotactic protein-1 and vascular endothelial growth factor-A. Melanoma Res 15: 417-425, 2005.
    OpenUrlCrossRefPubMed
  2. ↵
    1. Wong MP,
    2. Cheung KN,
    3. Yuen ST,
    4. Fu KH,
    5. Chan AS,
    6. Leung SY,
    7. Chung LP
    : Monocyte chemoattractant protein-1 (MCP-1) expression in primary lymphoepithelioma-like carcinomas (LELCs) of the lung. J Pathol 186: 372-377, 1998.
    OpenUrlCrossRefPubMed
  3. ↵
    1. Daurkin I,
    2. Eruslanov E,
    3. Stoffs T,
    4. Perrin GQ,
    5. Algood C,
    6. Gilbert SM,
    7. Rosser CJ,
    8. Su LM,
    9. Vieweg J,
    10. Kusmartsev S
    : Tumor-associated macrophages mediate immunosuppression in the renal cancer microenvironment by activating the 15-lipoxygenase-2 pathway. Cancer Res 71: 6400-6409, 2011.
    OpenUrlAbstract/FREE Full Text
  4. ↵
    1. Zollo M,
    2. Di Dato V,
    3. Spano D,
    4. De Martino D,
    5. Liguori L,
    6. Marino N,
    7. Vastolo V,
    8. Navas L,
    9. Garrone B,
    10. Mangano G,
    11. Biondi G,
    12. Guglielmotti A
    : Targeting monocyte chemotactic protein-1 synthesis with bindarit induces tumor regression in prostate and breast cancer animal models. Clin Exp Metastasis 29: 585-601, 2012.
    OpenUrlCrossRefPubMed
  5. ↵
    1. Soria G,
    2. Ben-Baruch A
    : The inflammatory chemokines CCL2 and CCL5 in breast cancer. Cancer Lett 267: 271-285, 2008.
    OpenUrlCrossRefPubMed
  6. ↵
    1. Craddock JA,
    2. Lu A,
    3. Bear A,
    4. Pule M,
    5. Brenner MK,
    6. Rooney CM,
    7. Foster AE
    : Enhanced tumor trafficking of GD2 chimeric antigen receptor T cells by expression of the chemokine receptor CCR2b. J Immunother 33: 780-788, 2010.
    OpenUrlCrossRef
  7. ↵
    1. Ksiazkiewicz M,
    2. Gottfried E,
    3. Kreutz M,
    4. Mack M,
    5. Hofstaedter F,
    6. Kunz-Schughart LA
    : Importance of CCL2-CCR2A/2B signaling for monocyte migration into spheroids of breast cancer-derived fibroblasts. Immunobiology 215: 737-747, 2010.
    OpenUrlPubMed
  8. ↵
    1. Leung SY,
    2. Wong MP,
    3. Chung LP,
    4. Chan AS,
    5. Yuen ST
    : Monocyte chemoattractant protein-1 expression and macrophage infiltration in gliomas. Acta Neuropathol 93: 518-527, 1997.
    OpenUrlCrossRefPubMed
  9. ↵
    1. de Vasconcellos JF,
    2. Laranjeira AB,
    3. Zanchin NI,
    4. Otubo R,
    5. Vaz TH,
    6. Cardoso AA,
    7. Brandalise SR,
    8. Yunes JA
    : Increased CCL2 and IL-8 in the bone marrow microenvironment in acute lymphoblastic leukemia. Pediatr Blood Cancer 56: 568-577, 2011.
    OpenUrlCrossRefPubMed
  10. ↵
    1. Allavena P,
    2. Sica A,
    3. Solinas G,
    4. Porta C,
    5. Mantovani A
    : The inflammatory micro-environment in tumor progression: the role of tumor-associated macrophages. Crit Rev Oncol Hematol 66: 1-9, 2008.
    OpenUrlCrossRefPubMed
    1. Steiner JL,
    2. Murphy EA
    : Importance of chemokine (CC-motif) ligand 2 in breast cancer. Int J Biol Markers 27: e179-185, 2012.
    OpenUrlPubMed
  11. ↵
    1. Peng L,
    2. Shu S,
    3. Krauss JC
    : Monocyte chemoattractant protein inhibits the generation of tumor-reactive T cells. Cancer Res 57: 4849-4854, 1997.
    OpenUrlAbstract/FREE Full Text
  12. ↵
    1. Perry JA,
    2. Thamm DH,
    3. Eickhoff J,
    4. Avery AC,
    5. Dow SW
    : Increased monocyte chemotactic protein-1 concentration and monocyte count independently associate with a poor prognosis in dogs with lymphoma. Vet Comp Oncol 9: 55-64, 2011.
    OpenUrlPubMed
  13. ↵
    1. Sanford DE,
    2. Belt BA,
    3. Panni RZ,
    4. Mayer A,
    5. Deshpande AD,
    6. Carpenter D,
    7. Mitchem JB,
    8. Plambeck-Suess SM,
    9. Worley LA,
    10. Goetz BD,
    11. Wang-Gillam A,
    12. Eberlein TJ,
    13. Denardo DG,
    14. Goedegebuure SP,
    15. Linehan DC
    : Inflammatory monocyte mobilization decreases patient survival in pancreatic cancer: a role for targeting the CCL2/CCR2 Axis. Clin Cancer Res 19: 3404-3415, 2013.
    OpenUrlAbstract/FREE Full Text
    1. Sandhu SK,
    2. Papadopoulos K,
    3. Fong PC,
    4. Patnaik A,
    5. Messiou C,
    6. Olmos D,
    7. Wang G,
    8. Tromp BJ,
    9. Puchalski TA,
    10. Balkwill F,
    11. Berns B,
    12. Seetharam S,
    13. de Bono JS,
    14. Tolcher AW
    : A first-in-human, first-in-class, phase I study of carlumab (CNTO 888), a human monoclonal antibody against CC-chemokine ligand 2 in patients with solid tumors. Cancer Chemother Pharmacol 71: 1041-1050, 2013.
    OpenUrlCrossRefPubMed
  14. ↵
    1. Kim EK,
    2. Kim YS,
    3. Milner JA,
    4. Wang TT
    : Indole-3-Carbinol and 3’,3’-Diindolylmethane Modulate Androgen's Effect on C-C Chemokine Ligand 2 and Monocyte Attraction to Prostate Cancer Cells. Cancer Prev Res (Phila) 6: 519-529, 2013.
    OpenUrlAbstract/FREE Full Text
  15. ↵
    1. Feng AL,
    2. Zhu JK,
    3. Sun JT,
    4. Yang MX,
    5. Neckenig MR,
    6. Wang XW,
    7. Shao QQ,
    8. Song BF,
    9. Yang QF,
    10. Kong BH,
    11. Qu X
    : CD16+ monocytes in breast cancer patients: expanded by monocyte chemoattractant protein-1 and may be useful for early diagnosis. Clin Exp Immunol 164: 57-65, 2011.
    OpenUrlCrossRefPubMed
  16. ↵
    1. Mishra P,
    2. Banerjee D,
    3. Ben-Baruch A
    : Chemokines at the crossroads of tumor-fibroblast interactions that promote malignancy. J Leukoc Biol 89: 31-39, 2011.
    OpenUrlAbstract/FREE Full Text
  17. ↵
    1. Zhang J,
    2. Patel L,
    3. Pienta KJ
    : CC chemokine ligand 2 (CCL2) promotes prostate cancer tumorigenesis and metastasis. Cytokine Growth Factor Rev 21: 41-48, 2010.
    OpenUrlCrossRefPubMed
  18. ↵
    1. Daurkin I,
    2. Eruslanov E,
    3. Stoffs T,
    4. Perrin GQ,
    5. Algood C,
    6. Gilbert SM,
    7. Rosser CJ,
    8. Su LM,
    9. Vieweg J,
    10. Kusmartsev S
    : Tumor-associated macrophages mediate immune suppression in the renal cancer microenvironment by activating the 15-lipoxygenase-2 pathway. Cancer Res 2011.
  19. ↵
    1. Mazur G,
    2. Jaskula E,
    3. Kryczek I,
    4. Dlubek D,
    5. Butrym A,
    6. Wrobel T,
    7. Lange A,
    8. Kuliczkowski K
    : Proinflammatory chemokine gene expression influences survival of patients with non-Hodgkin's lymphoma. Folia Histochem Cytobiol 49: 240-247, 2011.
    OpenUrlCrossRefPubMed
  20. ↵
    1. Mestdagt M,
    2. Polette M,
    3. Buttice G,
    4. Noel A,
    5. Ueda A,
    6. Foidart JM,
    7. Gilles C
    : Transactivation of MCP-1/CCL2 by beta-catenin/TCF-4 in human breast cancer cells. Int J Cancer 118: 35-42, 2006.
    OpenUrlCrossRefPubMed
  21. ↵
    1. Lugassy C,
    2. Wadehra M,
    3. Li X,
    4. Corselli M,
    5. Akhavan D,
    6. Binder SW,
    7. Peault B,
    8. Cochran AJ,
    9. Mischel PS,
    10. Kleinman HK,
    11. Barnhill RL
    : Pilot study on “pericytic mimicry” and potential embryonic/stem cell properties of angiotropic melanoma cells interacting with the abluminal vascular surface. Cancer Microenviron 6: 19-29, 2013.
    OpenUrlPubMed
  22. ↵
    1. Qian BZ,
    2. Li J,
    3. Zhang H,
    4. Kitamura T,
    5. Zhang J,
    6. Campion LR,
    7. Kaiser EA,
    8. Snyder LA,
    9. Pollard JW
    : CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis. Nature 475: 222-225, 2011.
    OpenUrlCrossRefPubMed
  23. ↵
    1. Zhang J,
    2. Patel L,
    3. Pienta KJ
    : Targeting chemokine (C-C motif) ligand 2 (CCL2) as an example of translation of cancer molecular biology to the clinic. Prog Mol Biol Transl Sci 95: 31-53, 2010.
    OpenUrlCrossRefPubMed
  24. ↵
    1. Rafei M,
    2. Galipeau J
    : A CCL2-based fusokine as a novel biopharmaceutical for the treatment of CCR2-driven autoimmune diseases. Crit Rev Immunol 30: 449-461, 2010.
    OpenUrlPubMed
  25. ↵
    1. Lai T,
    2. Wang K,
    3. Hou Q,
    4. Zhang J,
    5. Yuan J,
    6. Yuan L,
    7. You Z,
    8. Xi M
    : Interleukin 17 induces up-regulation of chemokine and cytokine expression via activation of the nuclear factor kappaB and extracellular signal-regulated kinase 1/2 pathways in gynecologic cancer cell lines. Int J Gynecol Cancer 2011.
  26. ↵
    1. Nalla AK,
    2. Gogineni VR,
    3. Gupta R,
    4. Dinh DH,
    5. Rao JS
    : Suppression of uPA and uPAR blocks radiation-induced MCP-1 mediated recruitment of endothelial cells in meningioma. Cell Signal 23: 1299-1310, 2011.
    OpenUrlPubMed
  27. ↵
    1. Ohanna M,
    2. Giuliano S,
    3. Bonet C,
    4. Imbert V,
    5. Hofman V,
    6. Zangari J,
    7. Bille K,
    8. Robert C,
    9. Bressac-de Paillerets B,
    10. Hofman P,
    11. Rocchi S,
    12. Peyron JF,
    13. Lacour JP,
    14. Ballotti R,
    15. Bertolotto C
    : Senescent cells develop a PARP-1 and nuclear factor-{kappa}B-associated secretome (PNAS). Genes Dev 25: 1245-1261, 2011.
    OpenUrlAbstract/FREE Full Text
  28. ↵
    1. Hatfield KJ,
    2. Reikvam H,
    3. Bruserud O
    : The crosstalk between the matrix metalloprotease system and the chemokine network in acute myeloid leukemia. Curr Med Chem 17: 4448-4461, 2010.
    OpenUrlCrossRefPubMed
  29. ↵
    1. Richardson VJ
    : Divergent and synergistic regulation of matrix metalloprotease production by cytokines in combination with C-C chemokines. Int J Immunopathol Pharmacol 23: 715-726, 2010.
    OpenUrlAbstract/FREE Full Text
  30. ↵
    1. Mueller L,
    2. Seggern LV,
    3. Schumacher J,
    4. Goumas F,
    5. Wilms C,
    6. Braun F,
    7. Broering DC
    : TNF-alpha similarly induces IL-6 and MCP-1 in fibroblasts from colorectal liver metastases and normal liver fibroblasts. Biochem Biophys Res Commun 2010.
  31. ↵
    1. Stairs DB,
    2. Bayne LJ,
    3. Rhoades B,
    4. Vega ME,
    5. Waldron TJ,
    6. Kalabis J,
    7. Klein-Szanto A,
    8. Lee JS,
    9. Katz JP,
    10. Diehl JA,
    11. Reynolds AB,
    12. Vonderheide RH,
    13. Rustgi AK
    : Deletion of p120-catenin results in a tumor microenvironment with inflammation and cancer that establishes it as a tumor suppressor gene. Cancer Cell 19: 470-483, 2011.
    OpenUrlCrossRefPubMed
  32. ↵
    1. Cendan JC,
    2. Topping DL,
    3. Pruitt J,
    4. Snowdy S,
    5. Copeland EM 3rd.,
    6. Lind DS
    : Inflammatory mediators stimulate arginine transport and arginine-derived nitric oxide production in a murine breast cancer cell line. J Surg Res 60: 284-288, 1996.
    OpenUrlPubMed
  33. ↵
    1. Shi CL,
    2. Yu CH,
    3. Zhang Y,
    4. Zhao D,
    5. Chang XH,
    6. Wang WH
    : Monocyte chemoattractant protein-1 modulates invasion and apoptosis of PC-3M prostate cancer cells via regulating expression of VEGF, MMP9 and caspase-3. Asian Pac J Cancer Prev 12: 555-559, 2011.
    OpenUrlPubMed
  34. ↵
    1. Low-Marchelli JM,
    2. Ardi VC,
    3. Vizcarra EA,
    4. van Rooijen N,
    5. Quigley JP,
    6. Yang J
    : Twist1 induces CCL2 and recruits macrophages to promote angiogenesis. Cancer Res 73: 662-671, 2013.
    OpenUrlAbstract/FREE Full Text
  35. ↵
    1. Lai KC,
    2. Hsu SC,
    3. Kuo CL,
    4. Yang JS,
    5. Ma CY,
    6. Lu HF,
    7. Tang NY,
    8. Hsia TC,
    9. Ho HC,
    10. Chung JG
    : 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. Environ Toxicol 2011.
  36. ↵
    1. Huang YS,
    2. Xie N,
    3. Su Q,
    4. Su J,
    5. Huang C,
    6. Liao QJ
    : Diallyl disulfide inhibits the proliferation of HT-29 human colon cancer cells by inducing differentially expressed genes. Mol Med Report 4: 553-559, 2011.
    OpenUrlPubMed
  37. ↵
    1. Yi L,
    2. Ji XX,
    3. Lin M,
    4. Tan H,
    5. Tang Y,
    6. Wen L,
    7. Ma YH,
    8. Su Q
    : Diallyl disulfide induces apoptosis in human leukemia HL-60 cells through activation of JNK mediated by reactive oxygen. Pharmazie 65: 693-698, 2010.
    OpenUrlPubMed
  38. ↵
    1. Sundaram SG,
    2. Milner JA
    : Diallyl disulfide induces apoptosis of human colon tumor cells. Carcinogenesis 17: 669-673, 1996.
    OpenUrlAbstract/FREE Full Text
PreviousNext
Back to top

In this issue

Anticancer Research: 34 (6)
Anticancer Research
Vol. 34, Issue 6
June 2014
  • Table of Contents
  • Table of Contents (PDF)
  • Index by author
  • Back Matter (PDF)
  • Ed Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on Anticancer Research.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Diallyl Disulfide Inhibits TNFα-induced CCL2 Release by MDA-MB-231 Cells
(Your Name) has sent you a message from Anticancer Research
(Your Name) thought you would like to see the Anticancer Research web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
5 + 0 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
Diallyl Disulfide Inhibits TNFα-induced CCL2 Release by MDA-MB-231 Cells
DAVID BAUER, ELIZABETH MAZZIO, KARAM FA SOLIMAN, EQUAR TAKA, EBENEZER ORIAKU, TRACEY WOMBLE, SELINA DARLING-REED
Anticancer Research Jun 2014, 34 (6) 2763-2770;

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
Diallyl Disulfide Inhibits TNFα-induced CCL2 Release by MDA-MB-231 Cells
DAVID BAUER, ELIZABETH MAZZIO, KARAM FA SOLIMAN, EQUAR TAKA, EBENEZER ORIAKU, TRACEY WOMBLE, SELINA DARLING-REED
Anticancer Research Jun 2014, 34 (6) 2763-2770;
Reddit logo Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Acknowledgements
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • Forkhead Box Q1 Is a Novel Target of Breast Cancer Stem Cell Inhibition by Diallyl Trisulfide
  • Google Scholar

More in this TOC Section

  • TIG1 Inhibits the mTOR Signaling Pathway in Malignant Melanoma Through the VAC14 Protein
  • Novel α-Trifluoromethyl Chalcone Exerts Antitumor Effects Against Prostate Cancer Cells
  • Differential Effects of Anti-PD-1/PD-L1 Checkpoint Inhibitors on Adhesion Molecules and Cytokine Secretion by THP-1 Monocytes
Show more Experimental Studies

Similar Articles

Keywords

  • Tumor-associated macrophages
  • monocyte chemotactic protein-1
  • garlic constituents
  • diallyl disulfide
Anticancer Research

© 2023 Anticancer Research

Powered by HighWire