Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 13(9)2021 Apr 27.
Article in English | MEDLINE | ID: mdl-33925607

ABSTRACT

The high concentrations of individual phytochemicals in vitro studies cannot be physiologically achieved in humans. Our solution for this concentration gap between in vitro and human studies is to combine two or more phytochemicals. We screened 12 phytochemicals by pairwise combining two compounds at a low level to select combinations exerting the synergistic inhibitory effect of breast cancer cell proliferation. A novel combination of luteolin at 30 µM (LUT30) and indole-3-carbinol 40 µM (I3C40) identified that this combination (L30I40) synergistically constrains ERα+ breast cancer cell (MCF7 and T47D) proliferation only, but not triple-negative breast cancer cells. At the same time, the individual LUT30 and I3C40 do not have this anti-proliferative effect in ERα+ breast cancer cells. Moreover, this combination L30I40 does not have toxicity on endothelial cells compared to the current commercial drugs. Similarly, the combination of LUT and I3C (LUT10 mg + I3C10 mg/kg/day) (IP injection) synergistically suppresses tumor growth in MCF7 cells-derived xenograft mice, but the individual LUT (10 mg/kg/day) and I3C (20 mg/kg/day) do not show an inhibitory effect. This combination synergistically downregulates two major therapeutic targets ERα and cyclin dependent kinase (CDK) 4/6/retinoblastoma (Rb) pathway, both in cultured cells and xenograft tumors. These results provide a solid foundation that a combination of LUT and I3C may be a practical approach to treat ERα+ breast cancer cells after clinical trials.

2.
Biores Open Access ; 7(1): 73-80, 2018.
Article in English | MEDLINE | ID: mdl-29862141

ABSTRACT

Breast cancer is the most common malignancy among Arab women in Eastern Mediterranean Region (EMR). The incidence of breast cancer has substantially increased in recent years among this women population, especially those younger than 50, and the incidence is expected to double by 2030. Considerable experimental evidence supports the potential role of dietary habits and lifestyle in cancer etiology and cancer prevention. In this review we examined the literature for evidence to link dietary choices and the rise in incidence and mortality of breast cancer among women in EMR. A literature search was conducted in PubMed and Ovid MEDLINE databases up to December 2017. The search terms used are breast cancer prevalence, breast cancer incidence worldwide, breast cancer and: nutrition, protein intake, vitamin D intake, fat intake, phytoestrogens, EMR, Arab, Middle East, Gulf countries, the UAE Arab women, breast cancer risk, diet, and chemoprevention. We found evidence to suggest that there is an alarming epidemic of obesity among women in most of the EMR countries, especially Gulf Cooperation Council (GCC) countries. The rise in the new breast cancer cases among women could be attributed to excess body weight. Their dietary pattern, which correlates with obesity, can be an important factor in the etiology of cancer. Although very few studies were found to support a direct causal relationship between obesity and breast cancer in the EMR, circumstantial evidence clearly points to the possible role of the epidemic, obesity, in this population and the startling rise in cases of breast cancer. Well-designed and systematic studies are urgently needed to confirm these associations and to elucidate potential mechanisms. More urgently, calls to action are needed in many sectors and at all levels of society, to establish intensive strategies for reducing obesity and promoting an overall healthy diet. Continued and expanded research on diet, lifestyle, and breast cancer risk is urgently needed to build the foundation for future progress in evidence-based public health efforts.

3.
Carcinogenesis ; 36 Suppl 1: S128-59, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26106135

ABSTRACT

The purpose of this review is to stimulate new ideas regarding low-dose environmental mixtures and carcinogens and their potential to promote invasion and metastasis. Whereas a number of chapters in this review are devoted to the role of low-dose environmental mixtures and carcinogens in the promotion of invasion and metastasis in specific tumors such as breast and prostate, the overarching theme is the role of low-dose carcinogens in the progression of cancer stem cells. It is becoming clearer that cancer stem cells in a tumor are the ones that assume invasive properties and colonize distant organs. Therefore, low-dose contaminants that trigger epithelial-mesenchymal transition, for example, in these cells are of particular interest in this review. This we hope will lead to the collaboration between scientists who have dedicated their professional life to the study of carcinogens and those whose interests are exclusively in the arena of tissue invasion and metastasis.


Subject(s)
Carcinogens, Environmental/adverse effects , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/pathology , Animals , Disease Progression , Environmental Exposure/adverse effects , Epithelial-Mesenchymal Transition/drug effects , Humans
4.
Toxicol Lett ; 233(2): 59-67, 2015 Mar 04.
Article in English | MEDLINE | ID: mdl-25637755

ABSTRACT

We examined the expression kinetics of some of the aryl hydrocarbon receptor (AhR)-regulated genes in LA1 variant cells compared to wild type (WT) Hepa-1 mouse hepatoma cell lines, and we investigated the stability of AhR protein as a key step in the function of this receptor. Treatment of both cell types with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) resulted in increased CYP1A1 and CYP1B1 mRNA with a subsequent down regulation of AhR. We show here that co-treatment with transcription inhibitor actinomycin D (ActD) has reversed the TCDD-induced depletion of AhR protein in WT. However, the proteolytic degradation of AhR in absence of TCDD was significantly higher in LA1 cells than in WT, and ActD treatment reduced this loss. Induction of CYP1A1 and CYP1B1 mRNA by TCDD in WT cells each exhibited bursts of activity in the initial hour which were about 3-fold greater than in LAI cells. The induced mRNA levels in LA1 exhibited a slow and sustained increase approximating the WT levels by 20h. The induction of two other AhR-regulated genes also showed comparable turnover differences between the two types of cell. Thus, altered regulation of the AhR responsive genes in LA1 may result from a difference in AhR stability.


Subject(s)
Gene Expression Regulation/genetics , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Animals , Cell Line, Tumor , Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 CYP1B1/biosynthesis , Dactinomycin/pharmacology , Environmental Pollutants/toxicity , Enzyme Induction/drug effects , Humans , Kinetics , Mice , Polychlorinated Dibenzodioxins/toxicity , Protein Synthesis Inhibitors/pharmacology , Translocation, Genetic
5.
PLoS One ; 9(6): e100103, 2014.
Article in English | MEDLINE | ID: mdl-24932473

ABSTRACT

The aryl hydrocarbon receptor (AhR), a transcription factor that is best known for its role in mediating the toxic responses elicited by poly aromatic hydrocarbons as well as many other environmental factors; is also involved in breast cancer progression. We previously reported that stable knockdown of AhR decreased the tumorigenic properties of the highly metastatic MDA-MB-231 breast cancer cell line; whereas ectopic overexpression of AhR was sufficient to transform immortalized human mammary epithelial cells to exhibit malignant phenotypes. In the present study we investigated the genes that are differentially regulated by AhR and are controlling cellular processes linked to breast cancer. We used Affymetrix Human GeneChip 1.0-ST whole transcriptome arrays to analyze alterations of gene expression resulting from stable AhR knockdown in the MDA-MB-231 breast cancer cell line. The expression of 144 genes was significantly altered with a ≥2.0-fold change and a multiple test corrected p-value ≤0.05, as a result of AhR knockdown. We demonstrate that AhR knockdown alters the expression of several genes known to be linked to cancer. These genes include those involved in tryptophan metabolism (KYNU), cell growth (MUC1 and IL8), cell survival (BIRC3 and BCL3), cell migration and invasion (S100A4 and ABI3), multi-drug resistance (ABCC3) and angiogenesis (VEGFA and CCL2). The identification of the genes and pathways affected by AhR depletion provides new insight into possible molecular events that could explain the reported phenotypic changes. In conclusion AhR knockdown alters the expression of genes known to enhance or inhibit cancer progression; tipping the balance towards a state that counteracts tumor progression.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , RNA, Small Interfering/genetics , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Triple Negative Breast Neoplasms/genetics , Female , Gene Regulatory Networks , Humans , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Aryl Hydrocarbon/genetics , Reverse Transcriptase Polymerase Chain Reaction , Triple Negative Breast Neoplasms/pathology , Tumor Cells, Cultured
6.
Int J Cancer ; 133(12): 2769-80, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-23733406

ABSTRACT

The aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor that belongs to the basic-helix-loop-helix (bHLH)-Per-ARNT-Sim (PAS) superfamily of transcription factors, mediates toxic response induced by environmental chemicals such as polycyclic aromatic hydrocarbons (PAH). AhR is expressed at high levels in several human breast carcinoma cell lines in direct correlation with the degree of their malignancy. Recent studies suggest a possible role for AhR in cancer independent of PAH. Therefore, we established stable AhR knockdown cells of the human breast cancer cell line MDA-MB-231 and analyzed their tumorigenic properties in in vitro and in vivo model systems. In addition we analyzed their response to radiation and chemotherapeutic treatment. AhR knockdown attenuated these cells tumorigenic properties in vitro including proliferation, anchorage independent growth, migration and apoptosis and reduced orthotopic xenograft tumor growth and lung metastasis in vivo. Notably, we observed that AhR knockdown enhanced radiation-induced apoptosis as well as significantly decreased cell clonogenic survival. Furthermore, AhR knockdown in MDA-MB-231 cells sensitized them to paclitaxel treatment, evident by a decrease in the required cytotoxic dose. Subsequent analysis revealed AhR knockdown significantly reduced phosphorylation of AKT, which impacts cell proliferation and survival. Apoptosis-focused gene expression analyses revealed an altered expression of genes regulating apoptosis in MDA-MB-231 cells. Collectively, our data identify AhR as a potential novel therapeutic target in the treatment of metastatic breast cancer.


Subject(s)
Breast Neoplasms/pathology , Cell Proliferation , Receptors, Aryl Hydrocarbon/physiology , Animals , Apoptosis , Cell Cycle , Cell Line, Tumor , Female , Humans , Lung Neoplasms/secondary , Mice , RNA Interference , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/genetics , Triple Negative Breast Neoplasms/pathology
7.
J Cancer Ther ; 4(7): 1177-1186, 2013 Sep.
Article in English | MEDLINE | ID: mdl-25068070

ABSTRACT

The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that regulates a battery of genes in response to exposure to a broad class of environmental poly aromatic hydrocarbons (PAH). AhR is historically characterized for its role in mediating the toxicity and adaptive responses to these chemicals, however mounting evidence has established a role for it in ligand-independent physiological processes and pathological conditions, including cancer. The AhR is overexpressed and constitutively activated in advanced breast cancer cases and was shown to drive the progression of breast cancer. In this article we will review the current state of knowledge on the possible role of AhR in breast cancer and how it will be exploited in targeting AhR for breast cancer therapy.

9.
Toxicol Lett ; 166(2): 150-9, 2006 Oct 10.
Article in English | MEDLINE | ID: mdl-16891067

ABSTRACT

The induction of CYP1A1 expression by oltipraz, a synthetic chemo-preventive agent, which increases intracellular calcium concentration, has previously been shown to result from transcriptional activation of CYP1A1 gene mediated by the Ah receptor (AhR), although oltipraz does not bind the receptor. The present study investigated the possible mechanisms of oltipraz-induced activation of AhR and the subsequent induction of CYP1A1 transcription. Treatment of the human metastatic breast cancer cell line MT-2 with oltipraz results in a concentration-dependent increase in the activity of the calcium-dependent calpain, as measured towards the BOC-LM-CMAC fluorescent substrate. This increase in calpain activity was coupled with the AhR activation, as evidenced by its nuclear localization and increased transcription of CYP1A1 gene. Treatment of cells with calpain specific inhibitor MDL 28170 completely blocked the oltipraz-induced nuclear translocation of AhR and subsequent CYP1A1 expression. Furthermore, treatment with oltipraz resulted in the classical ligand-dependent down-regulation of AhR protein, in a concentration dependent manner. The presented data established for the first time a mechanism of activating AhR and its transcription of CYP1A1 by oltipraz through activation of calcium-dependent calpain.


Subject(s)
Anticarcinogenic Agents/pharmacology , Calcium/metabolism , Calpain/metabolism , Cytochrome P-450 CYP1A1/biosynthesis , Pyrazines/pharmacology , Receptors, Aryl Hydrocarbon/metabolism , Animals , Cell Line, Tumor , Down-Regulation , Enzyme Induction/drug effects , Female , Humans , Immunohistochemistry , Mice , Microscopy, Fluorescence , Reverse Transcriptase Polymerase Chain Reaction , Thiones , Thiophenes
10.
Mol Pharmacol ; 70(5): 1481-7, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16891617

ABSTRACT

The aryl hydrocarbon receptor (AhR) is a ligand-activated basic-helix-loop-helix transcription factor that binds polyaromatic hydrocarbons (PAH), such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), and mediates their toxicity. Binding of PAH to AhR in the cytoplasm triggers a poorly defined transformation step of the receptor into a nuclear transcription factor. In this study, we show that the calcium-dependent cysteine protease calpain plays a major role in the ligand-induced transformation and signaling of AhR. Fluorescence imaging measurements showed that TCDD treatment elevates intracellular calcium, providing the trigger for calpain activation, as measured toward t-butoxycarbonyl-Leu-Met-chloromethylaminocoumarin, a calpain-specific substrate. Inhibition of calpain activity by the N-benzyloxycarbonyl-Val-Phe-aldehyde (MDL28170) blocked the TCDD-induced nuclear translocation of AhR in Hepa1c1c7 mouse hepatoma cell line. Treatment of the human metastatic breast carcinoma cell line MT-2 with MDL28170 and 3-(4-iodophenyl)-2-mercapto-(Z)-2-propenoic acid (PD 150606), two calpain-selective inhibitors, completely abolished the TCDD-induced transactivation of AhR as assessed by transcription of CYP1A1 gene. Previous studies have established that after TCDD-induced transactivation, the AhR undergoes a massive depletion; we show here that selective calpain inhibitors can block this step, which suggests that the ligand-induced down-regulation of the AhR is calpain-dependent. The data presented support a major role for calpain in the AhR transformation, transactivation, and subsequent down-regulation, and provide a possible explanation for many of the reported phenomena of ligand-independent activation of AhR.


Subject(s)
Calpain/metabolism , Down-Regulation/drug effects , Polychlorinated Dibenzodioxins/pharmacology , Receptors, Aryl Hydrocarbon/metabolism , Animals , Calcium/metabolism , Calpain/antagonists & inhibitors , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cytochrome P-450 CYP1A1/genetics , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Humans , Mice , Protein Processing, Post-Translational/drug effects , Protein Transport/drug effects , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects , Tumor Cells, Cultured
11.
Breast Cancer Res ; 6(3): R215-31, 2004.
Article in English | MEDLINE | ID: mdl-15084245

ABSTRACT

INTRODUCTION: Transforming growth factor (TGF)-beta1 is proposed to inhibit the growth of epithelial cells in early tumorigenesis, and to promote tumor cell motility and invasion in the later stages of carcinogenesis through the induction of an epithelial to mesenchymal transition (EMT). EMT is a multistep process that is characterized by changes in cell morphology and dissociation of cell-cell contacts. Although there is growing interest in TGF-beta1-mediated EMT, the phenotype is limited to only a few murine cell lines and mouse models. METHODS: To identify alternative cell systems in which to study TGF-beta1-induced EMT, 18 human and mouse established cell lines and cultures of two human primary epithelial cell types were screened for TGF-beta1-induced EMT by analysis of cell morphology, and localization of zonula occludens-1, E-cadherin, and F-actin. Sensitivity to TGF-beta1 was also determined by [3H]thymidine incorporation, flow cytometry, phosphorylation of Smad2, and total levels of Smad2 and Smad3 in these cell lines and in six additional cancer cell lines. RESULTS: TGF-beta1 inhibited the growth of most nontransformed cells screened, but many of the cancer cell lines were insensitive to the growth inhibitory effects of TGF-beta1. In contrast, TGF-beta1 induced Smad2 phosphorylation in the majority of cell lines, including cell lines resistant to TGF-beta1-mediated cell cycle arrest. Of the cell lines screened only two underwent TGF-beta1-induced EMT. CONCLUSION: The results presented herein show that, although many cancer cell lines have lost sensitivity to the growth inhibitory effect of TGF-beta1, most show evidence of TGF-beta1 signal transduction, but only a few cell lines undergo TGF-beta1-mediated EMT.


Subject(s)
Epithelial Cells/cytology , Neoplasm Invasiveness/physiopathology , Transforming Growth Factor beta/physiology , Actins/analysis , Animals , Cadherins/analysis , Cell Adhesion/physiology , Cell Cycle/physiology , Cell Line, Transformed/cytology , Cell Line, Tumor/cytology , Cell Movement/physiology , Cells, Cultured/cytology , DNA-Binding Proteins/analysis , DNA-Binding Proteins/metabolism , Disease Progression , Epithelial Cells/metabolism , Humans , In Vitro Techniques , Membrane Proteins/analysis , Mesoderm , Mice , Neoplasm Proteins/physiology , Phenotype , Phosphoproteins/analysis , Phosphorylation , Protein Processing, Post-Translational , Smad2 Protein , Smad3 Protein , Trans-Activators/analysis , Trans-Activators/metabolism , Transforming Growth Factor beta1 , Zonula Occludens-1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...