Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
2.
Cancer Res ; 82(20): 3718-3733, 2022 10 17.
Article in English | MEDLINE | ID: mdl-36007109

ABSTRACT

SIGNIFICANCE: Dll1+ breast cancer cells activate Notch signaling in cancer-associated fibroblasts that increases Wnt ligand secretion and leads to ß-catenin-driven radioresistance and metastasis, opening new therapeutic avenues for breast cancer.


Subject(s)
Breast Neoplasms , Cancer-Associated Fibroblasts , Breast Neoplasms/pathology , Breast Neoplasms/radiotherapy , Cancer-Associated Fibroblasts/pathology , Female , Humans , Intercellular Signaling Peptides and Proteins , Ligands , Receptors, Notch , beta Catenin
3.
Cancer Res ; 81(9): 2399-2414, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33514514

ABSTRACT

Inflammatory breast cancer (IBC) is a highly metastatic breast carcinoma with high frequency of estrogen receptor α (ERα) negativity. Here we explored the role of the second ER subtype, ERß, and report expression in IBC tumors and its correlation with reduced metastasis. Ablation of ERß in IBC cells promoted cell migration and activated gene networks that control actin reorganization, including G-protein-coupled receptors and downstream effectors that activate Rho GTPases. Analysis of preclinical mouse models of IBC revealed decreased metastasis of IBC tumors when ERß was expressed or activated by chemical agonists. Our findings support a tumor-suppressive role of ERß by demonstrating the ability of the receptor to inhibit dissemination of IBC cells and prevent metastasis. On the basis of these findings, we propose ERß as a potentially novel biomarker and therapeutic target that can inhibit IBC metastasis and reduce its associated mortality. SIGNIFICANCE: These findings demonstrate the capacity of ERß to elicit antimetastatic effects in highly aggressive inflammatory breast cancer and propose ERß and the identified associated genes as potential therapeutic targets in this disease.


Subject(s)
Actins/metabolism , Cell Movement/genetics , Estrogen Receptor beta/metabolism , Inflammatory Breast Neoplasms/metabolism , Signal Transduction/genetics , Actin Cytoskeleton/metabolism , Animals , Cohort Studies , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/genetics , Female , Gene Knockout Techniques , HEK293 Cells , Humans , Inflammatory Breast Neoplasms/genetics , Inflammatory Breast Neoplasms/pathology , MCF-7 Cells , Mice , Neoplasm Metastasis/genetics , Transfection , Tumor Burden/genetics , Xenograft Model Antitumor Assays
4.
Oncogene ; 39(22): 4344-4357, 2020 05.
Article in English | MEDLINE | ID: mdl-32335582

ABSTRACT

We explore a novel strategy of activating immune signaling through increased micronuclei formation utilizing a cell cycle checkpoint inhibitor to drive cell cycle progression following ionizing radiation. The Chk1/2 inhibitor AZD7762 is used to abrogate radiation therapy (RT)-induced G2/M cell cycle arrest in multiple cell lines and, we find that this therapeutic combination promotes increased micronuclei formation in vitro and subsequently drives increased type I interferon signaling and cytotoxic T-cell activation. In vivo studies using B16-F10 melanoma cancer cells implanted in C57/BL6 mice demonstrate improved rates of tumor control at the abscopal (unirradiated) site, located outside of the radiation field, only in the AZD7762 + RT group, with a corresponding reduction in mean tumor volume, increase in the CD8 T-cell population, and immune activated gene signaling. Our results demonstrate that targeted inhibition of cell cycle checkpoint activation following ionizing radiation drives increased production of immunogenic micronuclei, leading to systemic tumor response with potential future clinical benefit.


Subject(s)
Cell Nucleus/drug effects , Cell Nucleus/radiation effects , Checkpoint Kinase 1/antagonists & inhibitors , Checkpoint Kinase 2/antagonists & inhibitors , G2 Phase Cell Cycle Checkpoints/drug effects , G2 Phase Cell Cycle Checkpoints/radiation effects , Melanoma, Experimental/immunology , Neoplasm Proteins/antagonists & inhibitors , Thiophenes/pharmacology , Urea/analogs & derivatives , Animals , Cell Line, Tumor , Female , Humans , Interferon-beta/biosynthesis , Interferon-beta/genetics , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Melanoma, Experimental/radiotherapy , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice, Inbred C57BL , Micronucleus Tests , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , STAT1 Transcription Factor/biosynthesis , STAT1 Transcription Factor/genetics , Tumor Burden/drug effects , Tumor Burden/radiation effects , Urea/pharmacology , Xenograft Model Antitumor Assays
5.
Oncogene ; 38(12): 2092-2107, 2019 03.
Article in English | MEDLINE | ID: mdl-30442981

ABSTRACT

Aberrant Notch signaling is implicated in several cancers, including breast cancer. However, the mechanistic details of the specific receptors and function of ligand-mediated Notch signaling that promote breast cancer remains elusive. In our studies we show that DLL1, a Notch signaling ligand, is significantly overexpressed in ERα+ luminal breast cancer. Intriguingly, DLL1 overexpression correlates with poor prognosis in ERα+ luminal breast cancer, but not in other subtypes of breast cancer. In addition, this effect is specific to DLL1, as other Notch ligands (DLL3, JAGGED1, and JAGGED2) do not influence the clinical outcome of ERα+ patients. Genetic studies show that DLL1-mediated Notch signaling in breast cancer is important for tumor cell proliferation, angiogenesis, and cancer stem cell function. Consistent with prognostic clinical data, we found the tumor-promoting function of DLL1 is exclusive to ERα+ luminal breast cancer, as loss of DLL1 inhibits both tumor growth and lung metastasis of luminal breast cancer. Importantly, we find that estrogen signaling stabilizes DLL1 protein by preventing its proteasomal and lysososmal degradations. Moreover, estrogen inhibits ubiquitination of DLL1. Together, our results highlight an unexpected and novel subtype-specific function of DLL1 in promoting luminal breast cancer that is regulated by estrogen signaling. Our studies also emphasize the critical role of assessing subtype-specific mechanisms driving tumor growth and metastasis to generate effective subtype-specific therapeutics.


Subject(s)
Breast Neoplasms/pathology , Estrogens/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Receptors, Notch/metabolism , Signal Transduction , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/diagnosis , Breast Neoplasms/metabolism , Calcium-Binding Proteins , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic , Disease Progression , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Lysosomes/metabolism , Mice , Neoplasm Metastasis , Neoplastic Stem Cells/pathology , Neovascularization, Pathologic , Prognosis , Ubiquitination
6.
Oncotarget ; 9(32): 22509-22522, 2018 Apr 27.
Article in English | MEDLINE | ID: mdl-29854295

ABSTRACT

Estrogen receptor α (ERα)-positive breast cancers tend to develop resistance to both endocrine therapy and chemotherapy. Despite recent progress in defining molecular pathways that confer endocrine resistance, the mechanisms that regulate chemotherapy response in luminal tumors remain largely elusive. Luminal tumors often express wild-type p53 that is a major determinant of the cellular DNA damage response. Similar to p53, the second ER subtype, ERß, has been reported to inhibit breast tumorigenesis by acting alone or in collaboration with p53. However, a synergistic mechanism of action has not been described. Here, we suggest that ERß relies on p53 to elicit its tumor repressive actions in ERα-positive breast cancer cells. Upregulation of ERß and treatment with ERß agonists potentiates the tumor suppressor function of p53 resulting in decreased survival. This effect requires molecular interaction between the two proteins that disrupts the inhibitory action of ERα on p53 leading to increased transcriptional activity of p53. In addition, we show that the same interaction alters the chemosensitivity of endocrine-resistant cells including their response to tamoxifen therapy. Our results suggest a collaboration of ERß and p53 tumor suppressor activity in breast cancer cells that indicates the importance of ligand-regulated ERß as a tool to target p53 activity and improve the clinical management of resistant disease.

7.
Mol Cancer Res ; 16(2): 233-242, 2018 02.
Article in English | MEDLINE | ID: mdl-29117942

ABSTRACT

The expression of wild-type estrogen receptor ß (ESR2/ERß1) correlates with clinical outcome in patients with non-small cell lung cancer (NSCLC). However, the molecular mechanism that accounts for this association is currently poorly understood. ERß1 was previously linked to chemotherapy response in patients with breast cancer and in breast cancer cells. The effect of the receptor in NSCLC cells after chemotherapy treatment, a common remedy for advanced NSCLC, has not been studied. Here, upregulation of ERß1 increases the sensitivity of NSCLC cells to treatment with doxorubicin and etoposide. This effect was primarily observed in p53-defecient NSCLC cells. In these cells, ERß1 either enhanced G2-M cell-cycle arrest by activating the checkpoint kinase 1 (Chk1) and altering downstream signaling or induced apoptosis. The expression of p63 target genes that control G2-M checkpoint activation was altered by ERß1 suggesting an ERß1-p63 transcriptional cooperation in lung cancer cells that affects DNA damage response (DDR). These results suggest involvement of ERß1 in the mechanism that regulates DNA damage response in NSCLC cells and support the potential predictive and therapeutic value of the receptor in clinical management of the disease.Implications: This study demonstrating the impact of ERß1 on chemosensitivity of NSCLC cells suggests the predictive value of the receptor for successful response of tumors to chemotherapy and the potential benefit of chemotherapy-treated patients from the use of ER ligands. Mol Cancer Res; 16(2); 233-42. ©2017 AACR.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Doxorubicin/pharmacology , Estrogen Receptor beta/genetics , Etoposide/pharmacology , Lung Neoplasms/genetics , Up-Regulation , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Damage , DNA Repair/drug effects , Estrogen Receptor beta/metabolism , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Transfection , Tumor Suppressor Protein p53/genetics
8.
Breast Cancer Res ; 19(1): 79, 2017 Jul 03.
Article in English | MEDLINE | ID: mdl-28673316

ABSTRACT

BACKGROUND: Upregulation of estrogen receptor beta (ERß) in breast cancer cells is associated with epithelial maintenance, decreased proliferation and invasion, and a reduction in the expression of the receptor has been observed in invasive breast tumors. However, proof of an association between loss of ERß and breast carcinogenesis is still missing. METHODS: To study the role of ERß in breast oncogenesis, we generated mouse conditional mutants with specific inactivation of ERß and p53 in the mammary gland epithelium. For epithelium-specific knockout of ERß and p53, ERß F/F and p53 F/F mice were crossed to transgenic mice that express the Cre recombinase under the control of the human keratin 14 promoter. RESULTS: Somatic loss of ERß significantly accelerated formation of p53-deficient mammary tumors. Loss of the receptor also resulted in the development of less differentiated carcinomas with stronger spindle cell morphology and decreased expression of luminal epithelial markers. CONCLUSIONS: Our results show that synergism between ERß and p53 inactivation functions to determine important aspects of breast oncogenesis and cancer progression.


Subject(s)
Breast Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Estrogen Receptor beta/genetics , Tumor Suppressor Protein p53/genetics , Alleles , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Epithelial-Mesenchymal Transition/genetics , Epithelium/metabolism , Estrogen Receptor beta/metabolism , Female , Gene Expression , Gene Silencing , Genotype , Immunohistochemistry , Kaplan-Meier Estimate , Mice , Mice, Transgenic , Phenotype , Prognosis , Tumor Suppressor Protein p53/metabolism
9.
J Steroid Biochem Mol Biol ; 163: 45-50, 2016 10.
Article in English | MEDLINE | ID: mdl-27045680

ABSTRACT

Activation of the unfolded protein response (UPR) confers resistance to anti-estrogens and chemotherapeutics in estrogen receptor α (ERα)-positive and triple-negative breast cancers. Among the regulators of the UPR in breast cancer is estrogen signaling. Estrogen regulates major components of the UPR and ER expression is associated with the sensitivity of tumor cells to UPR-regulated apoptosis. Recent studies have confirmed the crosstalk between the ERs and UPR and suggest novel therapeutic strategies that combine targeting of both signaling pathways. These remedies may be more effective in repressing oncogenic adaptive mechanisms and benefit patients with resistant disease.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/metabolism , Estrogen Antagonists/therapeutic use , Estrogens/metabolism , Gene Expression Regulation, Neoplastic , Signal Transduction/genetics , Unfolded Protein Response/drug effects , Activating Transcription Factor 6/genetics , Activating Transcription Factor 6/metabolism , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Female , Humans , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
10.
Oncotarget ; 7(12): 13599-611, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26871946

ABSTRACT

Most (80%) of the triple-negative breast cancers (TNBCs) express mutant p53 proteins that acquire oncogenic activities including promoting metastasis. We previously showed that wild-type ERß (ERß1) impedes epithelial to mesenchymal transition (EMT) and decreases the invasiveness of TNBC cells. In the present study we searched for signaling pathways that ERß1 uses to inhibit EMT and invasion in TNBC cells. We show that ERß1 binds to and opposes the transcriptional activity of mutant p53 at the promoters of genes that regulate metastasis. p63 that transcriptionally cooperates with mutant p53 also binds to ERß1. Downregulation of p63 represses the epithelial phenotype of ERß1-expressing cells and alters the expression of mutant p53 target genes. These results describe a novel mechanism through which ERß1 can disturb oncogenic signals to inhibit aggressiveness in TNBCs.


Subject(s)
Epithelial-Mesenchymal Transition , Estrogen Receptor beta/metabolism , Gene Expression Regulation, Neoplastic , Mutation , Triple Negative Breast Neoplasms/pathology , Tumor Suppressor Protein p53/genetics , Apoptosis , Cell Movement , Cell Proliferation , Estrogen Receptor beta/genetics , Humans , Neoplasm Invasiveness , Signal Transduction , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
11.
Trends Endocrinol Metab ; 26(9): 453-4, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26277479

ABSTRACT

Copy number loss of the PGR gene and decreased expression of progesterone receptor (PR) may account for worse clinical outcomes in some individuals with estrogen receptor α (ERα)-positive breast cancer. A recent report shows that PR activation inhibits estrogen-driven breast tumor growth by altering ERα chromatin binding and transcriptional activity.


Subject(s)
Breast Neoplasms/genetics , Receptor Cross-Talk , Receptors, Estrogen/genetics , Receptors, Progesterone/genetics , Estrogen Receptor alpha/genetics , Female , Humans
12.
Trends Endocrinol Metab ; 26(9): 467-76, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26183887

ABSTRACT

A significant number of estrogen receptor α (ERα)-positive breast tumors develop resistance to endocrine therapy and recur with metastatic disease. Several mechanisms of endocrine resistance have been proposed, including genetic alterations that lead to ERs with altered protein sequence. By altering the conformation of the protein and increasing the interaction with coactivators, point mutations in ESR1, the gene encoding ERα, promote an active form of the receptor in the absence of hormone that assists tumor cells to evade hormonal treatments. Recent studies have confirmed that ESR1 point mutations frequently occur in metastatic breast tumors that are refractory to endocrine therapy, and suggest the development of novel strategies that may be more effective in controlling ER signaling and benefit patients with recurrent and metastatic disease.


Subject(s)
Breast Neoplasms/genetics , Estrogen Receptor alpha/genetics , Receptors, Estrogen/genetics , Breast Neoplasms/epidemiology , Female , Humans , Mutation/genetics
13.
Am J Physiol Renal Physiol ; 309(2): F98-108, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26017975

ABSTRACT

TNF has been implicated in glomerular diseases, but its actions on podocytes are not well understood. Endogenous TNF expression is markedly increased in mouse podocytes exposed to sera from patients with recurrent focal segmental glomerulosclerosis, and TNF is able to increase its own expression in these cells. Exposure of podocytes to TNF increased phosphorylation of NF-κB p65-RelA followed by increased tyrosine phosphorylation of STAT3. STAT3 activation was blocked by the NF-κB inhibitor JSH-23 and by the STAT3 inhibitor stattic, whereas TNF-evoked NF-κB activation was not affected by stattic. TNF treatment increased nuclear accumulation of nuclear factor of activated T cells (NFAT)c1 in podocytes, a process that occurred downstream of STAT3 activation. TNF also increased expression of cyclin D1 but had no effect on cyclin-dependent kinase 4, p27(kip), or podocin. Despite its effects on cyclin D1, TNF treatment for up to 72 h did not cause podocytes to reenter the cell cycle. TNF increased total expression of transient receptor potential (TRP)C6 channels through a pathway dependent on NFATc1 and increased the steady-state expression of TRPC6 subunits on the podocyte cell surface. TNF effects on TRPC6 trafficking required ROS. Consistent with this, La(3+)-sensitive cationic currents activated by a diacylglycerol analog were increased in TNF-treated cells. The effects of TNF on NFATc1 and TRPC6 expression were blocked by cyclosporine A but were not blocked by the pan-TRP inhibitor SKF-96365. TNF therefore influences multiple pathways previously implicated in podocyte pathophysiology and is likely to sensitize these cells to other insults.


Subject(s)
Podocytes/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Adult , Animals , Cell Line , Glomerulosclerosis, Focal Segmental/metabolism , Humans , Male , Mice , NFATC Transcription Factors/metabolism , STAT3 Transcription Factor/metabolism , TRPC Cation Channels/metabolism , TRPC6 Cation Channel
14.
Oncotarget ; 5(11): 3871-9, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-25003695

ABSTRACT

P53 mutations are associated with invasive tumors in mouse models. We assessed the p53mutations and survival in patients with advanced cancer treated in the Phase I Program. Of 691 tested patients, 273 (39.5%) had p53 mutations. Patients with p53 mutations were older (p<.0001) and had higher numbers of liver metastases (p=.005). P53 mutations were associated with higher numbers of other aberrations; PTEN (p=.0005) and HER2 (p=.003)aberrations were more common in the p53 mutation group. No survival difference was observed between patients with p53 mutations and those with wild-type p53. In patients with wild-type p53 and other aberrations, patients treated with matched-therapy against the additional aberrations had longer survival compared to those treated with non-matched-therapy or those who received no therapy (median survival, 26.0 vs. 11.8 vs. 9.8 months, respectively; p= .0007). Results were confirmed in a multivariate analysis (p= .0002). In the p53 mutation group with additional aberrations, those who received matched-therapy against the additional aberrations had survival similar to those treated with non-matched-therapy or those who received no therapy (p=.15). In conclusion, our results demonstrated resistance to matched-targeted therapy to the other aberrations in patients with p53 mutations and emphasize the need to overcome this resistance.


Subject(s)
Genes, p53 , Mutation , Neoplasms/genetics , Clinical Trials, Phase I as Topic , Female , Humans , Male , Middle Aged , Neoplasms/therapy , Prognosis , Survival Analysis , Survival Rate , Treatment Outcome
15.
Mol Cancer Res ; 12(6): 843-54, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24618619

ABSTRACT

UNLABELLED: Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related deaths worldwide. In addition to the aberrant growth factor signaling, dysregulation of other pathways, such as those mediated by estrogens and their receptors, has been linked to NSCLC initiation and progression. Although the expression of wild-type estrogen receptor ß (ERß1) has been associated with prolonged disease-free survival in patients with NSCLC, the molecular mechanism that accounts for this correlation is unknown. Here, upregulation of ERß1 reduced proliferation and enhanced apoptosis in the context of mutant RAS. ERß1 was found to induce apoptosis by stimulating the intrinsic apoptotic pathway that involves BIM, a Bcl-2 proapoptotic family member that is regulated by the extracellular signal-regulated kinase (ERK). Downregulation of EGFR and inactivation of RAS and the downstream components ERK1/2 were found to be involved in the ERß1-induced apoptosis. Manipulation of EGFR and RAS expression and activity in ERß1-expressing cells revealed the central role of oncogenic RAS inhibition in the ERß1-mediated proapoptotic phenotype and EGFR regulation. These results demonstrate that ERß1 decreases the survival of NSCLC cells by regulating oncogenic RAS signaling. IMPLICATIONS: The ability of ERß1 to regulate the oncogenic functions of RAS suggests its importance in the biology of NSCLC and its clinical management. Mol Cancer Res; 12(6); 843-54. ©2014 AACR.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Estrogen Receptor beta/metabolism , Lung Neoplasms/metabolism , ras Proteins/metabolism , Apoptosis/physiology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/physiology , Disease Progression , Disease-Free Survival , Estrogen Receptor beta/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Phenotype , Signal Transduction , Transfection , ras Proteins/genetics
16.
Breast Cancer Res ; 14(6): R148, 2012 Nov 16.
Article in English | MEDLINE | ID: mdl-23158001

ABSTRACT

INTRODUCTION: Epithelial to mesenchymal transition (EMT) is associated with the basal-like breast cancer phenotypes. 60% of basal-like cancers have been shown to express wild-type estrogen receptor beta (ERbeta1). However, it is still unclear whether the ERbeta expression is related to EMT, invasion and metastasis in breast cancer. In the present study, we examined whether ERbeta1 through regulating EMT can influence invasion and metastasis in basal-like cancers. METHODS: Basal-like breast cancer cells (MDA-MB-231 and Hs578T) in which ERbeta1 was either overexpressed or downregulated were analyzed for their ability to migrate and invade (wound-healing assay, matrigel-coated Transwell assay) as well as for the expression of EMT markers and components of the EGFR pathway (immunoblotting, RT-PCR). Coimmunoprecipitation and ubiquitylation assays were employed to examine whether ERbeta1 alters EGFR protein degradation and the interaction between EGFR and the ubiquitin ligase c-Cbl. The metastatic potential of the ERbeta1-expressing MDA-MB-231 cells was evaluated in vivo in a zebrafish xenotransplantation model and the correlation between ERbeta1 and E-cadherin expression was examined in 208 clinical breast cancer specimens by immunohistochemistry. RESULTS: Here we show that ERbeta1 inhibits EMT and invasion in basal-like breast cancer cells when they grow either in vitro or in vivo in zebrafish. The inhibition of EMT correlates with an ERbeta1-mediated upregulation of miR-200a/b/429 and the subsequent repression of ZEB1 and SIP1, which results in increased expression of E-cadherin. The positive correlation of ERbeta1 and E-cadherin expression was additionally observed in breast tumor samples. Downregulation of the basal marker EGFR through stabilization of the ubiquitin ligase c-Cbl complexes and subsequent ubiquitylation and degradation of the activated receptor is involved in the ERbeta1-mediated repression of EMT and induction of EGFR signaling abolished the ability of ERbeta1 to sustain the epithelial phenotype. CONCLUSIONS: Taken together, the results of our study strengthen the association of ERbeta1 with the regulation of EMT and propose the receptor as a potential crucial marker in predicting metastasis in breast cancer.


Subject(s)
Breast Neoplasms/pathology , Epithelial-Mesenchymal Transition , ErbB Receptors/metabolism , Estrogen Receptor beta/metabolism , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cadherins/biosynthesis , Cell Line, Tumor , Estrogen Receptor beta/biosynthesis , Estrogen Receptor beta/genetics , Female , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/biosynthesis , Humans , MCF-7 Cells , MicroRNAs/biosynthesis , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/pathology , Neoplasm Transplantation , Neoplasms, Basal Cell , Nerve Tissue Proteins/biosynthesis , Proto-Oncogene Proteins c-cbl/metabolism , RNA Interference , RNA, Small Interfering , RNA-Binding Proteins/biosynthesis , Repressor Proteins/biosynthesis , Snail Family Transcription Factors , Transcription Factors/biosynthesis , Transcription Factors/metabolism , Transplantation, Heterologous , Ubiquitination , Zebrafish , Zinc Finger E-box Binding Homeobox 2 , Zinc Finger E-box-Binding Homeobox 1
17.
J Clin Invest ; 122(8): 2771-3, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22820293

ABSTRACT

Alteration of the ERα/ERß balance is a critical step in breast cancer development and progression, and selective restoration of the activity of estrogen receptors has been proposed as one of the major therapeutic approaches for breast cancer. In this issue of JCI, Cheng et al. show that, by differentially modulating the stability of ERα and ERß, PES1 increases the ERα/ERß ratio and triggers breast tumor growth. These findings highlight PES1 as a potential target for the treatment of breast cancer.

18.
Nat Rev Cancer ; 11(8): 597-608, 2011 Jul 22.
Article in English | MEDLINE | ID: mdl-21779010

ABSTRACT

By eliciting distinct transcriptional responses, the oestrogen receptors (ERs) ERα and ERß exert opposite effects on cellular processes that include proliferation, apoptosis and migration and that differentially influence the development and the progression of cancer. Perturbation of ER subtype-specific expression has been detected in various types of cancer, and the differences in the expression of ERs are correlated with the clinical outcome. The changes in the bioavailability of ERs in tumours, together with their specific biological functions, promote the selective restoration of their activity as one of the major therapeutic approaches for hormone-dependent cancers.


Subject(s)
Estrogen Receptor alpha/physiology , Estrogen Receptor beta/physiology , Neoplasms/physiopathology , Animals , Humans , Neoplasms/drug therapy , Prognosis , Protein Isoforms
20.
Mol Endocrinol ; 25(8): 1326-36, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21680658

ABSTRACT

The growth of many human breast tumors requires the proliferative effect of estrogen acting via the estrogen receptor α (ERα). ERα signaling is therefore a clinically important target for breast cancer prevention and therapeutics. Although extensively studied, the mechanism by which ERα promotes proliferation remains to be fully established. We observed an up-regulation of transcript encoding the pH-sensitive two-pore domain potassium channel KCNK5 in a screen for genes stimulated by 17ß-estradiol (E2) in the ERα(+) breast cancer cell lines MCF-7 and T47D. KCNK5 mRNA increased starting 1 h after the onset of E2 treatment, and protein levels followed after 12 h. Estrogen-responsive elements are found in the enhancer region of KCNK5, and chromatin immunoprecipitation assays revealed binding of ERα to the KCNK5 enhancer in E2-treated MCF-7 cells. Cells treated with E2 also showed increases in the amplitude of pH-sensitive potassium currents, as assessed by whole-cell recordings. These currents are blocked by clofilium. Although confocal microscopy suggested that most of the channels are located in intracellular compartments, the increase in macroscopic currents suggests that E2 treatment increases the number of active channels at the cell surface. Application of small interfering RNA specific for KCNK5 decreased pH-sensitive potassium currents and also reduced the estrogen-induced proliferation of T47D cells. We conclude that E2 induces the expression of KCNK5 via ERα(+) in breast cancer cells, and this channel plays a role in regulating proliferation in these cell lines. KCNK5 may therefore represent a useful target for treatment, for example, of tamoxifen-resistant breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Estrogen Receptor alpha/metabolism , Potassium Channels, Tandem Pore Domain/chemistry , Potassium Channels, Tandem Pore Domain/metabolism , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Estrogens/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Half-Life , Humans , Hydrogen-Ion Concentration/drug effects , Ion Channel Gating/drug effects , Potassium Channels, Tandem Pore Domain/genetics , Protein Structure, Tertiary , Protein Transport/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...