Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 117(42): 26347-26355, 2020 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-33020300

RESUMEN

Loss of the tumor suppressor, PTEN, is one of the most common findings in prostate cancer (PCa). This loss leads to overactive Akt signaling, which is correlated with increased metastasis and androgen independence. However, another tumor suppressor, inositol-polyphosphate 4-phosphatase type II (INPP4B), can partially compensate for the loss of PTEN. INPP4B is up-regulated by androgens, and this suggests that androgen-deprivation therapy (ADT) would lead to hyperactivity of AKT. However, in the present study, we found that in PCa, samples from men treated with ADT, ERß, and INPP4B expression were maintained in some samples. To investigate the role of ERß1 in regulation of INPPB, we engineered the highly metastatic PCa cell line, PC3, to express ERß1. In these cells, INPP4B was induced by ERß ligands, and this induction was accompanied by inhibition of Akt activity and reduction in cell migration. These findings reveal that, in the absence of androgens, ERß1 induces INPP4B to dampen AKT signaling. Since the endogenous ERß ligand, 3ß-Adiol, is lost upon long-term ADT, to obtain the beneficial effects of ERß1 on AKT signaling, an ERß agonist should be added along with ADT.


Asunto(s)
Receptor beta de Estrógeno/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Antagonistas de Andrógenos/metabolismo , Andrógenos/farmacología , Línea Celular Tumoral , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Humanos , Masculino , Células PC-3 , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Neoplasias de la Próstata/patología , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Transducción de Señal
2.
Int J Mol Sci ; 24(6)2023 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-36982940

RESUMEN

Triple Negative Breast Cancer (TNBC) has the worst prognosis among all breast cancers, and survival in patients with recurrence is rarely beyond 12 months due to acquired resistance to chemotherapy, which is the standard of care for these patients. Our hypothesis is that Estrogen Receptor ß1 (ERß1) increases response to chemotherapy but is opposed by ERß4, which it preferentially dimerizes with. The role of ERß1 and ERß4 in influencing chemotherapy sensitivity has never been studied before. CRISPR/CAS9 was used to truncate ERß1 Ligand Binding Domain (LBD) and knock down the exon unique to ERß4. We show that the truncated ERß1 LBD in a variety of mutant p53 TNBC cell lines, where ERß1 ligand dependent function was inactivated, had increased resistance to Paclitaxel, whereas the ERß4 knockdown cell line was sensitized to Paclitaxel. We further show that ERß1 LBD truncation, as well as treatment with ERß1 antagonist 2-phenyl-3-(4-hydroxyphenyl)-5,7-bis(trifluoromethyl)-pyrazolo[1,5-a] pyrimidine (PHTPP), leads to increase in the drug efflux transporters. Hypoxia Inducible Factors (HIFs) activate factors involved in pluripotency and regulate the stem cell phenotype, both in normal and cancer cells. Here we show that the ERß1 and ERß4 regulate these stem cell markers like SOX2, OCT4, and Nanog in an opposing manner; and we further show that this regulation is mediated by HIFs. We show the increase of cancer cell stemness due to ERß1 LBD truncation is attenuated when HIF1/2α is knocked down by siRNA. Finally, we show an increase in the breast cancer stem cell population due to ERß1 antagonist using both ALDEFLUORTM and SOX2/OCT4 response element (SORE6) reporters in SUM159 and MDA-MB-231 cell lines. Since most TNBC cancers are ERß4 positive, while only a small proportion of TNBC patients are ERß1 positive, we believe that simultaneous activation of ERß1 with agonists and inactivation of ERß4, in combination with paclitaxel, can be more efficacious and yield better outcome for chemotherapy resistant TNBC patients.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Humanos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Receptores de Estrógenos , Ligandos , Receptor beta de Estrógeno/genética , Receptor beta de Estrógeno/metabolismo , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Células Madre Neoplásicas/metabolismo , Línea Celular Tumoral
3.
Proc Natl Acad Sci U S A ; 111(31): 11443-8, 2014 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-25056967

RESUMEN

The sodium-coupled neutral amino acid transporter 2 (SNAT2) translocates small neutral amino acids into the mammary gland to promote cell proliferation during gestation. It is known that SNAT2 expression increases during pregnancy, and in vitro studies indicate that this transporter is induced by 17ß-estradiol. In this study, we elucidated the mechanism by which 17ß-estradiol regulates the transcription of SNAT2. In silico analysis revealed the presence of a potential estrogen response element (ERE) in the SNAT2 promoter. Reporter assays showed an increase in SNAT2 promoter activity when cotransfected with estrogen receptor alpha (ER-α) after 17ß-estradiol stimulation. Deletion of the ERE reduced estradiol-induced promoter activity by 63%. Additionally, EMSAs and supershift assays showed that ER-α binds to the SNAT2 ERE and that this binding competes with the interaction of ER-α with its consensus ERE. An in vivo ChIP assay demonstrated that the binding of ER-α to the SNAT2 promoter gradually increased in the mammary gland during gestation and that maximal binding occurred at the highest 17ß-estradiol serum concentration. Liquid chromatography-elevated energy mass spectrometry and Western blot analysis revealed that the SNAT2 ER-α-ERE complex contained poly(ADP-ribose) polymerase 1, Lupus Ku autoantigen protein p70, and glyceraldehyde 3-phosphate dehydrogenase (GAPDH) proteins and that the silencing of each of these proteins nearly abolished 17ß-estradiol-stimulated SNAT2 promoter activity. Nuclear levels of GAPDH increased progressively during gestation in the mammary gland, and GAPDH binding was nucleotide-specific for the SNAT2 ERE. Thus, this study provides new insights into how the mammary epithelium adapts to control amino acid uptake through the transcriptional regulation of the SNAT2 transporter via 17ß-estradiol.


Asunto(s)
Sistemas de Transporte de Aminoácidos/genética , Estradiol/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Sistema de Transporte de Aminoácidos A , Sistemas de Transporte de Aminoácidos/metabolismo , Animales , Antígenos Nucleares/metabolismo , Secuencia de Bases , Núcleo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Epitelio/metabolismo , Receptor alfa de Estrógeno/metabolismo , Femenino , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Células HeLa , Humanos , Autoantígeno Ku , Glándulas Mamarias Animales/metabolismo , Datos de Secuencia Molecular , Fragmentos de Péptidos/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Embarazo , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Ratas , Elementos de Respuesta/genética
4.
Exp Cell Res ; 331(1): 1-10, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25499972

RESUMEN

We have previously demonstrated that the anti-apoptotic protein BAD is expressed in normal human breast tissue and shown that BAD inhibits expression of cyclin D1 to delay cell-cycle progression in breast cancer cells. Herein, expression of proteins in breast tissues was studied by immunohistochemistry and results were analyzed statistically to obtain semi-quantitative data. Biochemical and functional changes in BAD-overexpressing MCF7 breast cancer cells were evaluated using PCR, reporter assays, western blotting, ELISA and extracellular matrix invasion assays. Compared to normal tissues, Grade II breast cancers expressed low total/phosphorylated forms of BAD in both cytoplasmic and nuclear compartments. BAD overexpression decreased the expression of ß-catenin, Sp1, and phosphorylation of STATs. BAD inhibited Ras/MEK/ERK and JNK signaling pathways, without affecting the p38 signaling pathway. Expression of the metastasis-related proteins, MMP10, VEGF, SNAIL, CXCR4, E-cadherin and TlMP2 was regulated by BAD with concomitant inhibition of extracellular matrix invasion. Inhibition of BAD by siRNA increased invasion and Akt/p-Akt levels. Clinical data and the results herein suggest that in addition to the effect on apoptosis, BAD conveys anti-metastatic effects and is a valuable prognostic marker in breast cancer.


Asunto(s)
Movimiento Celular , Regulación hacia Abajo , Proteína Letal Asociada a bcl/metabolismo , Western Blotting , Proliferación Celular , Ensayo de Inmunoadsorción Enzimática , Transición Epitelial-Mesenquimal , Femenino , Citometría de Flujo , Humanos , Técnicas para Inmunoenzimas , Células MCF-7 , Fosforilación , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción STAT/genética , Factores de Transcripción STAT/metabolismo , Células Tumorales Cultivadas , Proteína Letal Asociada a bcl/antagonistas & inhibidores , beta Catenina/genética , beta Catenina/metabolismo
5.
Mol Cell Proteomics ; 10(1): M110.002170, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20884965

RESUMEN

The present study addresses, by transcriptomics and quantitative stable isotope labeling by amino acids in cell culture (SILAC)-based proteomics, the estrogen receptor α (ERα) and ß (ERß)-mediated effects on gene and protein expression in T47D breast cancer cells exposed to the phytoestrogen genistein. Using the T47D human breast cancer cell line with tetracycline-dependent ERß expression (T47D-ERß), the effect of a varying intracellular ERα/ERß ratio on genistein-induced gene and protein expression was characterized. Results obtained reveal that in ERα-expressing T47D-ERß cells with inhibited ERß expression genistein induces transcriptomics and proteomics signatures pointing at rapid cell growth and migration by dynamic activation of cytoskeleton remodeling. The data reveal an interplay between integrins, focal adhesion kinase, CDC42, and actin cytoskeleton signaling cascades, occurring upon genistein treatment, in the T47D-ERß breast cancer cells with low levels of ERα and no expression of ERß. In addition, data from our study indicate that ERß-mediated gene and protein expression counteracts ERα-mediated effects because in T47D-ERß cells expressing ERß and exposed to genistein transcriptomics and proteomics signatures pointing at a clear down-regulation of cell growth and induction of cell cycle arrest and apoptosis were demonstrated. These results suggest that ERß decreases cell motility and metastatic potential as well as cell survival of the breast cancer cell line. It is concluded that the effects of genistein on proteomics and transcriptomics end points in the T47D-ERß cell model are comparable with those reported previously for estradiol with the ultimate estrogenic effect being dependent on the relative affinity for both receptors and on the receptor phenotype (ERα/ERß ratio) in the cells or tissue of interest.


Asunto(s)
Neoplasias de la Mama/metabolismo , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Perfilación de la Expresión Génica/métodos , Genisteína/farmacología , Fitoestrógenos/farmacología , Proteómica/métodos , Neoplasias de la Mama/genética , Línea Celular Tumoral , Análisis por Conglomerados , Ensayo de Inmunoadsorción Enzimática , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Proteoma/metabolismo , Transducción de Señal/efectos de los fármacos
6.
Breast Cancer Res Treat ; 127(2): 417-27, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20623183

RESUMEN

Estrogen receptor beta (ERß) inhibits proliferation in different cellular systems by regulating components of the cell cycle machinery. Eukaryotic cells respond to DNA damage by arresting in G1, S, or G2 phases of the cell cycle to initiate DNA repair. Most tumor cells due to disruptions in the p53-dependent G1 pathway are dependent on S-phase and G2/M checkpoints to maintain genomic integrity in response to DNA damage. We report that induction of ERß expression causes abrogation of the S-phase, and the Chk1/Cdc25C-mediated G2/M checkpoints after cisplatin and doxorubicin exposure in p53-defective breast cancer cells but not in p53 wild-type mammary cells. This impairment of DNA damage response that involves BRCA1 downregulation and caspase-2 activation results in mitotic catastrophe and decreased cancer cell survival. These results indicate that in cancers where p53 is defective, assessment of the presence of ERß may be of predictive value for the successful response to chemotherapy.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Receptor beta de Estrógeno/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteína p53 Supresora de Tumor/deficiencia , Antineoplásicos/farmacología , Proteína BRCA1/metabolismo , Ciclo Celular/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Cisplatino/farmacología , Daño del ADN/genética , Células HeLa , Humanos , Ligandos , Mitosis/efectos de los fármacos , Mitosis/genética , Proteína p53 Supresora de Tumor/genética
7.
Proc Natl Acad Sci U S A ; 105(7): 2604-9, 2008 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-18272478

RESUMEN

In this article, we have applied the ChIP-on-chip approach to pursue a large scale identification of ERalpha- and ERbeta-binding DNA regions in intact chromatin. We show that there is a high degree of overlap between the regions identified as bound by ERalpha and ERbeta, respectively, but there are also regions that are bound by ERalpha only in the presence of ERbeta, as well as regions that are selectively bound by either receptor. Analysis of bound regions shows that regions bound by ERalpha have distinct properties in terms of genome landscape, sequence features, and conservation compared with regions that are bound by ERbeta. ERbeta-bound regions are, as a group, located more closely to transcription start sites. ERalpha- and ERbeta-bound regions differ in sequence properties, with ERalpha-bound regions having an overrepresentation of TA-rich motifs including forkhead binding sites and ERbeta-bound regions having a predominance of classical estrogen response elements (EREs) and GC-rich motifs. Differences in the properties of ER bound regions might explain some of the differences in gene expression programs and physiological effects shown by the respective estrogen receptors.


Asunto(s)
ADN/metabolismo , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Genoma/genética , Anticuerpos/inmunología , Secuencia de Bases , Sitios de Unión , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Receptor beta de Estrógeno/inmunología , Humanos , Unión Proteica
8.
J Mol Endocrinol ; 68(1): R1-R9, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34546964

RESUMEN

Summary: After the discovery of ERß, a novel role for dihydrotestosterone (DHT) in estrogen signaling was revealed. Instead of just being a better androgen, DHT was found to be a precursor of the ERß agonist 5α-androstane-3ß, 17ß-diol (3ßAdiol), an estrogen which does not require aromatase for its synthesis. ERß was found to oppose androgen signaling and thus is a potential target for treatment of prostate cancer. ERß was also found to have effects that were independent of androgen signaling, particularly in the CNS. Although in rodent models of neurodegenerative diseases (Parkinson's disease, multiple sclerosis, and Alzheimer's disease), ERß agonists are very effective in relieving symptoms and improving pathologies, this has not proven to be the case in humans. In this review we will focus on the main differences in ERß signaling between rodents and humans and will make the point that a very important difference between the two species is in the splice variants which are expressed in humans and not rodents. The main conclusion at this point is that before we think of using ERß agonists clinically, much more work on ERß signaling in the human or in primates needs to be done.


Asunto(s)
Receptor beta de Estrógeno/genética , Receptor beta de Estrógeno/metabolismo , Animales , Descubrimiento de Drogas , Estrógenos/metabolismo , Regulación de la Expresión Génica , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Ligandos , Investigación/historia , Transducción de Señal
9.
J Cell Physiol ; 222(1): 156-67, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19780039

RESUMEN

Estrogen effects on mammary gland development and differentiation are mediated by two receptors (ERalpha and ERbeta). Estrogen-bound ERalpha induces proliferation of mammary epithelial and cancer cells, while ERbeta is important for maintenance of the differentiated epithelium and inhibits proliferation in different cell systems. In addition, the normal breast contains higher ERbeta levels compared to the early stage breast cancers, suggesting that loss of ERbeta could be important in cancer development. Analysis of ERbeta-/- mice has consistently revealed reduced expression of cell adhesion proteins. As such, ERbeta is a candidate modulator of epithelial homeostasis and metastasis. Consequently, the aim of this study was to analyze estrogenic effects on adhesion of breast cancer cells expressing ERalpha and ERbeta. As ERbeta is widely found in breast cancer but not in cell lines, we used ERalpha positive T47-D and MCF-7 human breast cancer cells to generate cells with inducible ERbeta expression. Furthermore, the colon cancer cell lines SW480 and HT-29 were also used. Integrin alpha1 mRNA and protein levels increased following ERbeta expression. Integrin beta1-the unique partner for integrin alpha1-increased only at the protein level. ERbeta expression enhanced the formation of vinculin containing focal complexes and actin filaments, indicating a more adhesive potential. This was confirmed by adhesion assays where ERbeta increased adhesion to different extracellular matrix proteins, mostly laminin. In addition, ERbeta expression was associated to less cell migration. These results indicate that ERbeta affects integrin expression and clustering and consequently modulates adhesion and migration of breast cancer cells.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Receptor beta de Estrógeno/metabolismo , Integrina alfa1/metabolismo , Integrina beta1/metabolismo , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/metabolismo , Neoplasias de la Mama/genética , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Membrana Celular/metabolismo , Movimiento Celular/efectos de los fármacos , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Adhesiones Focales/efectos de los fármacos , Adhesiones Focales/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Integrina alfa1/genética , Integrina beta1/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Tetraciclina/farmacología , Factores de Tiempo , Vinculina/metabolismo
10.
Breast Cancer Res Treat ; 120(2): 317-26, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19322650

RESUMEN

We have previously shown that the anti-proliferative effect of retinoic acid in human breast cancer cell line MCF-7 is dependent on HES-1 expression. Here we show that retinoic acid induces HES-1 expression via upregulation of transcription factor SOX9. By expressing a dominant negative form of SOX9, disrupting endogenous SOX9 activity, the retinoic acid-induced HES-1 mRNA expression was inhibited. We found an enhancer regulating HES-1 expression: two SOX9 binding sites upstream of the HES-1 gene that were capable of binding SOX9 in vitro. By performing chromatin immunoprecipitation, we showed that SOX9 binding to the HES-1 enhancer was induced by retinoic acid in vivo. In reporter assays, transfection of a SOX9 expression plasmid increased the activity of the HES-1 enhancer. The enhancer responded to retinoic acid; furthermore, the expression of a dominant negative SOX9 abolished this response. Taken together, we present here a novel transcriptional mechanism in regulating hormone-dependent cancer cell proliferation.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Neoplasias de la Mama/genética , Regulación Neoplásica de la Expresión Génica/genética , Proteínas de Homeodominio/biosíntesis , Factor de Transcripción SOX9/metabolismo , Antineoplásicos/farmacología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/efectos de los fármacos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Western Blotting , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Ensayo de Cambio de Movilidad Electroforética , Elementos de Facilitación Genéticos/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas de Homeodominio/efectos de los fármacos , Proteínas de Homeodominio/genética , Humanos , Inmunoprecipitación , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción SOX9/efectos de los fármacos , Factor de Transcripción SOX9/genética , Factor de Transcripción HES-1 , Transfección , Tretinoina/farmacología
11.
Phys Rev Lett ; 104(25): 256804, 2010 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-20867409

RESUMEN

We analyze the dynamics of the helical edge modes of a quantum spin Hall state in the presence of a spatially nonuniform Rashba spin-orbit (SO) interaction. A randomly fluctuating Rashba SO coupling is found to open a scattering channel which causes localization of the edge modes for a weakly screened electron-electron (e-e) interaction. A periodic modulation of the SO coupling, with a wave number commensurate with the Fermi momentum, makes the edge insulating already at intermediate strengths of the e-e interaction. We discuss implications for experiments on edge state transport in a HgTe quantum well.

12.
PLoS One ; 15(5): e0226057, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32413024

RESUMEN

Estrogen receptor ß (ERß) was first identified in the rodent prostate and is abundantly expressed in human and rodent prostate epithelium, stroma, immune cells and endothelium of the blood vessels. In the prostates of mice with inactivated ERß, mutant phenotypes include epithelial hyperplasia and increased expression of androgen receptor (AR)-regulated genes, most of which are also upregulated in prostate cancer (PCa). ERß is expressed in both basal and luminal cells in the prostate while AR is expressed in luminal but not in the basal cell layer which harbors the prostate stem cells. To investigate the mechanisms of action of ERß and its potential cross-talk with AR, we used RNA-seq to study the effects of estradiol or the synthetic ligand, LY3201, in AR-positive LNCaP PCa cells which had been engineered to express ERß. Transcriptomic analysis indicated relatively few changes in gene expression with ERß overexpression, but robust responses following ligand treatments. There is significant overlap of responsive genes between the two ligands, estradiol and LY3201 as well as ligand-specific alterations. Gene set analysis of down-regulated genes identified an enrichment of androgen-responsive genes, such as FKBP5, CAMKK2, and TBC1D4. Consistently, AR transcript, protein levels, and transcriptional activity were down-regulated following ERß activation. In agreement with this, we find that the phosphorylation of the CAMKK2 target, AMPK, was repressed by ligand-activated ERß. These findings suggest that ERß-mediated signaling pathways are involved in the negative regulation of AR expression and activity, thus supporting a tumor suppressive role for ERß in PCa.


Asunto(s)
Receptor beta de Estrógeno/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Benzopiranos/farmacología , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/genética , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Estradiol/farmacología , Receptor beta de Estrógeno/agonistas , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Humanos , Masculino , Receptores Androgénicos/genética , Transducción de Señal , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/metabolismo
13.
Cancer Discov ; 10(4): 608-625, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32046984

RESUMEN

A hallmark of pancreatic ductal adenocarcinoma (PDAC) is an exuberant stroma comprised of diverse cell types that enable or suppress tumor progression. Here, we explored the role of oncogenic KRAS in protumorigenic signaling interactions between cancer cells and host cells. We show that KRAS mutation (KRAS*) drives cell-autonomous expression of type I cytokine receptor complexes (IL2rγ-IL4rα and IL2rγ-IL13rα1) in cancer cells that in turn are capable of receiving cytokine growth signals (IL4 or IL13) provided by invading Th2 cells in the microenvironment. Early neoplastic lesions show close proximity of cancer cells harboring KRAS* and Th2 cells producing IL4 and IL13. Activated IL2rγ-IL4rα and IL2rγ-IL13rα1 receptors signal primarily via JAK1-STAT6. Integrated transcriptomic, chromatin occupancy, and metabolomic studies identified MYC as a direct target of activated STAT6 and that MYC drives glycolysis. Thus, paracrine signaling in the tumor microenvironment plays a key role in the KRAS*-driven metabolic reprogramming of PDAC. SIGNIFICANCE: Type II cytokines, secreted by Th2 cells in the tumor microenvironment, can stimulate cancer cell-intrinsic MYC transcriptional upregulation to drive glycolysis. This KRAS*-driven heterotypic signaling circuit in the early and advanced tumor microenvironment enables cooperative protumorigenic interactions, providing candidate therapeutic targets in the KRAS* pathway for this intractable disease.


Asunto(s)
Citocinas/metabolismo , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Animales , Reprogramación Celular/genética , Humanos , Ratones , Oncogenes , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Transfección , Microambiente Tumoral
14.
Breast Cancer Res ; 11(6): R79, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19891787

RESUMEN

INTRODUCTION: Hes-6 is a member of the basic helix-loop-helix (bHLH) family of transcription factors, and its overexpression has been reported in metastatic cancers of different origins. Hes-6 has been described as an inhibitor of Hes-1 during neuronal development, although its function in cancer is not known. In this study, we investigated the function of Hes-6 in breast cancer and tested the hypothesis that Hes-6 enhances breast cancer cell proliferation and is regulated by estrogen. METHODS: To investigate the function of Hes-6, T47D cells stably expressing Hes-6 were generated by lentiviral transduction, and conversely, siRNA also was used to knock down Hes-6 expression in breast cancer cells. The Hes-6-expressing T47D cells were transplanted into immunodeficient mice to study effects on tumor growth. RESULTS: We found that Hes-6 expression was significantly higher in the high-grade, estrogen receptor (ER)alpha-negative SKBR3 and MDA-MB-231 cells compared with the ERalpha-positive, non-metastasizing T47D and MCF-7 breast carcinoma cells. Moreover, the level of Hes-6 mRNA was 28 times higher in breast cancer samples compared with normal breast samples. In Hes-6-expressing T47D cells, Hes-6 ectopic expression was shown to stimulate cell proliferation in vitro as well as breast tumor growth in xenografts. Moreover, expression of Hes-6 resulted in induction of E2F-1, a crucial target gene for the transcriptional repressor Hes-1. Consistently, silencing of Hes-6 by siRNA resulted in downregulation of E2F-1 expression, whereas estrogen treatment caused induction of Hes-6 and downstream targets hASH-1 and E2F-1 in MCF-7 cells. CONCLUSIONS: Together, the data suggest that Hes-6 is a potential oncogene overexpressed in breast cancer, with a tumor-promoting and proliferative function. Furthermore, Hes-6 is a novel estrogen-regulated gene in breast cancer cells. An understanding of the role and regulation of Hes-6 could provide insights into estrogen signaling and endocrine resistance in breast cancer and, hence, could be important for the development of novel anticancer drugs.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Neoplasias de la Mama/patología , Estradiol/farmacología , Proteínas de Homeodominio/antagonistas & inhibidores , Proteínas Represoras/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Procesos de Crecimiento Celular/efectos de los fármacos , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Factor de Transcripción E2F1/antagonistas & inhibidores , Factor de Transcripción E2F1/metabolismo , Receptor alfa de Estrógeno/biosíntesis , Receptor alfa de Estrógeno/metabolismo , Femenino , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Ratones SCID , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Proteínas Represoras/biosíntesis , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factor de Transcripción HES-1 , Transfección , Trasplante Heterólogo
15.
Cancer Res ; 67(8): 3955-62, 2007 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-17440111

RESUMEN

Estrogens, by binding to and activating two estrogen receptors (ERalpha and ERbeta), are critically involved in the development of the mammary gland and breast cancer. An isoform of ERbeta, ERbeta2 (also called ERbetacx), with an altered COOH-terminal region, is coexpressed with ERalpha in many human breast cancers. In this study, we generated a stable cell line from MCF7 breast cancer cells expressing an inducible version of ERbeta2, along with endogenous ERalpha, and examined the effects of ERbeta2 on the ERalpha protein levels and function. We showed that ERbeta2 inhibited ERalpha-mediated transactivation via estrogen response element and activator protein-1 sites of reporter constructs as well as the endogenous genes pS2 and MMP-1. Chromatin immunoprecipitation assays revealed that ERbeta2 expression caused a significant reduction in the recruitment of ERalpha to both the pS2 and MMP-1 promoters. Furthermore, ERbeta2 expression induced proteasome-dependent degradation of ERalpha. The inhibitory effects of ERbeta2 on ERalpha activity were further confirmed in HEK293 cells that lack functional endogenous ERs. We also showed that ERbeta2 can interact with ERalpha both in vitro and in mammalian cells, which is compatible with a model where ERbeta2/ERalpha heterodimers are targeted to the proteasome. Finally, in human breast cancer samples, we observed that expression of ERbeta2 significantly correlated with ERalpha-negative phenotype. Our data suggest that ERbeta2 could influence ERalpha-mediated effects relevant for breast cancer development, including hormone responsiveness.


Asunto(s)
Neoplasias de la Mama/genética , Receptor alfa de Estrógeno/antagonistas & inhibidores , Receptor beta de Estrógeno/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Regulación hacia Abajo , Receptor alfa de Estrógeno/biosíntesis , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/biosíntesis , Receptor beta de Estrógeno/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Regiones Promotoras Genéticas , Complejo de la Endopetidasa Proteasomal/metabolismo , Activación Transcripcional , Transfección
16.
J Steroid Biochem Mol Biol ; 191: 105312, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30995525

RESUMEN

ERbeta (ERß) celebrated its 20th birthday in 2016 and although the overwhelming data in the literature indicate a role for this receptor in the control of epithelial proliferation, neurodegeneration and immune function, no ERß agonists have yet made it to the clinics. This is the situation, despite the fact that very good safe ERß agonists have been synthesized and at least one has been donated to the NIH for distribution to researchers, who want to study its possible clinical use. Clinical trials are ongoing for the use of ERß agonists in prostate cancer and schizophrenia but even today reviewers of our grants still make comments like "The grant is excellent except that the focus of the grant is ERß". There are multiple reasons for the non-acceptance of the value of ERß and in this paper we will discuss issues raised by labs which do not support a role for ERß in physiology or pathology.


Asunto(s)
Receptor beta de Estrógeno/metabolismo , Animales , Anticuerpos/inmunología , Descubrimiento de Drogas , Receptor beta de Estrógeno/agonistas , Receptor beta de Estrógeno/genética , Receptor beta de Estrógeno/inmunología , Expresión Génica , Humanos , Inmunidad , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/metabolismo , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Isoformas de Proteínas/metabolismo
17.
Cancer Res ; 66(23): 11207-13, 2006 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-17145865

RESUMEN

Estrogens, which are stimulators of growth of both the normal breast and malignant breast, mediate their effects through two estrogen receptors (ER), namely ERalpha and ERbeta. ERalpha mediates the proliferative effect of estrogen in breast cancer cells, whereas ERbeta seems to be antiproliferative. We engineered ERalpha-positive T47D breast cancer cells to express ERbeta in a Tet-Off-regulated manner. These cells were then injected orthotopically into severe combined immunodeficient mice, and the growth of the resulting tumors was compared with tumors resulting from injecting the parental T47D cells that do not express ERbeta. The presence of ERbeta resulted in a reduction in tumor growth. Comparison of the ERbeta-expressing and non-ERbeta-expressing tumors revealed that the expression of ERbeta caused a reduction in the number of intratumoral blood vessels and a decrease in expression of the proangiogenic factors vascular endothelial growth factor (VEGF) and platelet-derived growth factor beta (PDGFbeta). In cell culture, with the Tet-Off-regulated ERbeta-expressing cells, expression of ERbeta decreased expression of VEGF and PDGFbeta mRNA under normoxic as well as hypoxic conditions and reduced secreted VEGF and PDGFbeta proteins in cell culture medium. Transient transfection assays with 1,026 bp VEGF and 1,006 bp PDGFbeta promoter constructs revealed a repressive effect of ERbeta at the promoter level of these genes. Taken together, these data show that introduction of ERbeta into malignant cells inhibits their growth and prevents tumor expansion by inhibiting angiogenesis.


Asunto(s)
Proliferación Celular , Receptor beta de Estrógeno/genética , Neoplasias Mamarias Experimentales/patología , Neovascularización Patológica/patología , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Western Blotting , Hipoxia de la Célula , Línea Celular Tumoral , Receptor beta de Estrógeno/análisis , Receptor beta de Estrógeno/fisiología , Femenino , Expresión Génica/genética , Terapia Genética , Humanos , Inmunohistoquímica , Antígeno Ki-67/análisis , Luciferasas/genética , Luciferasas/metabolismo , Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/terapia , Ratones , Ratones SCID , Neovascularización Patológica/metabolismo , Neovascularización Patológica/fisiopatología , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-sis/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factores de Tiempo , Transfección , Factor A de Crecimiento Endotelial Vascular/genética
18.
Oncotarget ; 9(91): 36273-36288, 2018 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-30555629

RESUMEN

Chemotherapy resistant prostate cancer is a major clinical problem. When the prostate cancer has become androgen deprivation resistant, one of the few treatment regimens left is chemotherapy. There is a strong connection between a cancer's stem cell like characteristics and drug resistance. By performing RNA-seq we observed several factors associated with stem cells being strongly up-regulated by the estrogen receptor ß variants, ß2 and ß5. In addition, most of these factors were also up-regulated by hypoxia. One mechanism of chemotherapy resistance was expression of the hypoxia-regulated, drug transporter genes, where especially ABCG2 and MDR1 were shown to be expressed in recurrent prostate cancer and to cause chemotherapy resistance by efficiently transporting drugs like docetaxel out of the cells. Another mechanism was expression of the hypoxia-regulated Notch3 gene, which causes chemotherapy resistance in urothelial carcinoma, although the mechanism is unknown. It is well known that hypoxic signaling is involved in increasing chemotherapy resistance. Regulation of the hypoxic factors, HIF-1α and HIF-2α is very complex and extends far beyond hypoxia itself. We have recently shown that two of the estrogen receptor ß variants, estrogen receptor ß2 and ß5, bind to and stabilize both HIF-1α and HIF-2α proteins leading to expression of HIF target genes. This study suggests that increased expression of the estrogen receptor ß variants, ß2 and ß5, could be involved in development of a cancer's stem cell characteristics and chemotherapy resistance, indicating that targeting these factors could prevent or reverse chemotherapy resistance and cancer stem cell expansion.

19.
Oncotarget ; 9(15): 12201-12211, 2018 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-29552303

RESUMEN

Triple negative breast cancer (TNBC) still remains a challenge to treat in the clinic due to a lack of good targets for treatment. Although TNBC lacks expression of ERα, the expression of ERß and its variants are detected quite frequently in this cancer type and can represent an avenue for treatment. We show that two of the variants of ERß, namely ERß2 and ERß5, control aggressiveness of TNBC by regulating hypoxic signaling through stabilization of HIF-1α. RNA-seq of patient derived xenografts (PDX) from TNBC shows expression of ERß2, ERß4 and ERß5 variants in more than half of the samples. Furthermore, expression of ERß4 in the immortalized, normal mammary epithelial cell line MCF-10A that is resistant to tumorsphere formation caused transformation and development of tumorspheres. By contrast, ERß1, ERß2 or ERß5 were unable to support tumorsphere formation. We have previously shown that all variants except ERß1 stabilize HIF-1α but only ERß4 appears to have the ability to transform normal mammary epithelial cells, pointing towards a unique property of ERß4. We propose that ERß variants may be good diagnostic tools and also serve as novel targets for treatment of breast cancer.

20.
Breast Cancer Res ; 9(2): R25, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17428314

RESUMEN

INTRODUCTION: The impact of interactions between the two estrogen receptor (ER) subtypes, ERalpha and ERbeta, on gene expression in breast cancer biology is not clear. The goal of this study was to examine transcriptomic alterations in cancer cells co-expressing both receptors and the association of gene expression signatures with disease outcome. METHODS: Transcriptional effects of ERbeta overexpression were determined in a stably transfected cell line derived from ERalpha-positive T-47D cells. Microarray analysis was carried out to identify differential gene expression in the cell line, and expression of key genes was validated by quantitative polymerase chain reaction. Microarray and clinical data from patient samples were then assessed to determine the in vivo relevance of the expression profiles observed in the cell line. RESULTS: A subset of 14 DNA replication and cell cycle-related genes was found to be specifically downregulated by ERbeta. Expression profiles of four genes, CDC2, CDC6, CKS2, and DNA2L, were significantly inversely correlated with ERbeta transcript levels in patient samples, consistent with in vitro observations. Kaplan-Meier analysis revealed better disease outcome for the patient group with an expression signature linked to higher ERbeta expression as compared to the lower ERbeta-expressing group for both disease-free survival (p = 0.00165) and disease-specific survival (p = 0.0268). These findings were further validated in an independent cohort. CONCLUSION: Our findings revealed a transcriptionally regulated mechanism for the previously described growth inhibitory effects of ERbeta in ERalpha-positive breast tumor cells and provide evidence for a functional and beneficial impact of ERbeta in primary breast tumors.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Receptor beta de Estrógeno/fisiología , Regulación Neoplásica de la Expresión Génica , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Supervivencia sin Enfermedad , Factores de Transcripción E2F/metabolismo , Receptor beta de Estrógeno/metabolismo , Femenino , Humanos , Familia de Multigenes , Análisis de Secuencia por Matrices de Oligonucleótidos , Transcripción Genética , Transfección , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA