Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 76
1.
BMB Rep ; 53(5): 284-289, 2020 May.
Article En | MEDLINE | ID: mdl-32317086

Tamoxifen, a nonsteroidal estrogen receptor (ER) antagonist, is used routinely as a chemotherapeutic agent for ER-positive breast cancer. However, it is also causes side effects, including retinotoxicity. The retinal pigment epithelium (RPE) has been recognized as the primary target of tamoxifen-induced retinotoxicity. The RPE plays an essential physiological role in the normal functioning of the retina. Nonetheless, potential therapeutic agents to prevent tamoxifen-induced retinotoxicity in breast cancer patients have not been investigated. Here, we evaluated the action mechanisms of sulfasalazine against tamoxifen- induced RPE cell death. Tamoxifen induced reactive oxygen species (ROS)-mediated autophagic cell death and caspase-1-mediated pyroptosis in RPE cells. However, sulfasalazine reduced tamoxifen-induced total ROS and ROS-mediated autophagic RPE cell death. Also, mRNA levels of tamoxifen-induced pyroptosis-related genes, IL-1ß, NLRP3, and procaspase-1, also decreased in the presence of sulfasalazine in RPE cells. Additionally, the mRNA levels of tamoxifen-induced AMD-related genes, such as complement factor I (CFI), complement factor H (CFH), apolipoprotein E (APOE), apolipoprotein J (APOJ), toll-like receptor 2 (TLR2) and toll-like receptor 4 (TLR4), were downregulated in RPE cells. Together, these data provide novel insight into the therapeutic effects of sulfasalazine against tamoxifen-induced RPE cell death. [BMB Reports 2020; 53(5): 284-289].


Retinal Pigment Epithelium/drug effects , Sulfasalazine/pharmacology , Tamoxifen/antagonists & inhibitors , Cell Death/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Reactive Oxygen Species/metabolism , Retinal Pigment Epithelium/metabolism , Structure-Activity Relationship , Tamoxifen/pharmacology
2.
Steroids ; 153: 108521, 2020 01.
Article En | MEDLINE | ID: mdl-31604073

Estrogen receptors (ERs) and the PTEN-Akt-mTor pathway are important growth regulators in human breast cancer cells, which both are known to affect response to tamoxifen therapy. Recently it was reported that ERß activates PTEN expression and tamoxifen sensitivity of human breast cancer cells. In this study we examined whether expression of ERß in turn might be affected by tumor suppressor PTEN, analyzed the effect of this interaction on tamoxifen response and the co-expression of both genes in human breast cancer samples. After siRNA-mediated PTEN knockdown, Western blot analysis revealed a reduction of ERß protein expression by 67.2% in MCF-7 cells and by 73.6% in T-47D cells (both p < 0.01), results which could be verified on the mRNA level. In cells with normal PTEN and ERß status, after 6 days of treatment with 1 µM 4-OH tamoxifen, E2-driven proliferation was decreased by 64.5% in MCF-7 and by 57.7% in T-47D cells (both p < 0.01). After knockdown of PTEN expression, the same concentration of 4-OH TAM reduced E2-triggered growth only by 34.9% (MCF-7) and by 41.8% (T-47D) (both p < 0.01 vs control siRNA). Importantly, treatment with ERß agonist DPN (5 nM) significantly decreased the inhibitory effect of a PTEN knockdown on tamoxifen response of both cell lines (p < 0.05). Additionally, Spearmans rank association analysis of PTEN and ERß 1 mRNA levels in 115 normal and malignant breast tissue samples revealed a strong positive correlation of both genes (rho = 0.6085, p < 0.0001). The data of previous studies reporting an important role of ERß in tamoxifen sensitivity and our findings suggest down-regulation of ERß triggered by PTEN knockdown contributed to the decreased response of breast cancer cells to tamoxifen observed in this study. Our data also suggest expression of ERß might be maintained by tumor suppressor PTEN in human breast cancer cells.


Antineoplastic Agents, Hormonal/antagonists & inhibitors , Breast Neoplasms/drug therapy , Estrogen Receptor beta/antagonists & inhibitors , PTEN Phosphohydrolase/antagonists & inhibitors , RNA, Small Interfering/pharmacology , Tamoxifen/antagonists & inhibitors , Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Humans , MCF-7 Cells , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Structure-Activity Relationship , Tamoxifen/pharmacology , Tumor Cells, Cultured
3.
J Virol ; 93(23)2019 12 01.
Article En | MEDLINE | ID: mdl-31511378

We previously reported that herpes simplex virus (HSV) glycoprotein K (gK) binds to signal peptide peptidase (SPP), also known as minor histocompatibility antigen H13. Binding of gK to SPP is required for HSV-1 infectivity in vitro SPP is a member of the γ-secretase family, and mice lacking SPP are embryonic lethal. To determine how SPP affects HSV-1 infectivity in vivo, the SPP gene was deleted using a tamoxifen-inducible Cre recombinase driven by the ubiquitously expressed ROSA26 promoter. SPP mRNA was reduced by more than 93% in the cornea and trigeminal ganglia (TG) and by 99% in the liver of tamoxifen-injected mice, while SPP protein expression was reduced by 90% compared to the level in control mice. Mice lacking SPP had significantly less HSV-1 replication in the eye as well as reduced gK, UL20, ICP0, and gB transcripts in the cornea and TG compared to levels in control mice. In addition, reduced infiltration of CD45+, CD4+, CD8+, F4/80+, CD11c+, and NK1.1+ T cells was observed in the cornea and TG of SPP-inducible knockout mice compared to that in control mice. Finally, in the absence of SPP, latency was significantly reduced in SPP-inducible knockout mice compared to that in control mice. Thus, in this study we have generated SPP-inducible knockout mice and shown that the absence of SPP affects virus replication in the eye of ocularly infected mice and that this reduction is correlated with the interaction of gK and SPP. These results suggest that blocking this interaction may have therapeutic potential in treating HSV-1-associated eye disease.IMPORTANCE Glycoprotein K (gK) is an essential and highly conserved HSV-1 protein. Previously, we reported that gK binds to SPP, an endoplasmic reticulum (ER) protein, and blocking this binding reduces virus infectivity in vitro and also affects gK and UL20 subcellular localization. To evaluate the function of gK binding to SPP in vivo, we generated SPP-inducible knockout mice and observed the following in the absence of SPP: (i) that significantly less HSV-1 replication was seen in ocularly infected mice than in control mice; (ii) that expression of various HSV-1 genes and cellular infiltrates in the eye and trigeminal ganglia of infected mice was less than that in control mice; and (iii) that latency was significantly reduced in infected mice. Thus, blocking of gK binding to SPP may be a useful tool to control HSV-1-induced eye disease in patients with herpes stromal keratitis (HSK).


Aspartic Acid Endopeptidases/metabolism , Herpes Simplex/immunology , Herpesvirus 1, Human/physiology , Viral Proteins/metabolism , Animals , Aspartic Acid Endopeptidases/drug effects , Aspartic Acid Endopeptidases/genetics , Cell Line , Cornea/virology , Disease Models, Animal , Eye Infections/immunology , Eye Infections/virology , Herpes Simplex/virology , Immediate-Early Proteins , Keratitis, Herpetic , Liver/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Tamoxifen/antagonists & inhibitors , Transcriptome , Trigeminal Ganglion/virology , Ubiquitin-Protein Ligases , Viral Load , Virus Latency , Virus Replication
4.
Nucleic Acids Res ; 47(5): 2322-2335, 2019 03 18.
Article En | MEDLINE | ID: mdl-30597111

Estrogen/ERα signaling is critical for breast cancer progression and therapeutic treatments. Thus, identifying new regulators of this pathway will help to develop new therapeutics to overcome chemotherapy resistance of the breast cancer cells. Here, we report Ajuba directly interacts with ERα to potentiate ERα target gene expression, and biologically Ajuba promotes breast cancer cell growth and contributes to tamoxifen resistance of these cells. Ajuba constitutively binds the DBD and AF2 regions of ERα, and these interactions can be markedly enhanced by estrogen treatment. Mechanistically, Ajuba recruits DBC1 and CBP/p300 and forms a ternary complex to co-activate ERα transcriptional activity and concomitantly enhances ERα acetylation. Moreover, components of this complex can be found at endogenous promoters containing functional ERα responsive elements. Taken together, these data demonstrate that Ajuba functions as a novel co-activator of ERα and that Ajuba/DBC1/CBP/p300 ternary complex may be a new target for developing therapeutics to treat breast cancer.


Breast Neoplasms/genetics , Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/metabolism , Gene Expression Regulation, Neoplastic , LIM Domain Proteins/metabolism , Tumor Suppressor Proteins/metabolism , p300-CBP Transcription Factors/metabolism , Acetylation , Breast Neoplasms/pathology , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation/drug effects , Estrogen Receptor alpha/agonists , Estrogens/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , LIM Domain Proteins/genetics , Nerve Tissue Proteins , Protein Binding/drug effects , Tamoxifen/antagonists & inhibitors , Tamoxifen/pharmacology , Transcription, Genetic/drug effects
5.
Life Sci ; 202: 78-88, 2018 Jun 01.
Article En | MEDLINE | ID: mdl-29626531

AIMS: This study investigated the hepatoprotective effects of both zafirlukast and vincamine and their possible role in the treatment of tamoxifen-induced liver injury in rats. MATERIALS AND METHODS: Female Wistar rats were divided into five groups (10 rats each). Groups I and II received 1% Tween 80 and served as normal and tamoxifen controls, respectively. Groups III, IV and V were treated with zafirlukast (80 mg/kg), vincamine (10 mg/kg) and a combination of zafirlukast (80 mg/kg) and vincamine (10 mg/kg), respectively for 10 successive days. Tamoxifen was given orally to all groups, except for 1st group, in the dose of 45 mg/kg for 10 days to induce liver injury. Subsequently, rats were sacrificed for biochemical, histopathological, Immunohistochemistry, PCR and western blot assessment. KEY FINDINGS: Tamoxifen-induced liver injury was reflected by alterations in estimated biochemical parameters, activation of JNK/ERK pathway, increased expression of NF-κB, liver oxidative stress and inflammatory markers parallel to histopathological changes in liver tissue. Treatment of rats with zafirlukast and vincamine ameliorated tamoxifen induced hepatic cell injury via suppressing oxidative stress, inflammatory markers, caspases-3, p-JNK/p-ERK and NF-κB pathways. SIGNIFICANCE: Zafirlukast and vincamine may be regarded as potential therapeutic strategies with antioxidant and anti-inflammatory activities against tamoxifen-induced oxidative damage in rat liver.


Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Chemical and Drug Induced Liver Injury/prevention & control , Estrogen Antagonists/toxicity , JNK Mitogen-Activated Protein Kinases/drug effects , Leukotriene Antagonists/pharmacology , MAP Kinase Signaling System/drug effects , Oxidative Stress/drug effects , Tamoxifen/antagonists & inhibitors , Tosyl Compounds/pharmacology , Vasodilator Agents/pharmacology , Vincamine/pharmacology , Animals , Caspase 3/biosynthesis , Caspase 3/genetics , Chemical and Drug Induced Liver Injury/pathology , Cytokines/blood , Female , Immunohistochemistry , Indoles , Liver Function Tests , NF-kappa B/drug effects , NF-kappa B/metabolism , Phenylcarbamates , Rats , Rats, Wistar , Sulfonamides , Tamoxifen/toxicity
6.
J Natl Cancer Inst ; 108(3)2016 Mar.
Article En | MEDLINE | ID: mdl-26631176

BACKGROUND: Controversy persists about whether certain antidepressants reduce tamoxifen's effectiveness on lowering breast cancer recurrence. We investigated whether taking tamoxifen and antidepressants (in particular, paroxetine) concomitantly is associated with an increased risk of recurrence or contralateral breast cancer. METHODS: We examined 16 887 breast cancer survivors (TNM stages 0-II) diagnosed between 1996 and 2007 and treated with tamoxifen in two California health plans. Women were followed-up through December 31, 2009, for subsequent breast cancer. The main exposure was the percent of days of overlap when both tamoxifen and an antidepressant (paroxetine, fluoxetine, other selective serotonin reuptake inhibitors, tricyclics, and other classes) were used. Hazard ratios (HRs) and 95% confidence intervals (CIs) were estimated using multivariable Cox regression models with time-varying medication variables. RESULTS: Of the 16 887 women, half (n = 8099) used antidepressants and 2946 women developed subsequent breast cancer during the 14-year study period. We did not find a statistically significant increased risk of subsequent breast cancer in women who concurrently used paroxetine and tamoxifen. For 25%, 50%, and 75% increases in percent overlap days between paroxetine and tamoxifen, hazard ratios were 1.06 (95% CI = 0.98 to 1.14, P = .09), 1.13 (95% CI = 0.98 to 1.30, P = .09), and 1.20 (95% CI = 0.97 to 1.49, P = .09), respectively, in the first year of tamoxifen treatment but were not statistically significant. Hazard ratios decreased to 0.94 (95% CI = 0.81 to 1.10, P = .46), 0.89 (95% CI = 0.66 to 1.20, P = .46), and 0.85 (95% CI = 0.54 to 1.32, P = .46) by the fifth year (all non-statistically significantly). Absolute subsequent breast cancer rates were similar among women who used paroxetine concomitantly with tamoxifen vs tamoxifen-only users. For the other antidepressants, we again found no such associations. CONCLUSIONS: Using the comprehensive electronic health records of insured patients, we did not observe an increased risk of subsequent breast cancer in women who concurrently used tamoxifen and antidepressants, including paroxetine.


Antidepressive Agents, Second-Generation/therapeutic use , Antineoplastic Agents, Hormonal/antagonists & inhibitors , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Depression/drug therapy , Neoplasm Recurrence, Local/prevention & control , Paroxetine/therapeutic use , Tamoxifen/antagonists & inhibitors , Tamoxifen/therapeutic use , Adult , Aged , Breast Neoplasms/epidemiology , Breast Neoplasms/pathology , Breast Neoplasms/prevention & control , Breast Neoplasms/psychology , Cohort Studies , Depression/etiology , Female , Health Resources/statistics & numerical data , Humans , Middle Aged , Neoplasm Recurrence, Local/epidemiology , Neoplasm Staging , Odds Ratio , Proportional Hazards Models , Survivors , United States/epidemiology
8.
Drug Metab Dispos ; 43(6): 889-97, 2015 Jun.
Article En | MEDLINE | ID: mdl-25795461

We previously reported upregulation of UGT2B15 by 17ß-estradiol in breast cancer MCF7 cells via binding of the estrogen receptor α (ERα) to an estrogen response unit (ERU) in the proximal UGT2B15 promoter. In the present study, we show that this ERα-mediated upregulation was significantly reduced by two ER antagonists (fulvestrant and raloxifene) but was not affected by a third ER antagonist, 4-hydroxytamoxifen (4-OHTAM), a major active tamoxifen (TAM) metabolite. Furthermore, we found that, similar to 17ß-estradiol, 4-OHTAM and endoxifen (another major active TAM metabolite) elevated UGT2B15 mRNA levels, and that this stimulation was significantly abrogated by fulvestrant. Further experiments using 4-OHTAM revealed a critical role for ERα in this regulation. Specifically; knockdown of ERα expression by anti-ERα small interfering RNA reduced the 4-OHTAM-mediated induction of UGT2B15 expression; 4-OHTAM activated the wild-type but not the ERU-mutated UGT2B15 promoter; and chromatin immunoprecipitation assays showed increased ERα occupancy at the UGT2B15 ERU in MCF7 cells upon exposure to 4-OHTAM. Together, these data indicate that both 17ß-estradiol and the antiestrogen 4-OHTAM upregulate UGT2B15 in MCF7 cells via the same ERα-signaling pathway. This is consistent with previous observations that both 17ß-estradiol and TAM upregulate a common set of genes in MCF7 cells via the ER-signaling pathway. As 4-OHTAM is a UGT2B15 substrate, the upregulation of UGT2B15 by 4-OHTAM in target breast cancer cells is likely to enhance local metabolism and inactivation of 4-OHTAM within the tumor. This represents a potential mechanism that may reduce TAM therapeutic efficacy or even contribute to the development of acquired TAM resistance.


Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/drug therapy , Drugs, Investigational/pharmacology , Enzyme Induction/drug effects , Estrogen Receptor Antagonists/pharmacology , Glucuronosyltransferase/metabolism , Tamoxifen/analogs & derivatives , Antineoplastic Agents, Hormonal/antagonists & inhibitors , Antineoplastic Agents, Hormonal/metabolism , Breast Neoplasms/metabolism , Drugs, Investigational/chemistry , Drugs, Investigational/metabolism , Estrogen Receptor Antagonists/chemistry , Estrogen Receptor Antagonists/metabolism , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Genes, Reporter/drug effects , Glucuronosyltransferase/antagonists & inhibitors , Glucuronosyltransferase/genetics , Humans , MCF-7 Cells , Mutation , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Promoter Regions, Genetic/drug effects , RNA Interference , Response Elements/drug effects , Signal Transduction/drug effects , Substrate Specificity , Tamoxifen/antagonists & inhibitors , Tamoxifen/metabolism , Tamoxifen/pharmacology
9.
Indian J Exp Biol ; 52(11): 1052-61, 2014 Nov.
Article En | MEDLINE | ID: mdl-25434100

Wide spread use of Di-(2-ethylhexyl) phthalate (DEHP) has made it a ubiquitous contaminant in today's environment, responsible for possible carcinogenic and endocrine disrupting effects. In the present investigation an integrative toxicoproteomic approach was made to study the estrogenic potential of DEHP. In vitro experiments carried out with DEHP (0.1-100 microM) induced proliferations (E-screen assay) in human estrogen receptors-alpha (ERalpha) positive MCF-7 and ERalpha negative MDA-MB-231 breast cancer cells irrespective of their ERa status. Further, DEHP suppressed tamoxifen (a potent anti-breast cancer drug) induced apoptosis in both cell types as shown by flowcytometric cell cycle analysis. Label-free quantitative proteomics analysis of the cell secretome of both the cell lines indicated a wide array of stress related, structural and receptor binding proteins that were affected due to DEHP exposure. The secretome of DEHP treated MCF-7 cells revealed the down regulation of lactotransferrin, an ERalpha responsive iron transport protein. The results indicated that toxicological effects of DEHP did not follow an ERa signaling pathway. However, the differential effects in MCF-7 and MDA-MB-231 cell lines indicate that ERa might have an indirect modulating effect on DEHP induced toxicity.


Breast Neoplasms/pathology , Diethylhexyl Phthalate/toxicity , Environmental Pollutants/toxicity , Estrogen Receptor alpha/physiology , Estrogens , Neoplasm Proteins/physiology , Neoplasms, Hormone-Dependent/pathology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Division/drug effects , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Estrogen Receptor alpha/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lactoferrin/biosynthesis , Lactoferrin/genetics , Lactoferrin/metabolism , MCF-7 Cells/drug effects , MCF-7 Cells/metabolism , Mass Spectrometry/instrumentation , Microchemistry/instrumentation , Neoplasm Proteins/drug effects , Neoplasm Proteins/metabolism , Proteomics , Tamoxifen/antagonists & inhibitors , Tamoxifen/pharmacology
10.
BMC Cancer ; 14: 119, 2014 Feb 22.
Article En | MEDLINE | ID: mdl-24559095

BACKGROUND: Yes-associated protein (YAP1) is frequently reported to function as an oncogene in many types of cancer, but in breast cancer results remain controversial. We set out to clarify the role of YAP1 in breast cancer by examining gene and protein expression in subgroups of patient material and by downregulating YAP1 in vitro and studying its role in response to the widely used anti-estrogen tamoxifen. METHODS: YAP1 protein intensity was scored as absent, weak, intermediate or strong in two primary breast cancer cohorts (n = 144 and n = 564) and mRNA expression of YAP1 was evaluated in a gene expression dataset (n = 1107). Recurrence-free survival was analysed using the log-rank test and Cox multivariate analysis was used to test for independence. WST-1 assay was employed to measure cell viability and a luciferase ERE (estrogen responsive element) construct was used to study the effect of tamoxifen, following downregulation of YAP1 using siRNAs. RESULTS: In the ER+ (Estrogen Receptor α positive) subgroup of the randomised cohort, YAP1 expression was inversely correlated to histological grade and proliferation (p = 0.001 and p = 0.016, respectively) whereas in the ER- (Estrogen Receptor α negative) subgroup YAP1 expression correlated positively to proliferation (p = 0.005). Notably, low YAP1 mRNA was independently associated with decreased recurrence-free survival in the gene expression dataset, specifically for the luminal A subgroup (p < 0.001) which includes low proliferating tumours of lower grade, usually associated with a good prognosis. This subgroup specificity led us to hypothesize that YAP1 may be important for response to endocrine therapies, such as tamoxifen, extensively used for luminal A breast cancers. In a tamoxifen randomised patient material, absent YAP1 protein expression was associated with impaired tamoxifen response which was significant upon interaction analysis (p = 0.042). YAP1 downregulation resulted in increased progesterone receptor (PgR) expression and a delayed and weaker tamoxifen in support of the clinical data. CONCLUSIONS: Decreased YAP1 expression is an independent prognostic factor for recurrence in the less aggressive luminal A breast cancer subgroup, likely due to the decreased tamoxifen sensitivity conferred by YAP1 downregulation.


Adaptor Proteins, Signal Transducing/biosynthesis , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Phosphoproteins/biosynthesis , Tamoxifen , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Breast Neoplasms/diagnosis , Cell Line, Tumor , Cohort Studies , Female , Follow-Up Studies , Humans , Middle Aged , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/metabolism , Phosphoproteins/antagonists & inhibitors , Tamoxifen/antagonists & inhibitors , Tamoxifen/therapeutic use , Transcription Factors , Treatment Outcome , YAP-Signaling Proteins
11.
J Cell Mol Med ; 18(2): 305-13, 2014 Feb.
Article En | MEDLINE | ID: mdl-24266867

Vitamin C is generally thought to enhance immunity and is widely taken as a supplement especially during cancer treatment. Tamoxifen (TAM) has both cytostatic and cytotoxic properties for breast cancer. TAM engaged mitochondrial oestrogen receptor beta in MCF-7 cells and induces apoptosis by activation of pro-caspase-8 followed by downstream events, including an increase in reactive oxygen species and the release of pro-apoptotic factors from the mitochondria. In addition to that, TAM binds with high affinity to the microsomal anti-oestrogen-binding site and inhibits cholesterol esterification at therapeutic doses. This study aimed to investigate the role of vitamin C in TAM-mediated apoptosis. Cells were loaded with vitamin C by exposure to dehydroascorbic acid, thereby circumventing in vitro artefacts associated with the poor transport and pro-oxidant effects of ascorbic acid. Pre-treatment with vitamin C caused a dose-dependent attenuation of cytotoxicity, as measured by acridine-orange/propidium iodide (AO/PI) and Annexin V assay after treatment with TAM. Vitamin C dose-dependently protected cancer cells against lipid peroxidation caused by TAM treatment. By real-time PCR analysis, an impressive increase in FasL and tumour necrosis factor-α (TNF-α) mRNA was detected after TAM treatment. In addition, a decrease in mitochondrial transmembrane potential was observed. These results support the hypothesis that vitamin C supplementation during cancer treatment may detrimentally affect therapeutic response.


Antineoplastic Agents, Hormonal/pharmacology , Dehydroascorbic Acid/pharmacology , Tamoxifen/pharmacology , Annexin A5 , Caspase 8/genetics , Caspase 8/metabolism , Cell Death/drug effects , Cell Line, Tumor , Dehydroascorbic Acid/metabolism , Dose-Response Relationship, Drug , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , Female , Humans , Lipid Peroxidation/drug effects , Membrane Potential, Mitochondrial/drug effects , Propidium , Reactive Oxygen Species/metabolism , Tamoxifen/antagonists & inhibitors , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
12.
Ned Tijdschr Geneeskd ; 157(40): A6069, 2013.
Article Nl | MEDLINE | ID: mdl-24004923

OBJECTIVE: Adjuvant hormonal therapy using tamoxifen or aromatase inhibitors result in menopausal symptoms. Non-prescription dietary supplements containing plant-based phytoestrogens are often used to relieve menopausal symptoms. Phytoestrogens may reduce the action of tamoxifen and aromatase inhibitors. The purpose of this study is to gain insight into the potential risks of phytoestrogen supplementation during adjuvant hormonal therapy for breast cancer using tamoxifen or aromatase inhibitors. DESIGN: Literature survey. METHOD: We performed a PubMed search for articles about studies on the effects of phytoestrogens on breast cancer treatment efficacy. We also searched on the basis of references in the articles we found in PubMed. RESULTS: Studies based on in-vitro models as well as experimental animal models were identified. The studies on the interaction between phytoestrogen and tamoxifen showed contradictory results. The few studies pertaining to the interactive effect of phytoestrogens on aromatase inhibitors all showed a reduction in therapeutic efficacy of aromatase inhibitors caused by phytoestrogens. CONCLUSION: The outcomes of the studies we found suggest that caution should be exercised when taking phytoestrogen in combination with either tamoxifen or aromatase inhibitors. We believe that patients with breast cancer should be informed of this as a matter of routine.


Aromatase Inhibitors/adverse effects , Breast Neoplasms/therapy , Menopause/drug effects , Phytoestrogens/therapeutic use , Tamoxifen/adverse effects , Aromatase Inhibitors/therapeutic use , Dietary Supplements , Drug Interactions , Female , Humans , Tamoxifen/antagonists & inhibitors , Tamoxifen/therapeutic use , Treatment Outcome
13.
J Enzyme Inhib Med Chem ; 28(4): 734-40, 2013 Aug.
Article En | MEDLINE | ID: mdl-22524678

Apocynin (APO), curcumin (CUR) and vanillin (VAN) are o-methyl catechols widely studied due their antioxidant and antitumour properties. The effect of treatment with these o-methyl catechols on tamoxifen (TAM)-induced cytotoxicity in normal and tumour cells was studied. The cytotoxicity of TAM on red blood cells (RBC) was performed by haemoglobin or K(+)release and on polymorphonuclear leukocytes (PMNs) by trypan blue dye exclusion method. Cytotoxic activity was assessed in human chronic myeloid leukemia (K562) cell line by (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide). According the release of haemoglobin and K(+), the CUR showed a decrease in TAM cytotoxicity on RBC; however, in PMN, APO, CUR and VAN showed increased of these cells viability. VAN presented the highest cytotoxicity on K562 cells, followed by APO and CUR. These results point the potential therapeutic value of these o-methyl catechols with TAM, particularly of CUR, which potentiates the cytotoxic effects of TAM on K562 cells and also decreases TAM-associated cytotoxicity on RBC and PMN.


Acetophenones/pharmacology , Antineoplastic Agents/pharmacology , Benzaldehydes/pharmacology , Curcumin/pharmacology , Erythrocytes/drug effects , Neutrophils/drug effects , Tamoxifen/pharmacology , Acetophenones/chemistry , Antineoplastic Agents/chemistry , Benzaldehydes/chemistry , Cell Survival/drug effects , Curcumin/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , K562 Cells , Molecular Structure , Structure-Activity Relationship , Tamoxifen/antagonists & inhibitors , Tamoxifen/chemistry , Tumor Cells, Cultured
14.
Carcinogenesis ; 33(4): 895-901, 2012 Apr.
Article En | MEDLINE | ID: mdl-22266527

The present study examined the effect of dietary genistein, a soy isoflavone, on breast cancer patients who take tamoxifen, an antiestrogen treatment, using a preclinical model. The interaction of various doses of genistein with tamoxifen on the growth of estrogen receptor-positive breast cancer MCF-7 cells was investigated by subcutaneously injecting MCF-7 cells into the flank of ovariectomized athymic mice. Animals were randomized into eight experimental groups with 10-13 mice per group: control (C), estrogen (E) (0.08 mg E implant), tamoxifen (T) (3 mg T implant), estrogen + tamoxifen (E + T), tamoxifen + 500 p.p.m. genistein (T + G500), estrogen + tamoxifen + 250 p.p.m. genistein (E + T + G250), estrogen + tamoxifen + 500 p.p.m. genistein (E + T + G500) and estrogen + tamoxifen + 1000 p.p.m. genistein (E + T + G1000). Treatment of tamoxifen significantly reduced the estrogen-induced MCF-7 tumor prevalence and tumor size. This inhibitory effect of tamoxifen was significantly negated by the low doses of dietary genistein (250 and 500 p.p.m.), whereas the 1000 p.p.m. genistein did not have the same effect. Cells harvested from tamoxifen-treated tumors retained estrogen responsiveness of their progenitor MCF-7 cells, indicating that the abrogating effect of genistein on tamoxifen-treated tumor growth was not caused by a diminished tamoxifen response but directly by genistein. The low doses of dietary genistein abrogated the inhibitory effect of tamoxifen potentially by acting on the tumor cell proliferation/apoptosis ratio and the messenger RNA (mRNA) expression of cyclin D1 in addition to regulating the mRNA expression of progesterone receptor. Therefore, data from the current study suggest that caution is warranted regarding the consumption of dietary genistein by breast cancer patients while on tamoxifen therapy.


Antineoplastic Agents, Hormonal/antagonists & inhibitors , Diet , Genistein/pharmacology , Tamoxifen/antagonists & inhibitors , Animals , Apoptosis/drug effects , Base Sequence , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Primers , Dose-Response Relationship, Drug , Drug Interactions , Estrogens/blood , Genistein/administration & dosage , Mice , Mice, Nude
15.
Environ Toxicol Pharmacol ; 31(1): 143-52, 2011 Jan.
Article En | MEDLINE | ID: mdl-21787679

The genotoxic actions of anti-neoplastic drugs can lead to the development of secondary cancers in patients in extended remission. One of the most attractive approaches to disease prevention involves the use of natural antioxidants to protect tissue against toxic injury. We investigated the modulatory effects of exogenously administered taurine, on the genotoxicity of two well known anti-neoplastic drugs methotrexate (MTX) and tamoxifen (TAM) in Swiss albino mice. The animals were randomly divided into six groups consisting of ten mice each. Two groups were received single intraperitoneal injection of MTX (10 mg/kgb.wt.) and TAM (50 mg/kgb.wt.) to induce genotoxicity. Two other groups were treated orally with taurine (100 mg/kgb.wt.) for nine days prior to MTX and TAM administration. A vehicle treated control group and taurine control groups were also included. The protective effects of taurine were monitored by apoptosis assays and level of reduced glutathione (GSH), a key antioxidant, in liver, chromosomal aberrations in somatic and germ cells as well as sperm count, motility and morphology. The results indicated that taurine pre-treatment showed significant increment in the levels of GSH content, reduction in DNA fragmentation and ladder formation in hepatic tissue, suggesting the antioxidant activity of taurine may reduce the toxic effects of MTX and TAM. Treatment with taurine showed also significant reduction in the frequency of chromosomal aberrations in both somatic and germ cells. Moreover, it increases sperm count and motility, and decreases the incidence of sperm abnormalities. In conclusion, it appears that taurine protects against anti-neoplastic drugs-induced genotoxicity in somatic and germ tissues and may be of therapeutic potential in alleviating the risk of secondary tumors in chemotherapy.


Antimetabolites, Antineoplastic/toxicity , Antimutagenic Agents , Antineoplastic Agents, Hormonal/antagonists & inhibitors , Antineoplastic Agents, Hormonal/toxicity , Methotrexate/antagonists & inhibitors , Methotrexate/toxicity , Tamoxifen/antagonists & inhibitors , Tamoxifen/toxicity , Taurine/pharmacology , Androgens/pharmacology , Animals , Apoptosis/drug effects , Bone Marrow Cells/drug effects , Chromosome Aberrations/drug effects , DNA Damage , DNA Fragmentation/drug effects , Electrophoresis, Agar Gel , Germ Cells/drug effects , Glutathione/metabolism , Liver/cytology , Liver/drug effects , Liver/metabolism , Male , Mice , Sperm Motility/drug effects , Spermatocytes/drug effects , Sulfhydryl Compounds/metabolism
16.
Protein Pept Lett ; 18(3): 305-17, 2011 Mar.
Article En | MEDLINE | ID: mdl-21121897

The combination of several drugs is often necessary, especially during long-term therapy. A competitive binding of the drugs can cause a decrease of the amount of drugs actually bound to the protein and increase the biologically active fraction of the drug. The aim of this study has been to analyze the interactions of tamoxifen (TMX) and aspirin (ASA) with human serum albumin (HSA) and to evaluate the mechanism of a simultaneous binding of TMX and ASA to the protein. Fluorescence analysis was used to estimate the effect of the drugs on the protein fluorescence and to define the binding and quenching properties of drug-HSA complexes. The binding sites for TMX and ASA were identified in ternary structures of HSA by means of spectrofluroscence. The analysis of the fluorescence quenching of HSA in binary and ternary systems pointed at TMX and ASA having an effect on the HSA-ASA and HSA-TMX complexes. Furthermore, the results of synchronous fluorescence, resonance light scattering and circular dichroism of the binary and ternary systems showed that the binding of TMX and ASA to HSA could induce conformational changes in HSA. Moreover, the simultaneous presence of TMX and ASA during binding to HSA should be taken into account in multi-drug therapy, as it induces the necessity of a monitoring therapy owing to the possible increase of uncontrolled toxic effects. Competitive site marker experiments demonstrated that the binding site of ASA and TMX to HSA differed in the binary system as opposed to in its ternary counterpart. Finally, molecular modeling of the possible binding sites of TMX and ASA in binary and ternary systems to HSA confirmed the experimental results.


Aspirin/metabolism , Drug Antagonism , Models, Molecular , Serum Albumin/chemistry , Serum Albumin/metabolism , Spectrum Analysis , Tamoxifen/metabolism , Aspirin/antagonists & inhibitors , Aspirin/pharmacology , Binding Sites , Humans , Protein Binding , Protein Conformation/drug effects , Tamoxifen/antagonists & inhibitors , Tamoxifen/pharmacology
17.
J Pharmacol Sci ; 113(1): 66-75, 2010.
Article En | MEDLINE | ID: mdl-20472984

Tamoxifen inhibits transmembrane currents of the Kir2.x inward rectifier potassium channels by interfering with the interaction of the channels with membrane phosphatidylinositol 4,5-bisphosphate (PIP(2)). We tested the hypothesis that Kir channels with low affinity for PIP(2), like the adenosine triphosphate (ATP)-sensitive K(+) channel (K(ATP)) and acetylcholine (ACh)-activated K(+) channel (K(ACh)), have at least the same sensitivity to tamoxifen as Kir2.3. We investigated the effects of tamoxifen (0.1 - 10 microM) on Kir6.2/SUR2A (K(ATP)) and Kir3.1/3.4 (K(ACh)) channels expressed in HEK-293 cells and ATP-sensitive K(+) current (I(KATP)) and ACh-activated K(+) current (I(KACh)) in feline atrial myocytes. The onset of tamoxifen inhibition of both I(KATP) and I(KACh) was slow (T(1/2) approximately 3.5 min) and concentration-dependent but voltage-independent. The time course and degree of inhibition was independent of external or internal drug application. Tamoxifen interacts with the pore forming subunit, Kir6.2, rather than with the SUR subunit. The inhibitory potency of tamoxifen on the Kir6.2/SUR2A channel was decreased by the mutation (C166S) on Kir6.2 and in the continuous presence of PIP(2). In atrial myocytes, the mechanism and potency of the effects of tamoxifen on K(ATP) and K(ACh) channels were comparable to those in HEK-293 cells. These data suggest that, similar to its effects on Kir2.x currents, tamoxifen inhibits K(ATP) and K(ACh) currents by interfering with the interaction between the channel and PIP(2).


G Protein-Coupled Inwardly-Rectifying Potassium Channels/drug effects , KATP Channels/drug effects , Membrane Potentials/drug effects , Phosphatidylinositol Phosphates/pharmacology , Tamoxifen/pharmacology , Animals , Cats , Cell Line , Dose-Response Relationship, Drug , Drug Interactions , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , Humans , KATP Channels/genetics , Membrane Potentials/genetics , Mutagenesis, Site-Directed/methods , Myocytes, Cardiac/drug effects , Patch-Clamp Techniques , Phosphatidylinositol 4,5-Diphosphate , Tamoxifen/antagonists & inhibitors
18.
Thromb Res ; 125(4): 340-50, 2010 Apr.
Article En | MEDLINE | ID: mdl-20172594

Calmodulin (CaM) antagonists induce apoptosis in various tumor models and inhibit tumor cell invasion and metastasis, thus some of which have been extensively used as anti-cancer agents. In platelets, CaM has been found to bind directly to the cytoplasmic domains of several platelet receptors. Incubation of platelets with CaM antagonists impairs the receptors-related platelet functions. However, it is still unknown whether CaM antagonists induce platelet apoptosis. Here we show that CaM antagonists N-(6-aminohexyl)-5-chloro-1-naphthalene sulfonamide (W7), tamoxifen (TMX), and trifluoperazine (TFP) induce apoptotic events in human platelets, including depolarization of mitochondrial inner transmembrane potential, caspase-3 activation, and phosphatidylserine exposure. CaM antagonists did not incur platelet activation as detected by P-selectin surface expression and PAC-1 binding. However, ADP-, botrocetin-, and alpha-thrombin-induced platelet aggregation, platelet adhesion and spreading on von Willebrand factor surface were significantly reduced in platelets pre-treated with CaM antagonists. Furthermore, cytosolic Ca(2+) levels were obviously elevated by both W7 and TMX, and membrane-permeable Ca(2+) chelator BAPTA-AM significantly reduced apoptotic events in platelets induced by W7. Therefore, these findings indicate that CaM antagonists induce platelet apoptosis. The elevation of the cytosolic Ca(2+) levels may be involved in the regulation of CaM antagonists-induced platelet apoptosis.


Apoptosis/drug effects , Blood Platelets/metabolism , Calmodulin/antagonists & inhibitors , Caspase 3/metabolism , Thrombin/metabolism , Apoptosis/physiology , Blood Platelets/cytology , Calmodulin/metabolism , Calmodulin/physiology , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Carrier Proteins/pharmacology , Caspase Inhibitors , Egtazic Acid/analogs & derivatives , Humans , Membrane Potentials/drug effects , Mitochondria/metabolism , P-Selectin/metabolism , P-Selectin/pharmacology , Phosphatidylserines/antagonists & inhibitors , Phosphatidylserines/metabolism , Phosphatidylserines/pharmacology , Platelet Activation/drug effects , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/metabolism , Platelet Aggregation Inhibitors/pharmacology , Tamoxifen/antagonists & inhibitors , Tamoxifen/metabolism , Tamoxifen/pharmacology , Thrombin/antagonists & inhibitors , Thrombin/pharmacology , Trifluoperazine/antagonists & inhibitors , Trifluoperazine/metabolism , Trifluoperazine/pharmacology , von Willebrand Factor/antagonists & inhibitors , von Willebrand Factor/metabolism , von Willebrand Factor/pharmacology
20.
Food Chem Toxicol ; 47(7): 1689-95, 2009 Jul.
Article En | MEDLINE | ID: mdl-19394397

Tamoxifen (TAM) is widely used in the treatment and prevention of breast cancer. Adverse effects of TAM include hepatotoxicity. Caffeic acid phenethyl ester (CAPE), an active component of propolis, has been used in folk medicine for diverse ailments. In the current study, the protective effects of CAPE against TAM-induced hepatotoxicity in female rats were evaluated. TAM (45 mg/kg/day, i.p., for 10 consecutive days) resulted in an elevation of serum alanine aminotransferase (ALT), aspartate aminotransferase (AST) and alkaline phosphatase (ALP), depletion of liver reduced glutathione (GSH) and accumulation of oxidized glutathione (GSSG) and lipid peroxidation (LPO). Also, TAM treatment resulted in inhibition of hepatic activity of glutathione reductase (GR), glutathione peroxidase (GPx), superoxide dismutase (SOD) and catalase (CAT). Further, it raised liver tumor necrosis factor-alpha (TNF-alpha) level and induced histopathological changes. Pretreatment with CAPE (2.84 mg/kg/day; i.p., for 20 consecutive days, starting 10 days before TAM injection) significantly prevented the elevation in serum activity of the assessed enzymes. CAPE significantly inhibited TAM-induced hepatic GSH depletion and GSSG and LPO accumulation. Consistently, CAPE normalized the activity of GR, GPx, SOD and CAT, inhibited the rise in TNF-alpha and ameliorated the histopathological changes. In conclusion, CAPE protects against TAM-induced hepatotoxicity.


Antineoplastic Agents, Hormonal/antagonists & inhibitors , Antineoplastic Agents, Hormonal/toxicity , Caffeic Acids/pharmacology , Chemical and Drug Induced Liver Injury/prevention & control , Phenylethyl Alcohol/analogs & derivatives , Tamoxifen/antagonists & inhibitors , Tamoxifen/toxicity , Animals , Antioxidants/metabolism , Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/pathology , Female , Glutathione/metabolism , Lipid Peroxidation/drug effects , Liver/enzymology , Liver/pathology , Liver Function Tests , Neutrophils/drug effects , Neutrophils/enzymology , Phenylethyl Alcohol/pharmacology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species , Tumor Necrosis Factor-alpha/metabolism , Xanthine Oxidase/metabolism
...