Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Mol Cell ; 65(6): 1122-1135.e5, 2017 Mar 16.
Article in English | MEDLINE | ID: mdl-28306507

ABSTRACT

Human breast cancers that exhibit high proportions of immune cells and elevated levels of pro-inflammatory cytokines predict poor prognosis. Here, we demonstrate that treatment of human MCF-7 breast cancer cells with pro-inflammatory cytokines results in ERα-dependent activation of gene expression and proliferation, in the absence of ligand or presence of 4OH-tamoxifen (TOT). Cytokine activation of ERα and endocrine resistance is dependent on phosphorylation of ERα at S305 in the hinge domain. Phosphorylation of S305 by IKKß establishes an ERα cistrome that substantially overlaps with the estradiol (E2)-dependent ERα cistrome. Structural analyses suggest that S305-P forms a charge-linked bridge with the C-terminal F domain of ERα that enables inter-domain communication and constitutive activity from the N-terminal coactivator-binding site, revealing the structural basis of endocrine resistance. ERα therefore functions as a transcriptional effector of cytokine-induced IKKß signaling, suggesting a mechanism through which the tumor microenvironment controls tumor progression and endocrine resistance.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/drug therapy , Cytokines/metabolism , Drug Resistance, Neoplasm , Estrogen Receptor alpha/drug effects , Inflammation Mediators/metabolism , Neoplasms, Hormone-Dependent/drug therapy , Selective Estrogen Receptor Modulators/pharmacology , Tamoxifen/analogs & derivatives , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/genetics , Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation, Neoplastic , HeLa Cells , Hep G2 Cells , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Interleukin-1beta/metabolism , MCF-7 Cells , Molecular Dynamics Simulation , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Phosphorylation , Protein Conformation , RNA Interference , Signal Transduction/drug effects , Structure-Activity Relationship , Tamoxifen/pharmacology , Transcription, Genetic , Transfection , Tumor Microenvironment , Tumor Necrosis Factor-alpha/metabolism
2.
Breast Cancer Res Treat ; 201(3): 561-570, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37477798

ABSTRACT

PURPOSE: The oncogenic factor ZNF217 promotes aggressive estrogen receptor (ER)+breast cancer disease suggesting that its inhibition may be useful in the clinic. Unfortunately, no direct pharmacological inhibitor is available. Dimethyl fumarate (DMF) exhibits anti-breast cancer activities, in vitro and in pre-clinical in vivo models. Its therapeutic benefits stem from covalent modification of cellular thiols such as protein cysteines, but the full profile of molecular targets mediating its anti-breast cancer effects remains to be determined. METHODS: ER+breast cancer cells were treated with DMF followed by cysteine-directed proteomics. Cells with modulated ZNF217 levels were used to probe the efficacy of DMF. RESULTS: Covalent modification of ZNF217 by DMF identified by proteomics was confirmed by using a DMF-chemical probe. Inhibition of ZNF217's transcriptional activity by DMF was evident on reported ZNF217-target genes. ZNF217 as an oncogene has been shown to enhance stem-like properties, survival, proliferation, and invasion. Consistent with ZNF217 inhibition, DMF was more effective at blocking these ZNF217-driven phenotypes in cells with elevated ZNF217 expression. Furthermore, partial knockdown of ZNF217 led to a reduction in DMF's efficacy. DMF's in vivo activity was evaluated in a xenograft model of MCF-7 HER2 cells that have elevated expression of ZNF217 and DMF treatment resulted in significant inhibition of tumor growth. CONCLUSION: These data indicate that DMF's anti-breast cancer activities in the ER+HER2+models, at least in part, are due to inhibition of ZNF217. DMF is identified as a new covalent inhibitor of ZNF217.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Dimethyl Fumarate/pharmacology , Dimethyl Fumarate/therapeutic use , Receptors, Estrogen , Trans-Activators/genetics , Trans-Activators/metabolism , Trans-Activators/therapeutic use , MCF-7 Cells
3.
Mol Carcinog ; 62(12): 1803-1816, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37555760

ABSTRACT

The levels of the SELENOF selenoprotein are dramatically reduced in prostate cancer compared to adjacent benign tissue and reducing SELENOF in prostate epithelial cells results in the acquisition of features of the transformed phenotype. It was hypothesized that the aberrant increase in the eiF4a3 translation factor, which has an established role in RNA splicing and the regulation of selenoprotein translation, contributes to the lower levels of SELENOF. Using the available databases, eIF4a3 messenger RNA (mRNA) levels are elevated in prostate cancer compared to normal tissue as is the hypomethylation of the corresponding gene. Using a prostate cancer tissue microarray, we established that eiF4a3 levels are higher in prostate cancer tissue. Ectopic expression of eIF4a3 in prostate cancer cells reduced SELENOF levels and attenuated the readthrough of the UGA codon using a specialized reporter construct designed to examine UGA decoding, with the opposite effects observed using eIF4a3 knock-down constructs. Direct binding of eIF4a3 to the regulatory regions of SELENOF mRNA was established with pull-down experiments. Lastly, we show that an eIF4a3 inhibitor, eIF4a3-IN-2, increases SELENOF levels, UGA readthrough, and reduces binding of eIF4a3 to the SELENOF mRNA 3'-UTR in exposed cells. These data establish eIF4a3 as a likely prostate cancer oncogene and a regulator of SELENOF translation.


Subject(s)
Prostate , Prostatic Neoplasms , Male , Humans , Prostate/metabolism , Selenoproteins/genetics , Prostatic Neoplasms/genetics , Codon, Terminator , RNA, Messenger/genetics , RNA, Messenger/metabolism
4.
Breast Cancer Res ; 24(1): 88, 2022 12 08.
Article in English | MEDLINE | ID: mdl-36482488

ABSTRACT

BACKGROUND: Up to 40% of patients with estrogen receptor-positive (ER+) breast cancer experience relapse. This can be attributed to breast cancer stem cells (BCSCs), which are known to be involved in therapy resistance, relapse, and metastasis. Therefore, there is an urgent need to identify genes/pathways that drive stem-like cell properties in ER+ breast tumors. METHODS: Using single-cell RNA sequencing and various bioinformatics approaches, we identified a unique stem-like population and established its clinical relevance. With follow-up studies, we validated our bioinformatics findings and confirmed the role of ER and NFĸB in the promotion of stem-like properties in breast cancer cell lines and patient-derived models. RESULTS: We identified a novel quiescent stem-like cell population that is driven by ER and NFĸB in multiple ER+ breast cancer models. Moreover, we found that a gene signature derived from this stem-like population is expressed in primary ER+ breast tumors, endocrine therapy-resistant and metastatic cell populations and predictive of poor patient outcome. CONCLUSIONS: These findings indicate a novel role for ER and NFĸB crosstalk in BCSCs biology and understanding the mechanism by which these pathways promote stem properties can be exploited to improve outcomes for ER+ breast cancer patients at risk of relapse.


Subject(s)
Breast Neoplasms , Mammary Neoplasms, Animal , Animals , Humans , Female , Antineoplastic Agents, Hormonal/therapeutic use , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Breast Neoplasms/pathology , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/drug therapy , MCF-7 Cells , Mammary Neoplasms, Animal/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic
5.
Arch Biochem Biophys ; 732: 109465, 2022 12 15.
Article in English | MEDLINE | ID: mdl-36379313

ABSTRACT

There is an urgent need for new and improved therapeutic strategies in breast cancer, which is the most common malignancy affecting women in the United States and worldwide. Selenium (Se) is an essential trace element of the human diet and plays a critical role in many aspects of human health. Clinical and epidemiological studies summarized here clearly demonstrate that Se status correlates with breast cancer survival. As a result, one way to curb breast cancer mortality would be via Se supplementation, especially in patients with severely deplete Se status. Se manifests its biological activity through incorporation into selenoproteins as selenocysteine. However, a better understanding of tissue-specific mechanisms and roles for selenoproteins in general is required. Additionally, many human selenoproteins harbor single nucleotide polymorphisms, which impact protein expression and activity and have been associated with cancer susceptibility or impacting survival. Increasing evidence indicates that these genetic variations impinge on the interactions between Se and breast cancer. This highlights the importance of integrating the Se status with genetic factors to fully define the benefit of Se in breast cancer. While Se supplementation would clearly benefit a subset of patients, this requires first the identification of at-risk patients and warrants validation through intervention trials.


Subject(s)
Breast Neoplasms , Selenium , Humans , Female , Breast Neoplasms/epidemiology , Breast Neoplasms/genetics , Selenoproteins/genetics , Selenoproteins/metabolism , Selenocysteine/genetics , Selenocysteine/metabolism , Polymorphism, Single Nucleotide
6.
Int J Mol Sci ; 22(21)2021 Nov 07.
Article in English | MEDLINE | ID: mdl-34769469

ABSTRACT

SELENOF is a member of the class of selenoproteins in which the amino acid selenocysteine is co-translationally inserted into the elongating peptide in response to an in-frame UGA codon located in the 3'-untranslated (3'-UTR) region of the SELENOF mRNA. Polymorphisms in the 3'-UTR are associated with an increased risk of dying from prostate cancer and these variations are functional and 10 times more frequent in the genomes of African American men. SELENOF is dramatically reduced in prostate cancer compared to benign adjacent regions. Using a prostate cancer tissue microarray, it was previously established that the reduction of SELENOF in the cancers from African American men was significantly greater than in cancers from Caucasian men. When SELENOF levels in human prostate immortalized epithelial cells were reduced with an shRNA construct, those cells acquired the ability to grow in soft agar, increased the ability to migrate in a scratch assay and acquired features of energy metabolism associated with prostate cancer. These results support a role of SELENOF loss in prostate cancer progression and further indicate that SELENOF loss and genotype may contribute to the disparity in prostate cancer mortality experienced by African American men.


Subject(s)
Cell Transformation, Neoplastic/genetics , Epithelial Cells/pathology , Prostate/pathology , Selenoproteins/genetics , Adult , Aged , Case-Control Studies , Cell Line, Transformed , Cells, Cultured , Epithelial Cells/metabolism , Genotype , Humans , Male , Middle Aged , Phenotype , Prostate/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology
7.
Nat Chem Biol ; 13(1): 111-118, 2017 01.
Article in English | MEDLINE | ID: mdl-27870835

ABSTRACT

Resistance to endocrine therapies remains a major clinical problem for the treatment of estrogen receptor-α (ERα)-positive breast cancer. On-target side effects limit therapeutic compliance and use for chemoprevention, highlighting an unmet need for new therapies. Here we present a full-antagonist ligand series lacking the prototypical ligand side chain that has been universally used to engender antagonism of ERα through poorly understood structural mechanisms. A series of crystal structures and phenotypic assays reveal a structure-based design strategy with separate design elements for antagonism and degradation of the receptor, and access to a structurally distinct space for further improvements in ligand design. Understanding structural rules that guide ligands to produce diverse ERα-mediated phenotypes has broad implications for the treatment of breast cancer and other estrogen-sensitive aspects of human health including bone homeostasis, energy metabolism, and autoimmunity.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Receptors, Estrogen/antagonists & inhibitors , Antineoplastic Agents/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Female , Humans , Ligands , Models, Molecular , Molecular Structure , Receptors, Estrogen/metabolism , Structure-Activity Relationship
8.
J Biol Chem ; 291(7): 3639-47, 2016 Feb 12.
Article in English | MEDLINE | ID: mdl-26683377

ABSTRACT

In breast tumors, activation of the nuclear factor κB (NFκB) pathway promotes survival, migration, invasion, angiogenesis, stem cell-like properties, and resistance to therapy--all phenotypes of aggressive disease where therapy options remain limited. Adding an anti-inflammatory/anti-NFκB agent to breast cancer treatment would be beneficial, but no such drug is approved as either a monotherapy or adjuvant therapy. To address this need, we examined whether dimethyl fumarate (DMF), an anti-inflammatory drug already in clinical use for multiple sclerosis, can inhibit the NFκB pathway. We found that DMF effectively blocks NFκB activity in multiple breast cancer cell lines and abrogates NFκB-dependent mammosphere formation, indicating that DMF has anti-cancer stem cell properties. In addition, DMF inhibits cell proliferation and significantly impairs xenograft tumor growth. Mechanistically, DMF prevents p65 nuclear translocation and attenuates its DNA binding activity but has no effect on upstream proteins in the NFκB pathway. Dimethyl succinate, the inactive analog of DMF that lacks the electrophilic double bond of fumarate, is unable to inhibit NFκB activity. Also, the cell-permeable thiol N-acetyl l-cysteine, reverses DMF inhibition of the NFκB pathway, supporting the notion that the electrophile, DMF, acts via covalent modification. To determine whether DMF interacts directly with p65, we synthesized and used a novel chemical probe of DMF by incorporating an alkyne functionality and found that DMF covalently modifies p65, with cysteine 38 being essential for the activity of DMF. These results establish DMF as an NFκB inhibitor with anti-tumor activity that may add therapeutic value in the treatment of aggressive breast cancers.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Breast Neoplasms/drug therapy , Dimethyl Fumarate/pharmacology , NF-kappa B/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Signal Transduction/drug effects , Transcription Factor RelA/antagonists & inhibitors , Active Transport, Cell Nucleus/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cysteine/chemistry , Dimethyl Fumarate/chemistry , Dimethyl Fumarate/therapeutic use , Female , Gene Expression Regulation, Neoplastic/drug effects , Genes, Reporter/drug effects , Humans , Mice, Nude , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Random Allocation , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
9.
BMC Cancer ; 15: 845, 2015 Nov 04.
Article in English | MEDLINE | ID: mdl-26530254

ABSTRACT

INTRODUCTION: Activation of cyclooxygenase (COX)/prostaglandin and nuclear factor κB (NFκB) pathways can promote breast tumor initiation, growth, and progression to drug resistance and metastasis. Thus, anti-inflammatory drugs have been widely explored as chemopreventive and antineoplastic agents. Aspirin (ASA), in particular, is associated with reduced breast cancer incidence but gastrointestinal toxicity has limited its usefulness. To improve potency and minimize toxicity, ASA ester prodrugs have been developed, in which the carboxylic acid of ASA is masked and ancillary pharmacophores can be incorporated. To date, the effects of ASA and ASA prodrugs have been largely attributed to COX inhibition and reduced prostaglandin production. However, ASA has also been reported to inhibit the NFκB pathway at very high doses. Whether ASA prodrugs can inhibit NFκB signaling remains relatively unexplored. METHODS: A library of ASA prodrugs was synthesized and screened for inhibition of NFκB activity and cancer stem-like cell (CSC) properties, an important PGE2-and NFκB-dependent phenotype of aggressive breast cancers. Inhibition of NFκB activity was determined by dual luciferase assay, RT-QPCR, p65 DNA binding activity and Western blots. Inhibition of CSC properties was determined by mammosphere growth, CD44(+)CD24(-)immunophenotype and tumorigenicity at limiting dilution. RESULTS: While we identified multiple ASA prodrugs that are capable of inhibiting the NFκB pathway, several were associated with cytotoxicity. Of particular interest was GTCpFE, an ASA prodrug with fumarate as the ancillary pharmacophore. This prodrug potently inhibits NFκB activity without innate cytotoxicity. In addition, GTCpFE exhibited selective anti-CSC activity by reducing mammosphere growth and the CD44(+)CD24(-)immunophenotype. Moreover, GTCpFE pre-treated cells were less tumorigenic and, when tumors did form, latency was increased and growth rate was reduced. Structure-activity relationships for GTCpFE indicate that fumarate, within the context of an ASA prodrug, is essential for anti-NFκB activity, whereas both the ASA and fumarate moieties contributed to attenuated mammosphere growth. CONCLUSIONS: These results establish GTCpFE as a prototype for novel ASA-and fumarate-based anti-inflammatory drugs that: (i) are capable of targeting CSCs, and (ii) may be developed as chemopreventive or therapeutic agents in breast cancer.


Subject(s)
Aspirin/administration & dosage , Breast Neoplasms/drug therapy , NF-kappa B/genetics , Prodrugs/administration & dosage , Aspirin/chemical synthesis , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Fumarates/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , NF-kappa B/antagonists & inhibitors , NF-kappa B/biosynthesis , Neoplastic Stem Cells/drug effects , Prodrugs/chemical synthesis , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
12.
Biomolecules ; 13(3)2023 03 06.
Article in English | MEDLINE | ID: mdl-36979420

ABSTRACT

SELENOF, previously known as SEP15, is a selenoprotein that contains selenium in the form of the amino acid selenocysteine. Like other selenoproteins, the role for SELENOF in carcinogenesis has been investigated due to its altered expression compared to the corresponding normal tissue, its molecular function, and the association of genetic variations in the SELENOF gene to cancer risk or outcome. This review summarizes SELENOF's discovery, structure, cellular localization, and expression. SELENOF belongs to a new family of thioredoxin-like proteins. Published data summarized here indicate a likely role for SELENOF in redox protein quality control, and in the regulation of lipids, glucose, and energy metabolism. Current evidence indicates that loss of SELENOF contributes to the development of prostate and breast cancer, while its loss may be protective against colon cancer. Additional investigation into SELENOF's molecular mechanisms and its impact on cancer is warranted.


Subject(s)
Neoplasms , Selenium , Humans , Male , Neoplasms/genetics , Neoplasms/metabolism , Oxidation-Reduction , Prostate/metabolism , Selenium/metabolism , Selenoproteins/genetics , Selenoproteins/metabolism , Female
13.
Cancers (Basel) ; 15(14)2023 Jul 19.
Article in English | MEDLINE | ID: mdl-37509331

ABSTRACT

SELENOF expression is significantly lower in aggressive breast tumors compared to normal tissue, indicating that its reduction or loss may drive breast tumorigenesis. Deletion of SELENOF in non-tumorigenic immortalized breast epithelial MCF-10A cells resulted in enhanced proliferation, both in adherent culture and matrix-assisted three-dimmensional (3D) growth. Modulation of SELENOF in vitro through deletion or overexpression corresponded to changes in the cell-cycle regulators p21 and p27, which is consistent with breast tumor expression data from the METABRIC patient database. Together, these findings indicate that SELENOF affects both proliferation and cell death in normal epithelial and breast cancer cells, largely through the regulation of p21 and p27. In glandular cancers like breast cancer, the filling of luminal space is one of the hallmarks of early tumorigenesis. Loss of SELENOF abrogated apoptosis and autophagy, which are required for the formation of hollow acini in MCF-10A cells in matrix-assisted 3D growth, resulting in luminal filling. Conversely, overexpression of SELENOF induced cell death via apoptosis and autophagy. In conclusion, these findings are consistent with the notion that SELENOF is a breast tumor suppressor, and its loss contributes to breast cancer etiology.

14.
Oncogene ; 41(9): 1263-1268, 2022 02.
Article in English | MEDLINE | ID: mdl-35082382

ABSTRACT

Epidemiological evidence has indicated an inverse association between selenium status and various types of cancer, including breast cancer. Selenoproteins are the primary mediators of selenium effects in human health. We have previously reported loss of heterozygosity in breast tumor samples of the gene for one of the selenoproteins, SELENOF. The function of SELENOF remains unclear and whether SELENOF levels impact breast cancer risk or outcome is unknown. The mining of breast cancer patient databases revealed that SELENOF mRNA is significantly lower in late-stage tumor samples and lower levels of SELENOF also predict poor patient outcome from breast cancer. Genetically manipulating SELENOF in human breast cancer cells or in the murine mammary gland by overexpression, silencing or knockout impacted cell viability by affecting both proliferation and cell death. Restoring SELENOF can attenuate a number of aggressive cancer phenotypes in breast cancer cells, including clonogenic survival, and enhance the response to drugs or radiation used in breast cancer therapy. Importantly, enhancing SELENOF expression reduced in vivo tumor growth in a murine xenograft model of breast cancer. These data indicate that SELENOF is a new tumor suppressor in breast cancer.


Subject(s)
Breast Neoplasms
15.
Mol Cancer Res ; 18(7): 1018-1027, 2020 07.
Article in English | MEDLINE | ID: mdl-32245803

ABSTRACT

The purpose of this study was to identify critical pathways promoting survival of tamoxifen-tolerant, estrogen receptor α positive (ER+) breast cancer cells, which contribute to therapy resistance and disease recurrence. Gene expression profiling and pathway analysis were performed in ER+ breast tumors of patients before and after neoadjuvant tamoxifen treatment and demonstrated activation of the NF-κB pathway and an enrichment of epithelial-to mesenchymal transition (EMT)/stemness features. Exposure of ER+ breast cancer cell lines to tamoxifen, in vitro and in vivo, gives rise to a tamoxifen-tolerant population with similar NF-κB activity and EMT/stemness characteristics. Small-molecule inhibitors and CRISPR/Cas9 knockout were used to assess the role of the NF-κB pathway and demonstrated that survival of tamoxifen-tolerant cells requires NF-κB activity. Moreover, this pathway was essential for tumor recurrence following tamoxifen withdrawal. These findings establish that elevated NF-κB activity is observed in breast cancer cell lines under selective pressure with tamoxifen in vitro and in vivo, as well as in patient tumors treated with neoadjuvant tamoxifen therapy. This pathway is essential for survival and regrowth of tamoxifen-tolerant cells, and, as such, NF-κB inhibition offers a promising approach to prevent recurrence of ER+ tumors following tamoxifen exposure. IMPLICATIONS: Understanding initial changes that enable survival of tamoxifen-tolerant cells, as mediated by NF-κB pathway, may translate into therapeutic interventions to prevent resistance and relapse, which remain major causes of breast cancer lethality.


Subject(s)
Breast Neoplasms/pathology , Drug Resistance, Neoplasm , Estrogen Receptor alpha/metabolism , Gene Regulatory Networks/drug effects , Neoplasm Recurrence, Local/pathology , Tamoxifen/administration & dosage , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Mice , NF-kappa B/metabolism , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/metabolism , Neoplasm Transplantation , Signal Transduction/drug effects , Tamoxifen/pharmacology
16.
Drug Metab Dispos ; 37(1): 161-9, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18936111

ABSTRACT

Raloxifene and arzoxifene are benzothiophene selective estrogen receptor modulators (SERMs) of clinical use in postmenopausal osteoporosis and treatment of breast cancer and potentially in hormone replacement therapy. The benefits of arzoxifene are attributed to improved bioavailability over raloxifene, whereas the arzoxifene metabolite, desmethylarzoxifene (DMA) is a more potent antiestrogen. As polyaromatic phenolics, benzothiophene SERMs undergo oxidative metabolism to electrophilic quinoids. The long-term clinical use of SERMs demands increased understanding of correlations between structure and toxicity, with metabolism being a key component. A homologous series of 4'-substituted 4'-desmethoxyarzoxifene derivatives was developed, and metabolism was studied in liver and intestinal microsomes. Formation of glutathione conjugates was assayed in rat liver microsomes and novel adducts were characterized by liquid chromatography-tandem mass spectrometry. Formation of glucuronide conjugates was assayed in human intestine and liver microsomes, demonstrating formation of glucuronides ranging from 5 to 100% for the benzothiophene SERMs: this trend was inversely correlated with the loss of parent SERM in rat liver microsomal incubations. Molecular orbital calculations generated thermodynamic parameters for oxidation that correlated with Hammett substituent constants; however, metabolism in liver microsomes correlated with a combination of both Hammett and Hansch lipophilicity parameters. The results demonstrate a rich oxidative chemistry for the benzothiophene SERMs, the amplitude of which can be powerfully modulated, in a predictable manner, by structural tuning of the 4'-substituent. The predicted extensive metabolism of DMA was confirmed in vivo and compared with the relatively stable arzoxifene and F-DMA.


Subject(s)
Selective Estrogen Receptor Modulators/metabolism , Thiophenes/metabolism , Animals , Biotransformation , Chromatography, High Pressure Liquid , Drug Design , Humans , Microsomes, Liver/metabolism , Molecular Structure , Oxidation-Reduction , Rats , Selective Estrogen Receptor Modulators/chemistry , Tandem Mass Spectrometry , Thiophenes/chemistry
17.
Mol Cell Endocrinol ; 483: 97-101, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30659843

ABSTRACT

Estrogen receptor (ER) is the most important factor in the pathophysiology of breast cancer. Consequently, modulation of ER activity has been exploited to develop drugs against ER + breast cancer, such as tamoxifen, referred to as endocrine therapies. With deeper understanding of ER mechanism of action, posttranslational modifications (PTMs) are increasingly recognized as important in mediating ER activity. Some ER PTMs such as phosphorylation, are studied in the context of ligand-independent ER activity. However, they also play a pivotal role in defining the actions and outcome of the antiestrogen-bound ER. The complexity of these actions is increasing as new PTMs are identified, yet the functional consequences and clinical implications are not fully understood. This review will examine and summarize new emerging mechanistic knowledge and clinical data in breast cancer on how these PTMs affect antiestrogen-ER activity, with an emphasis on phosphorylation of serine 305 (S305). This phosphorylation site represents an integrated hub of oncogenic signaling to modulate ER conformation, dimerization, coregulators, and DNA binding to profoundly reduce sensitivity to endocrine therapy. Consequently, (i) S305 has the potential to become a useful marker of tamoxifen response, and (ii) blocking S305 phosphorylation defines a new therapeutic strategy to overcome tamoxifen resistance in breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Receptors, Estrogen/metabolism , Serine/chemistry , Tamoxifen/pharmacology , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm , Female , Humans , Phosphorylation/drug effects , Protein Conformation , Protein Processing, Post-Translational , Receptors, Estrogen/chemistry , Tamoxifen/therapeutic use
18.
Adv Exp Med Biol ; 617: 601-7, 2008.
Article in English | MEDLINE | ID: mdl-18497087

ABSTRACT

Oxidative metabolism of estrogens has been associated with genotoxicity. O-methylation of catechol estrogens is considered as a protective mechanism. 4-Methoxyequilenin (4-MeOEN) is the O-methylated product of 4-hydroxyequilenin (4-OHEN). 4-OHEN, the major catechol metabolite of the equine estrogens present in the most widely prescribed hormone replacement therapeutics, causes DNA damage via quinone formation. In this study, estrogen receptor (ERa) binding of 4-MeOEN was compared with estradiol (E2) and equilenin derivatives including 4-BrEN using computer modeling, estrogen response element (ERE)-luciferase induction in MCF-7 cells, and alkaline phosphatase (AP) induction in Ishikawa cells. 4-MeOEN induced AP and luciferase with nanomolar potency and displayed a similar profile of activity to E2. Molecular modeling indicated that MeOEN could be a ligand for ERa despite no binding being observed in the ERa competitive binding assay. Methylation of 4-OHEN may not represent a detoxification pathway, since 4-MeOEN is a full estrogen agonist with nanomolar potency.


Subject(s)
Breast Neoplasms/drug therapy , Endometrial Neoplasms/drug therapy , Equilenin/analogs & derivatives , Transcriptional Activation/drug effects , Alkaline Phosphatase/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Computer Simulation , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Equilenin/pharmacology , Estradiol/chemistry , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation , Horses , Humans , Luciferases/metabolism , Response Elements/genetics , Transcription, Genetic , Tumor Cells, Cultured
19.
Mol Cancer Ther ; 6(9): 2418-28, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17876041

ABSTRACT

The benzothiophene selective estrogen receptor modulators (SERM) raloxifene and arzoxifene are in clinical use and clinical trials for chemoprevention of breast cancer and other indications. These SERMs are "oxidatively labile" and therefore have potential to activate antioxidant responsive element (ARE) transcription of genes for cytoprotective phase II enzymes such as NAD(P)H-dependent quinone oxidoreductase 1 (NQO1). To study this possible mechanism of cancer chemoprevention, a family of benzothiophene SERMs was developed with modulated redox activity, including arzoxifene and its metabolite desmethylarzoxifene (DMA). The relative antioxidant activity of these SERMs was assayed and correlated with induction of NQO1 in murine and human liver cells. DMA was found to induce NQO1 and to activate ARE more strongly than other SERMs, including raloxifene and 4-hydroxytamoxifen. Livers from female, juvenile rats treated for 3 days with estradiol and/or with the benzothiophene SERMs arzoxifene, DMA, and F-DMA showed substantial induction of NQO1 by the benzothiophene SERMs. No persuasive evidence in this assay or in MCF-7 breast cancer cells was obtained of a major role for the estrogen receptor in induction of NQO1 by the benzothiophene SERMs. These results suggest that arzoxifene might provide chemopreventive benefits over raloxifene and other SERMs via metabolism to DMA and stimulation of ARE-mediated induction of phase II enzymes. The correlation of SERM structure with antioxidant activity and NQO1 induction also suggests that oxidative bioactivation of SERMs may be modulated to enhance chemopreventive activity.


Subject(s)
Antioxidants/pharmacology , Breast Neoplasms/drug therapy , Carcinoma, Hepatocellular/drug therapy , Liver/drug effects , Selective Estrogen Receptor Modulators/chemistry , Selective Estrogen Receptor Modulators/pharmacology , Animals , Chemoprevention , Female , Humans , Liver Neoplasms/drug therapy , Luciferases , Mice , NAD(P)H Dehydrogenase (Quinone)/metabolism , Oxidation-Reduction , Peroxides , Piperidines/pharmacology , Raloxifene Hydrochloride/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Estrogen/metabolism , Selective Estrogen Receptor Modulators/chemical synthesis , Thiophenes/pharmacology , Tumor Cells, Cultured/drug effects
20.
Cancer Res ; 78(4): 974-984, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29229606

ABSTRACT

A growing body of evidence suggests that the inflammatory NFκB pathway is associated with the progression of ER+ tumors to more aggressive stages. However, it is unknown whether NFκB is a driver or a consequence of aggressive ER+ disease. To investigate this question, we developed breast cancer cell lines expressing an inducible, constitutively active form of IκB kinase ß (CA-IKKß), a key kinase in the canonical NFκB pathway. We found that CA-IKKß blocked E2-dependent cell proliferation in vitro and tumor growth in vivo in a reversible manner, suggesting that IKKß may contribute to tumor dormancy and recurrence of ER+ disease. Moreover, coactivation of ER and IKKß promoted cell migration and invasion in vitro and drove experimental metastasis in vivo Gene expression profiling revealed a strong association between ER and CA-IKKß-driven gene expression and clinically relevant invasion and metastasis gene signatures. Mechanistically, the invasive phenotype appeared to be driven by an expansion of a basal/stem-like cell population rather than EMT. Taken together, our findings suggest that coactivation of ER and the canonical NFκB pathway promotes a dormant, metastatic phenotype in ER+ breast cancer and implicates IKKß as a driver of certain features of aggressive ER+ breast cancer.Significance: The canonical NFκB pathway promotes expansion of stem/basal-like cells and a dormant, metastatic phenotype in ER+ breast cancer cells. Cancer Res; 78(4); 974-84. ©2017 AACR.


Subject(s)
Breast Neoplasms/genetics , I-kappa B Kinase/metabolism , Receptors, Estrogen/metabolism , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Female , Humans , Mice , Mice, Nude , Phenotype , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL