Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters











Publication year range
1.
Commun Biol ; 7(1): 1086, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39232115

ABSTRACT

Cell-free DNA (cfDNA) has emerged as a pivotal player in precision medicine, revolutionizing the diagnostic and therapeutic landscape. While its clinical applications have significantly increased in recent years, current cfDNA assays have limited ability to identify the active transcriptional programs that govern complex disease phenotypes and capture the heterogeneity of the disease. To address these limitations, we have developed a non-invasive platform to enrich and examine the active chromatin fragments (cfDNAac) in peripheral blood. The deconvolution of the cfDNAac signal from traditional nucleosomal chromatin fragments (cfDNAnuc) yields a catalog of features linking these circulating chromatin signals in blood to specific regulatory elements across the genome, including enhancers, promoters, and highly transcribed genes, mirroring the epigenetic data from the ENCODE project. Notably, these cfDNAac counts correlate strongly with RNA polymerase II activity and exhibit distinct expression patterns for known circadian genes. Additionally, cfDNAac signals across gene bodies and promoters show strong correlations with whole blood gene expression levels defined by GTEx. This study illustrates the utility of cfDNAac analysis for investigating epigenomics and gene expression, underscoring its potential for a wide range of clinical applications in precision medicine.


Subject(s)
Cell-Free Nucleic Acids , Chromatin , Chromatin/genetics , Chromatin/metabolism , Humans , Cell-Free Nucleic Acids/blood , Cell-Free Nucleic Acids/genetics , Promoter Regions, Genetic , Epigenesis, Genetic , Epigenomics/methods , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Nucleosomes/metabolism , Nucleosomes/genetics
2.
Front Oncol ; 10: 1703, 2020.
Article in English | MEDLINE | ID: mdl-33224868

ABSTRACT

Cancer growth is predicted to require substantial rates of substrate catabolism and ATP turnover to drive unrestricted biosynthesis and cell growth. While substrate limitation can dramatically alter cell behavior, the effects of substrate limitation on total cellular ATP production rate is poorly understood. Here, we show that MCF7 breast cancer cells, given different combinations of the common cell culture substrates glucose, glutamine, and pyruvate, display ATP production rates 1.6-fold higher than when cells are limited to each individual substrate. This increase occurred mainly through faster oxidative ATP production, with little to no increase in glycolytic ATP production. In comparison, non-transformed C2C12 myoblast cells show no change in ATP production rate when substrates are limited. In MCF7 cells, glutamine allows unexpected access to oxidative capacity that pyruvate, also a strictly oxidized substrate, does not. Pyruvate, when added with other exogenous substrates, increases substrate-driven oxidative ATP production, by increasing both ATP supply and demand. Overall, we find that MCF7 cells are highly flexible with respect to maintaining total cellular ATP production under different substrate-limited conditions, over an acute (within minutes) timeframe that is unlikely to result from more protracted (hours or more) transcription-driven changes to metabolic enzyme expression. The near-identical ATP production rates maintained by MCF7 and C2C12 cells given single substrates reveal a potential difficulty in using substrate limitation to selectively starve cancer cells of ATP. In contrast, the higher ATP production rate conferred by mixed substrates in MCF7 cells remains a potentially exploitable difference.

3.
Sci Rep ; 9(1): 12135, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31431634

ABSTRACT

Cadmium is an environmental contaminant that can activate estrogen receptor alpha (ERα) and contribute to the development and progression of breast cancer. Our lab previously demonstrated that chronic cadmium exposure alters the expression of several ERα-responsive genes and increases the malignancy of breast cancer cells. Although these studies support cadmium's function as a hormone disrupter, the role of ERα in cadmium-induced breast cancer progression remains unclear. To address this, we modulated the expression of ERα and found that while the loss of ERα significantly impaired cancer cell growth, migration, invasion and anchorage-independent growth in both MCF7 and MCF7-Cd cells, cadmium-exposed cells retained a significant advantage in cell growth, migration, and invasion, and partially circumvented the loss of ERα. ERα knockout in MCF7 and MCF7-Cd cells significantly reduced the expression of classical ERα-regulated genes, while non-classical ERα-regulated genes were less impacted by the loss of ERα in MCF7-Cd cells. This is the first study to show that chronic cadmium exposure, even at low levels, can increase the malignancy of breast cancer cells by decreasing their dependency on ERα and increasing the adaptability of the cancer cells.


Subject(s)
Adenocarcinoma/metabolism , Breast Neoplasms/metabolism , Cadmium Chloride/adverse effects , Estrogen Receptor alpha/metabolism , Adenocarcinoma/genetics , Breast Neoplasms/genetics , CRISPR-Cas Systems , Cell Movement/physiology , Cell Proliferation/physiology , Estrogen Receptor alpha/genetics , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Humans , MCF-7 Cells , Neoplasm Invasiveness/physiopathology
4.
Clin Cancer Res ; 25(14): 4255-4263, 2019 07 15.
Article in English | MEDLINE | ID: mdl-30992300

ABSTRACT

PURPOSE: Up to 30% of patients with breast cancer relapse after primary treatment. There are no sensitive and reliable tests to monitor these patients and detect distant metastases before overt recurrence. Here, we demonstrate the use of personalized circulating tumor DNA (ctDNA) profiling for detection of recurrence in breast cancer. EXPERIMENTAL DESIGN: Forty-nine primary patients with breast cancer were recruited following surgery and adjuvant therapy. Plasma samples (n = 208) were collected every 6 months for up to 4 years. Personalized assays targeting 16 variants selected from primary tumor whole-exome data were tested in serial plasma for the presence of ctDNA by ultradeep sequencing (average >100,000X). RESULTS: Plasma ctDNA was detected ahead of clinical or radiologic relapse in 16 of the 18 relapsed patients (sensitivity of 89%); metastatic relapse was predicted with a lead time of up to 2 years (median, 8.9 months; range, 0.5-24.0 months). None of the 31 nonrelapsing patients were ctDNA-positive at any time point across 156 plasma samples (specificity of 100%). Of the two relapsed patients who were not detected in the study, the first had only a local recurrence, whereas the second patient had bone recurrence and had completed chemotherapy just 13 days prior to blood sampling. CONCLUSIONS: This study demonstrates that patient-specific ctDNA analysis can be a sensitive and specific approach for disease surveillance for patients with breast cancer. More importantly, earlier detection of up to 2 years provides a possible window for therapeutic intervention.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/diagnosis , Circulating Tumor DNA/genetics , High-Throughput Nucleotide Sequencing/methods , Mutation , Neoplasm Recurrence, Local/diagnosis , Precision Medicine , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/blood , Breast Neoplasms/blood , Breast Neoplasms/genetics , Breast Neoplasms/secondary , Circulating Tumor DNA/blood , Female , Humans , Middle Aged , Neoplasm Metastasis , Neoplasm Recurrence, Local/blood , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Prognosis , Prospective Studies
5.
Cancer Lett ; 443: 157-166, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30503556

ABSTRACT

SL-1-39 [1-(4-chloro-3-methylphenyl)-3-(4-nitrophenyl)thiourea] is a new flexible heteroarotinoid (Flex-Het) analog derived from the parental compound, SHetA2, previously shown to inhibit cell growth across multiple cancer types. The current study aims to determine growth inhibitory effects of SL-1-39 across the different subtypes of breast cancer cells and delineate its molecular mechanism. Our results demonstrate that while SL-1-39 blocks cell proliferation of all breast cancer subtypes tested, it has the highest efficacy against HER2+ breast cancer cells. Molecular analyses suggest that SL-1-39 prevents S phase progression of HER2+ breast cancer cells (SKBR3 and MDA-MB-453), which is consistent with reduced expression of key cell-cycle regulators at both the protein and transcriptional levels. SL-1-39 treatment also decreases the protein levels of HER2 and pHER2 as well as its downstream effectors, pMAPK and pAKT. Reduction of HER2 and pHER2 at the protein level is attributed to increased lysosomal degradation of total HER2 levels. This is the first study to show that a flexible heteroarotinoid analog modulates the HER2 signaling pathway through lysosomal degradation, and thus further warrants the development of SL-1-39 as a therapeutic option for HER2+ breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Chromans/chemical synthesis , Lysosomes/metabolism , Receptor, ErbB-2/chemistry , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Catechols/chemistry , Catechols/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Chromans/chemistry , Chromans/pharmacology , Drug Screening Assays, Antitumor , Female , Humans , MCF-7 Cells , Proteolysis , Receptor, ErbB-2/genetics , Thiones/chemistry
6.
Am J Cancer Res ; 7(8): 1617-1636, 2017.
Article in English | MEDLINE | ID: mdl-28861319

ABSTRACT

Despite the existence of many promising anti-cancer therapies, not all breast cancers are equally treatable, due partly to the fact that focus has been primarily on a few select breast cancer biomarkers- notably ERα, PR and HER2. In cases like triple negative breast cancer (ERα-, PR-, and HER2-), there is a complete lack of available biomarkers for prognosis and therapeutic purposes. The goal of this review is to determine if other steroid receptors, like ERß and AR, could play a prognostic and/or therapeutic role. Data from various in vitro, in vivo, and clinical breast cancer studies were examined to analyze the presence and function of ERß, PR, and AR in the presence and absence of ERα. Additionally, we focused on studies that examined how expression of the various steroid receptor isoforms affects breast cancer progression. Our findings suggest that while we have a solid understanding of how these receptors work individually, how they interact and behave in the presence and absence of other receptors requires further research. Furthermore, there is an incomplete understanding of how the various steroid receptor isoforms interact and impact receptor function and breast cancer progression, partly due to the difficulty in detecting all the various isoforms. More large-scale clinical studies must be made to analyze systematically the expression of steroid hormone receptors and their respective isoforms in breast cancer patients in order to determine how these receptors interact with each other and in turn affect cancer progression.

7.
Cancer Lett ; 408: 82-91, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28844711

ABSTRACT

SL-1-18 (1-(chrysen-6-yl)-3-(4-nitrophenyl)thiourea) is new flexible heteroarotinoid (Flex-Het) analog derived from the parent compound, SHetA2, and our previous study showed comparable activity to SHetA2 in terms of inhibiting ER+ breast cancer cell growth. This current study aims to determine the molecular mechanism underlying SL-1-18's effect on breast cancer cell growth. Our results indicate that SL-1-18 inhibits cell proliferation of ER+ breast cancer cells (MCF-7 and T-47D) by preventing cell cycle progression. SL-1-18 treatment correlated positively with decreased expression of key cell-cycle regulators, such as cyclin D1, as well as other ERα-target genes at both the transcript and protein levels. Interestingly, decreased expression of ERα was also observed, with a significant reduction at the protein level within 2 h of SL-1-18 treatment, while the decrease in mRNA occurred at a later time point. ERα degradation was shown to be mediated by the ubiquitination-proteasome pathway. In summary, this is the first study to show that a Flex-Het- SL-1-18- can promote the degradation of ERα via the ubiquitin-proteasome pathway and should be further developed as a therapeutic option for ER+ breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Chromans/pharmacology , Chrysenes/pharmacology , Estrogen Receptor alpha/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Proteolysis/drug effects , Thiourea/analogs & derivatives , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Proliferation/drug effects , Female , Humans , Proteasome Endopeptidase Complex , Thiones/pharmacology , Thiourea/pharmacology , Tumor Cells, Cultured , Ubiquitination
9.
Nat Med ; 22(5): 488-96, 2016 05.
Article in English | MEDLINE | ID: mdl-27019329

ABSTRACT

The androgen receptor (AR) is overexpressed and hyperactivated in human castration-resistant prostate cancer (CRPC). However, the determinants of AR overexpression in CRPC are poorly defined. Here we show that retinoic acid receptor-related orphan receptor γ (ROR-γ) is overexpressed and amplified in metastatic CRPC tumors, and that ROR-γ drives AR expression in the tumors. ROR-γ recruits nuclear receptor coactivator 1 and 3 (NCOA1 and NCOA3, also known as SRC-1 and SRC-3) to an AR-ROR response element (RORE) to stimulate AR gene transcription. ROR-γ antagonists suppress the expression of both AR and its variant AR-V7 in prostate cancer (PCa) cell lines and tumors. ROR-γ antagonists also markedly diminish genome-wide AR binding, H3K27ac abundance and expression of the AR target gene network. Finally, ROR-γ antagonists suppressed tumor growth in multiple AR-expressing, but not AR-negative, xenograft PCa models, and they effectively sensitized CRPC tumors to enzalutamide, without overt toxicity, in mice. Taken together, these results establish ROR-γ as a key player in CRPC by acting upstream of AR and as a potential therapeutic target for advanced PCa.


Subject(s)
Gene Expression Regulation, Neoplastic , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Prostatic Neoplasms, Castration-Resistant/genetics , Receptors, Androgen/genetics , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzamides , Cell Survival/drug effects , Databases, Factual , Gene Knockdown Techniques , Glucose-6-Phosphate Isomerase , Humans , Immunoblotting , Immunohistochemistry , Male , Mice , Neoplasm Transplantation , Nitriles , Nuclear Receptor Coactivator 1/metabolism , Nuclear Receptor Coactivator 3/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Piperazines/pharmacology , Propanols/pharmacology , Prostatic Neoplasms, Castration-Resistant/metabolism , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Androgen/metabolism , Response Elements , Tumor Stem Cell Assay
10.
Bioorg Med Chem Lett ; 25(6): 1301-5, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25701251

ABSTRACT

Ten p-nitrodiarylthiourea analogs were designed, synthesized and evaluated in breast (MCF-7, T-47D, MDA-MB-453) and prostate (DU-145, PC-3, LNCaP) cancer cell lines for their anticancer activities. The majority of the compounds were able to inhibit the growth of these six cancer cell lines at low micromolar concentrations. Compound 7 was found to be the most potent anticancer agent in this series with GI50 values of 3.16µM for MCF-7, 2.53µM for T-47D, 4.77µM for MDA-MB-453 breast cancer lines and 3.54µM for LNCaP prostate cancer cell line. These GI50 values were comparable to the parent compound, SHetA2.


Subject(s)
Antineoplastic Agents/chemical synthesis , Thiourea/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , MCF-7 Cells , Structure-Activity Relationship , Thiourea/chemical synthesis , Thiourea/pharmacology
11.
Mol Carcinog ; 54(10): 1014-25, 2015 Oct.
Article in English | MEDLINE | ID: mdl-24798214

ABSTRACT

Cadmium is an environmental carcinogen that usually enters the body at minute concentrations through diet or cigarette smoke and bioaccumulates in soft tissues. In past studies, cadmium has been shown to contribute to the development of more aggressive cancer phenotypes including increased cell migration and invasion. This study aims to determine if cadmium exposure-both acute and chronic-contributes to breast cancer progression by interfering with the normal functional relationship between E-cadherin and ß-catenin. An MCF7 breast cancer cell line (MCF7-Cd) chronically exposed to 10(-7) M CdCl2 was previously developed and used as a model system to study chronic exposures, whereas parental MCF7 cells exposed to 10(-6) M CdCl2 for short periods of time were used to study acute exposures. Cadmium exposure of MCF7 cells led to the degradation of the E-cadherin protein via the ubiquitination pathway. This resulted in fewer E-cadherin/ß-catenin complexes and the relocation of active ß-catenin to the nucleus, where it interacted with transcription factor TCF-4 to modulate gene expression. Interestingly, only cells chronically exposed to cadmium showed a significant decrease in the localization of ß-catenin to the plasma membrane and an increased distance between cells. Our data suggest that cadmium exposure promotes breast cancer progression by (1) down-regulating E-cadherin, thus decreasing the number of E-cadherin/ß-catenin adhesion complexes, and (2) enhancing the nuclear translocation of ß-catenin to increase expression of cancer-promoting proteins (i.e., c-Jun and cyclin D1).


Subject(s)
Breast Neoplasms/metabolism , Cadherins/metabolism , Cadmium/adverse effects , Proteolysis/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Disease Progression , Down-Regulation/drug effects , Female , Gene Expression/drug effects , Humans , MCF-7 Cells , Transcription Factor 4 , Transcription Factors/metabolism , Ubiquitination/drug effects , beta Catenin/metabolism
12.
Am J Pharm Educ ; 78(10): 180, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25657367

ABSTRACT

OBJECTIVE: To identify differences among faculty members in various health professional training programs in perceived benefits and challenges of implementing interprofessional education (IPE). METHODS: A 19-item survey using a 5-point Likert scale was administered to faculty members across different health disciplines at a west coast, multicollege university with osteopathic medicine, pharmacy, and physician assistant programs. RESULTS: Sixty-two of 103 surveys (60.2%) were included in the study. Faculty members generally agreed that there were benefits of IPE on patient outcomes and that implementing IPE was feasible. However, group differences existed in belief that IPE improves care efficiency (p=0.001) and promotes team-based learning (p=0.001). Program divergence was also seen in frequency of stressing importance of IPE (p=0.009), preference for more IPE opportunities (p=0.041), and support (p=0.002) within respective college for IPE. CONCLUSIONS: Despite consensus among faculty members from 3 disciplines that IPE is invaluable to their curricula and training of health care students, important program level differences existed that would likely need to be addressed in advance IPE initiatives.


Subject(s)
Education, Pharmacy/organization & administration , Faculty/statistics & numerical data , Osteopathic Medicine/education , Physician Assistants/education , Adult , Attitude of Health Personnel , Curriculum , Data Collection , Female , Humans , Interprofessional Relations , Male , Middle Aged , Universities
13.
PLoS One ; 8(12): e84646, 2013.
Article in English | MEDLINE | ID: mdl-24376830

ABSTRACT

Cadmium is a metalloestrogen known to activate the estrogen receptor and promote breast cancer cell growth. Previous studies have implicated cadmium in the development of more malignant tumors; however the molecular mechanisms behind this cadmium-induced malignancy remain elusive. Using clonal cell lines derived from exposing breast cancer cells to cadmium for over 6 months (MCF-7-Cd4, -Cd6, -Cd7, -Cd8 and -Cd12), this study aims to identify gene expression signatures associated with chronic cadmium exposure. Our results demonstrate that prolonged cadmium exposure does not merely result in the deregulation of genes but actually leads to a distinctive expression profile. The genes deregulated in cadmium-exposed cells are involved in multiple biological processes (i.e. cell growth, apoptosis, etc.) and molecular functions (i.e. cadmium/metal ion binding, transcription factor activity, etc.). Hierarchical clustering demonstrates that the five clonal cadmium cell lines share a common gene expression signature of breast cancer associated genes, clearly differentiating control cells from cadmium exposed cells. The results presented in this study offer insights into the cellular and molecular impacts of cadmium on breast cancer and emphasize the importance of studying chronic cadmium exposure as one possible mechanism of promoting breast cancer progression.


Subject(s)
Cadmium/toxicity , Gene Expression Regulation, Neoplastic/drug effects , MCF-7 Cells/drug effects , Cluster Analysis , DNA Primers/genetics , Female , Gene Expression Profiling/methods , Humans , Microarray Analysis , Reverse Transcriptase Polymerase Chain Reaction
14.
PLoS One ; 8(8): e72639, 2013.
Article in English | MEDLINE | ID: mdl-24015267

ABSTRACT

Cadmium is an omnipotent environmental contaminant associated with the development of breast cancer. Studies suggest that cadmium functions as an endocrine disruptor, mimicking the actions of estrogen in breast cancer cells and activating the receptor to promote cell growth. Although acute cadmium exposure is known to promote estrogen receptor-mediated gene expression associated with growth, the consequence of chronic cadmium exposure is unclear. Since heavy metals are known to bioaccumulate, it is necessary to understand the effects of prolonged cadmium exposure. This study aims to investigate the effects of chronic cadmium exposure on breast cancer progression. A MCF7 breast cancer cell line chronically exposed to 10(-7) M CdCl2 serves as our model system. Data suggest that prolonged cadmium exposures result in the development of more aggressive cancer phenotypes - increased cell growth, migration and invasion. The results from this study show for the first time that chronic cadmium exposure stimulates the expression of SDF-1 by altering the molecular interactions between ERα, c-jun and c-fos. This study provides a mechanistic link between chronic cadmium exposure and ERα and demonstrates that prolonged, low-level cadmium exposure contributes to breast cancer progression.


Subject(s)
Breast Neoplasms/metabolism , Cadmium/toxicity , Chemokine CXCL12/biosynthesis , Estrogen Receptor alpha/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Female , Humans , Neoplasm Invasiveness , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism
15.
Article in English | MEDLINE | ID: mdl-22970719

ABSTRACT

During the past half-century, incidences of breast cancer have increased globally. Various factors--genetic and environmental--have been implicated in the initiation and progression of this disease. One potential environmental risk factor that has not received a lot of attention is the exposure to heavy metals. While several mechanisms have been put forth describing how high concentrations of heavy metals play a role in carcinogenesis, it is unclear whether chronic, low-level exposure to certain heavy metals (i.e., cadmium and nickel) can directly result in the development and progression of cancer. Cadmium and nickel have been hypothesized to play a role in breast cancer development by acting as metalloestrogens--metals that bind to estrogen receptors and mimic the actions of estrogen. Since the lifetime exposure to estrogen is a well-established risk factor for breast cancer, anything that mimics its activity will likely contribute to the etiology of the disease. However, heavy metals, depending on their concentration, are capable of binding to a variety of proteins and may exert their toxicities by disrupting multiple cellular functions, complicating the analysis of whether heavy metal-induced carcinogenesis is mediated by the estrogen receptor. The purpose of this review is to discuss the various epidemiological, in vivo, and in vitro studies that show a link between the heavy metals, cadmium and nickel, and breast cancer development. We will particularly focus on the studies that test whether these two metals act as metalloestrogens in order to assess the strength of the data supporting this hypothesis.


Subject(s)
Breast Neoplasms/metabolism , Cadmium/toxicity , Nickel/toxicity , Receptors, Estrogen/metabolism , Signal Transduction/drug effects , Breast Neoplasms/etiology , Cadmium/chemistry , Chromatin/drug effects , Environmental Exposure , Female , Humans , Nickel/chemistry , Receptors, Estrogen/chemistry , Risk Factors
16.
Pharmacol Res ; 65(4): 445-50, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22245433

ABSTRACT

Prostanoids play an important role in a variety of physiological and pathophysiological processes including inflammation and cancer. The rate-limiting step in the prostanoid biosynthesis pathway is catalyzed by cyclooxygenase-2 (COX-2). COX-2 exists as two glycoforms, 72 and 74 kDa, the latter resulting from an additional glycosylation at Asn(580). In this study, Asn(580) was mutated, and the mutant and wild-type COX-2 genes were expressed in COS-1 cells to determine how glycosylation affects the inhibition of COX-2 activity by aspirin, flurbiprofen, ibuprofen, celecoxib, and etoricoxib. Results indicate that certain inhibitors were 2-5 times more effective at inhibiting COX-2 activity when the glycosylation site was eliminated, indicating that glycosylation of COX-2 at Asn(580) decreases the efficacy of some inhibitors.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2/metabolism , Animals , COS Cells , Chlorocebus aethiops , Cyclooxygenase 2/genetics , Glycosylation , Humans , Transfection
17.
Mol Endocrinol ; 24(5): 981-92, 2010 May.
Article in English | MEDLINE | ID: mdl-20219890

ABSTRACT

Cadmium is an environmental contaminant that enters the body through diet or cigarette smoke. It affects multiple cellular processes, including cell proliferation, differentiation, and apoptosis. Recently, cadmium has been shown to function as an endocrine disruptor, to stimulate estrogen receptor alpha (ERalpha) activity and promote uterine and mammary gland growth in mice. Although cadmium exposure has been associated with the development of breast cancer, the mechanism of action of cadmium remains unclear. To address this deficit, we examined the effects of cadmium treatment on breast cancer cells. We found that ERalpha is required for both cadmium-induced cell growth and modulation of gene expression. We also determined that ERalpha translocates to the nucleus in response to cadmium exposure. Additionally, we provide evidence that cadmium potentiates the interaction between ERalpha and c-Jun and enhances recruitment of this transcription factor complex to the proximal promoters of cyclin D1 and c-myc, thus increasing their expression. This study provides a mechanistic link between cadmium exposure and ERalpha and demonstrates that cadmium plays an important role in the promotion of breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Cadmium/pharmacology , Cell Proliferation/drug effects , Estrogen Receptor alpha/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Blotting, Western , Breast Neoplasms/chemically induced , Cell Line, Tumor , Chromatin Immunoprecipitation , Cyclin D1/metabolism , Female , Humans , Immunoprecipitation , Protein Binding/drug effects , Reverse Transcriptase Polymerase Chain Reaction
18.
Mol Cancer Res ; 8(3): 343-52, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20215421

ABSTRACT

Antiestrogen resistance often develops with prolonged exposure to hormone therapies, including tamoxifen, and is a major problem in the treatment of breast cancer. Understanding the mechanisms involved in the development of antiestrogen resistance is an important step in the development of new targeted therapies. Two forms of antiestrogen resistance exist: de novo resistance and acquired resistance. To mimic acquired resistance, we have established a tamoxifen-resistant breast cancer cell line (MCF-7TamR) by treating parental MCF-7 cells with tamoxifen over a period of 6 months to select for cells with the resistant phenotype. Characterization of the MCF-7TamR cells under normal, hormone-deprived, and tamoxifen-treated conditions suggests that these cells continue to grow in the presence of tamoxifen. Additionally, a greater percentage of resistant cells enter the S phase under tamoxifen conditions, compared with parental MCF-7 cells. Consistent with these growth results, molecular analysis indicates that tamoxifen-resistant cells express higher levels of cyclin E1, cdk2, ACTR, and E2F1. Faslodex or ICI 182, 780 (ICI)-mediated degradation of estrogen receptor (ER) reduced the proliferation of these cells, as well as the level of E2F1 expression in tamoxifen-resistant cells, suggesting that tamoxifen resistance and E2F1 expression are in part dependent on ER. We further showed that tamoxifen enhances the ERalpha/Sp-1 interaction and promotes the recruitment of ERalpha and Sp-1 to the proximal promoter of E2F1 in resistant cells. Collectively, our findings suggest that tamoxifen resistance is a result of increased ERalpha/Sp-1 interaction, which enhances the expression of E2F1 to promote tamoxifen resistance.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , E2F1 Transcription Factor/genetics , Estrogen Receptor alpha/genetics , Gene Expression Regulation, Neoplastic/genetics , Tamoxifen/pharmacology , Breast Neoplasms/metabolism , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , E2F1 Transcription Factor/drug effects , E2F1 Transcription Factor/metabolism , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Receptor alpha/drug effects , Estrogen Receptor alpha/metabolism , Fulvestrant , Genes, cdc/drug effects , Genes, cdc/physiology , Humans , Immunoglobulins/drug effects , Immunoglobulins/metabolism , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , S Phase/drug effects , S Phase/physiology , Selective Estrogen Receptor Modulators/pharmacology
19.
Cancer Lett ; 261(1): 64-73, 2008 Mar 08.
Article in English | MEDLINE | ID: mdl-18162290

ABSTRACT

Overexpression of ACTR/AIB1 is frequently found in different cancers with distant metastasis. To address its possible involvement in tumor metastasis, we performed invasion assays to examine the effect of ACTR alteration on the invasiveness of breast cancer cells (MDA-MB-231 or T-47D) and found that high levels of ACTR are required for their strong invasiveness. Molecular analysis indicates that ACTR functions as a coactivator of AP-1 to up-regulate the expression of matrix metalloproteinases such as MMP-7 and MMP-10 and reduce cell adhesion to specific extracellular matrix proteins. These novel findings provide a mechanistic link between ACTR and MMPs, and suggest that ACTR may also play an important role in cancer progression by facilitating tumor invasion.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Matrix Metalloproteinase 10/metabolism , Matrix Metalloproteinase 7/genetics , Matrix Metalloproteinases/metabolism , Neoplasm Invasiveness , Proto-Oncogenes , Transcription Factors/genetics , Breast Neoplasms/pathology , Female , Gene Expression , Humans , Nuclear Receptor Coactivator 3 , Proto-Oncogene Mas , Up-Regulation
20.
Mol Cell Biol ; 26(10): 3810-23, 2006 May.
Article in English | MEDLINE | ID: mdl-16648476

ABSTRACT

ACTR (also called AIB1 and SRC-3) was identified as a coactivator for nuclear receptors and is linked to multiple types of human cancer due to its frequent overexpression. However, the molecular mechanism of ACTR oncogenicity and its function independent of nuclear receptors remain to be defined. We demonstrate here that ACTR is required for both normal and malignant human cells to effectively enter S phase. RNA interference-mediated depletion and chromatin immunoprecipitation assays show that endogenous ACTR directly controls the expression of genes important for initiation of DNA replication, which include cdc6, cdc25A, MCM7, cyclin E, and Cdk2. Moreover, consistent with its critical role in cell cycle control, ACTR expression appears to be cell cycle regulated, which involves E2F. Interestingly, ACTR is recruited to its own promoter at the G1/S transition and activates its own expression, suggesting a positive feedback mechanism for ACTR action in the control of cell cycle progression and for its aberrant expression in cancers. Importantly, overexpression of ACTR alone transforms human mammary epithelial cells, which requires its association with E2F. These findings reveal a novel role for ACTR in cell cycle control and support the notion that the ability of aberrant ACTR to deregulate the cell cycle through E2F underlies its oncogenicity in human cancers.


Subject(s)
Acetyltransferases/metabolism , Cell Cycle , Gene Expression Regulation, Neoplastic , Gene Expression Regulation , Genes, cdc , Oncogene Proteins/metabolism , Trans-Activators/metabolism , Acetyltransferases/genetics , Adenoviridae/genetics , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle Proteins/antagonists & inhibitors , Cell Line , Cell Line, Tumor , Cell Proliferation , Chromatin Immunoprecipitation , Diploidy , Female , Fibroblasts/metabolism , Fluorescein-5-isothiocyanate , Fluorescent Dyes , Genes, Reporter , Glioblastoma/genetics , Glioblastoma/pathology , Green Fluorescent Proteins/metabolism , HeLa Cells , Histone Acetyltransferases , Humans , Indoles , Luciferases/metabolism , Microscopy, Fluorescence , Nuclear Receptor Coactivator 3 , Oncogene Proteins/genetics , Promoter Regions, Genetic , Proto-Oncogene Mas , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/genetics , Tumor Stem Cell Assay
SELECTION OF CITATIONS
SEARCH DETAIL