Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 110(16): E1470-9, 2013 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-23533274

RESUMEN

Lung cancer is a heterogeneous disease at both clinical and molecular levels, posing conceptual and practical bottlenecks in defining key pathways affecting its initiation and progression. Molecules with a central role in lung carcinogenesis are likely to be targeted by multiple deregulated pathways and may have prognostic, predictive, and/or therapeutic value. Here, we report that Tumor Progression Locus 2 (TPL2), a kinase implicated in the regulation of innate and adaptive immune responses, fulfils a role as a suppressor of lung carcinogenesis and is subject to diverse genetic and epigenetic aberrations in lung cancer patients. We show that allelic imbalance at the TPL2 locus, up-regulation of microRNA-370, which targets TPL2 transcripts, and activated RAS (rat sarcoma) signaling may result in down-regulation of TPL2 expression. Low TPL2 levels correlate with reduced lung cancer patient survival and accelerated onset and multiplicity of urethane-induced lung tumors in mice. Mechanistically, TPL2 was found to antagonize oncogene-induced cell transformation and survival through a pathway involving p53 downstream of cJun N-terminal kinase (JNK) and be required for optimal p53 response to genotoxic stress. These results identify multiple oncogenic pathways leading to TPL2 deregulation and highlight its major tumor-suppressing function in the lung.


Asunto(s)
Transformación Celular Neoplásica/inmunología , Regulación Neoplásica de la Expresión Génica/inmunología , Neoplasias Pulmonares/fisiopatología , Quinasas Quinasa Quinasa PAM/metabolismo , MicroARNs/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas ras/metabolismo , Animales , Secuencia de Bases , Transformación Celular Neoplásica/genética , Metilación de ADN , Análisis Mutacional de ADN , Cartilla de ADN/genética , Citometría de Flujo , Humanos , Immunoblotting , Neoplasias Pulmonares/inmunología , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/inmunología , Ratones , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/inmunología , Análisis de Secuencia de ADN
2.
J Immunol ; 189(10): 4748-58, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23071284

RESUMEN

The TNF superfamily ligands APRIL and BAFF bind with different affinity to two receptors, BCMA and TACI, and induce cell survival and/or proliferation, whereas BAFF also binds specifically to BAFFR. These molecules were considered specific for the immune system. Recently, however, they were also found in epithelial and mesenchymal noncancerous and cancerous tissues and cell lines. In this article, we report that hepatocellular carcinoma (HCC) cell lines HepG2 and Hep3B and HCC specimens express APRIL and BAFF and their receptors BCMA and BAFFR, but not TACI; APRIL/BCMA is enhanced in HCC, compared with normal liver tissue. In contrast to previous reports, APRIL binding to BCMA decreases cell proliferation by inducing G(2)/M cell cycle arrest, whereas BAFF has no effect on cell growth. HCC cells therefore represent a rare system in which these two ligands (APRIL and BAFF) exert a differential effect and may serve as a model for specific APRIL/BCMA actions. We show that the effect of APRIL is mediated via BCMA, which does not activate the classical NF-κB pathway, whereas it induces a novel signaling pathway, which involves JNK2 phosphorylation, FOXO3A activation, and GADD45 transcription. In addition, JNK2 mediates the phosphorylation of Akt, which is activated but does not participate in the antiproliferative effect of APRIL. Furthermore, transcriptome analysis revealed that APRIL modifies genes specifically related to cell cycle modulation, including MCM2/4/5/6, CDC6, PCNA, and POLE2. Our data, therefore, identify a novel APRIL/BCMA signaling pathway in HCC and suggest that APRIL could have a pleiotropic role in tumor biology.


Asunto(s)
Antígeno de Maduración de Linfocitos B/inmunología , Proteínas de Ciclo Celular/inmunología , Proteínas de Unión al ADN/inmunología , Factores de Transcripción Forkhead/inmunología , Puntos de Control de la Fase G2 del Ciclo Celular/inmunología , Hígado/inmunología , Puntos de Control de la Fase M del Ciclo Celular/inmunología , MAP Quinasa Quinasa 7/inmunología , Proteínas Nucleares/inmunología , Factores de Transcripción/inmunología , Factor Activador de Células B/genética , Factor Activador de Células B/inmunología , Factor Activador de Células B/metabolismo , Antígeno de Maduración de Linfocitos B/genética , Antígeno de Maduración de Linfocitos B/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Puntos de Control de la Fase G2 del Ciclo Celular/genética , Células Hep G2 , Humanos , Hígado/citología , Puntos de Control de la Fase M del Ciclo Celular/genética , MAP Quinasa Quinasa 7/genética , MAP Quinasa Quinasa 7/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosforilación/genética , Fosforilación/inmunología , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/inmunología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Unión Proteica , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética/genética , Transcripción Genética/inmunología
3.
J Cell Biochem ; 112(12): 3786-96, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21826705

RESUMEN

Recently, our knowledge on estrogen receptor alpha (ERα) functions and fate has progressed: ERα enters in repeated transcription-modulating cycles (nucleus/cytoplasm/membrane trafficking processes and proteasomal degradation) that are governed by specific protein-protein interactions. Receptor fragments, especially those resulting from the proteolysis of its ligand binding domain, as well as corresponding synthetic peptides, have been studied with respect to their estrogenic/antiestrogenic potency. A peptide, corresponding to the human ERα P(295) -T(311) sequence (ERα17p) has been shown to alter breast cancer cell fate, triggering proliferation, or apoptosis. The aim of this work was to explore the effect of ERα17p on breast cancer cell migration and actin cytoskeleton dynamics and further analyze the mechanism of its membrane action. We show that ERα17p increases (MCF-7 and SK-BR-3 cells) or decreases (T47D and MDA-MB-231 cells) migration of breast cancer cells, in an ERα-independent manner, by mechanism(s) depending on Rho/ROCK and PI3K/Akt signaling pathways. Moreover, the peptide enhances the association of both estrogens and androgens to membranes and modifies cell migration, induced by E(2) -BSA. Additionally, initial evidence of a possible agonistic action of the peptide on GPR30 is also provided. ERα17p can be considered as a cell migration-modulator and could therefore constitute a therapeutic challenge, even in anti-estrogen-resistant tumors.


Asunto(s)
Actinas/metabolismo , Neoplasias de la Mama/patología , Citoesqueleto/metabolismo , Receptor alfa de Estrógeno/fisiología , Metástasis de la Neoplasia/patología , Biopolímeros/metabolismo , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Receptor alfa de Estrógeno/química , Receptor alfa de Estrógeno/metabolismo , Femenino , Humanos , Unión Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
4.
J Pharmacol Exp Ther ; 337(1): 24-32, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21205921

RESUMEN

Prostate cancer is the most common malignancy among men in Western societies, and current therapeutic approaches are evolving to manage growth, recurrence, and mortality neoplasia. Membrane androgen receptors (mARs) have been characterized in human prostate cancer, being preferentially expressed in tumor rather than benign gland areas. Furthermore, mAR agonists (protein-conjugated testosterone) decrease in vitro prostate cancer cell growth and induce apoptosis, whereas in vivo they regress growth of tumor xenografts alone or in combination with taxane drugs. In this respect, targeting mARs might be a novel therapeutic approach in prostate cancer. In our search for new small-molecule ligands of mAR, we report that flavanol dimers B1-B4 (oligomeric procyanidins) decrease in vitro growth of the androgen-sensitive (LnCaP) and androgen-resistant (DU145) human prostate cancer cell lines in the following order: B3 = B4 > B2 ≫ B1 (LnCaP) and B2 ≫ B3 = B4 ≫ B1 (DU145). Some of these analogs were previously shown to trigger signaling cascades similar to testosterone-bovine serum albumin (BSA) conjugate. Galloylation does not confer an additional advantage; however, oleylation increases the dimers' antiproliferative potency by a factor of 100. In addition, we report that B2, oleylated or not, displaces testosterone from mARs with an IC(50) value at the nanomolar range and induces DU145 tumor xenograft regression by 50% (testosterone-BSA 40%). In this respect, oleylated B2 is a potent small-molecule agonist of mAR and could be a novel therapeutic agent for advanced prostate cancer, especially when taking into account the absence of androgenic actions and (liver) toxicity.


Asunto(s)
Extracto de Semillas de Uva/metabolismo , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Neoplasias Hormono-Dependientes/metabolismo , Proantocianidinas/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Testosterona/metabolismo , Animales , Sitios de Unión/fisiología , Bovinos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Extracto de Semillas de Uva/aislamiento & purificación , Extracto de Semillas de Uva/uso terapéutico , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Hormono-Dependientes/patología , Proantocianidinas/aislamiento & purificación , Proantocianidinas/fisiología , Proantocianidinas/uso terapéutico , Neoplasias de la Próstata/patología , Ensayos Antitumor por Modelo de Xenoinjerto
5.
J Immunol ; 183(9): 5948-56, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19828625

RESUMEN

Adipose tissue represents a rich source of multipotent stem cells. Mesenchymal cells, isolated from this source, can differentiate to other cell types in vitro and therefore can be used for a number of regenerative therapies. Our view of adipose tissue has recently changed, establishing adipocytes as new members of the immune system, as they produce a number of proinflammatory cytokines (such as IL-6 and TNFalpha and chemokines, in addition to adipokines (leptin, adiponectin, resistin) and molecules associated with the innate immune system. In this paper, we report the differential expression of TNF-superfamily members B cell activating factor of the TNF Family (BAFF), a proliferation inducing ligand (APRIL), and TNF-like weak inducer of apoptosis (TWEAK) in immature-appearing and mature adipocytes and in benign and malignant adipose tissue-derived tumors. These ligands act through their cognitive receptors, BAFF receptor, transmembrane activator and calcium signal-modulating cyclophilic ligand (TACI), B cell maturation Ag (BCMA), and fibroblast growth factor-inducible 14 (Fn14), which are also expressed in these cells. We further report the existence of functional BCMA, TACI, and Fn14 receptors and their ligands BAFF, APRIL, and TWEAK on adipose tissue-derived mesenchymal cells, their interaction modifying the rate of adipogenesis. Our data integrate BAFF, APRIL, and TWEAK and their receptors BCMA, TACI, and Fn14 as novel potential mediators of adipogenesis, in addition to their specific role in immunity, and define immature and mature adipocytes as source of immune mediators.


Asunto(s)
Adipocitos/inmunología , Adipocitos/metabolismo , Factor Activador de Células B/biosíntesis , Receptor del Factor Activador de Células B/biosíntesis , Antígeno de Maduración de Linfocitos B/biosíntesis , Receptores del Factor de Necrosis Tumoral/biosíntesis , Proteína Activadora Transmembrana y Interactiva del CAML/biosíntesis , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/biosíntesis , Factores de Necrosis Tumoral/biosíntesis , Adipocitos/citología , Adipocitos/patología , Tejido Adiposo/citología , Tejido Adiposo/inmunología , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Diferenciación Celular/inmunología , Células Cultivadas , Citocina TWEAK , Humanos , Receptor de TWEAK , Células Tumorales Cultivadas
6.
BMC Cancer ; 8: 76, 2008 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-18366696

RESUMEN

BACKGROUND: Recent studies suggest an association between chronic inflammation, modulating the tissue microenvironment, and tumor biology. Tumor environment consists of tumor, stromal and endothelial cells and infiltrating macrophages, T lymphocytes, and dendritic cells, producing an array of cytokines, chemokines and growth factors, accounting for a complex cell interaction and regulation of differentiation, activation, function and survival of tumor and surrounding cells, responsible for tumor progression and spreading or induction of antitumor immune responses and rejection. Tumor Necrosis Factor (TNF) family members (19 ligands and 29 receptors) represent a pleiotropic family of agents, related to a plethora of cellular events from proliferation and differentiation to apoptosis and tumor reduction. Among these members, BAFF and APRIL (CD257 and CD256 respectively) gained an increased interest, in view of their role in cell protection, differentiation and growth, in a number of lymphocyte, epithelial and mesenchymal structures. METHODS: We have assayed by immunohistochemistry 52 human breast cancer biopsies for the expression of BAFF and APRIL and correlated our findings with clinicopathological data and the evolution of the disease. RESULTS: BAFF was ubiquitely expressed in breast carcinoma cells, DCIS, normal-appearing glands and ducts and peritumoral adipocytes. In contrast, APRIL immunoreactive expression was higher in non-malignant as compared to malignant breast structures. APRIL but not BAFF immunoreactivity was higher in N+ tumors, and was inversely related with the grade of the tumors. Neither parameter was related to DFS or the OS of patients. CONCLUSION: Our data show, for the first time, an autocrine secretion of BAFF and APRIL from breast cancer cells, offering new perspectives for their role in neoplastic and normal breast cell biology and offering new perspectives for possible selective intervention in breast cancer.


Asunto(s)
Factor Activador de Células B/biosíntesis , Neoplasias de la Mama/metabolismo , Carcinoma Ductal de Mama/metabolismo , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/biosíntesis , Adulto , Anciano , Femenino , Humanos , Inmunohistoquímica , Persona de Mediana Edad
7.
Steroids ; 73(9-10): 953-60, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18249430

RESUMEN

The mode of action of steroid hormones has been extended in recent years. In addition to their classical nuclear action (acting as transcription factors), they can also regulate cell-signaling phosphorylation cascades and exert actions that are initiated at the membrane and which, in most cases, are rapid. Even though research in this field was intensified during the last decade the nature of the up-stream receptor targets that mediates these rapid non-genomic actions remains to be better established. However, it became obvious that steroid signaling is not uniform, with a variety of modes of rapid action being described. There are several studies speculating a classical steroid receptor involvement in the rapid effects of steroids, localized at the cytoplasmic membrane and mediating effects directly or indirectly, via interactions with specific membrane structures (estrogen receptor (ER) isoforms have been shown to localize in caveolae) and/or other membrane receptors (like growth factor receptor). In addition, there are reports that suggest the existence of a distinct receptor, associated to the plasma membrane, being different from the classical, intracellular one. Non-genomic/extranuclear actions of steroids have been described in a number of different normal or cancer tissues independently of the presence of classical nuclear steroid receptors. In the present work, we review briefly the identification and signaling events of membrane-initiated steroid (androgen and estrogen) action in breast and prostate cancer cell lines and clinical specimens. Furthermore, we discuss the interaction of cytokine/growth factor receptors with membrane-acting steroids and their potential clinical implications.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Próstata/metabolismo , Esteroides/metabolismo , Animales , Núcleo Celular/metabolismo , Citocinas/metabolismo , Femenino , Humanos , Masculino , Modelos Biológicos , Progestinas/metabolismo , Isoformas de Proteínas , Receptores de Estrógenos/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Transducción de Señal
8.
Cancer Epidemiol Biomarkers Prev ; 16(10): 2016-23, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17932349

RESUMEN

Autocrine/paracrine erythropoietin (EPO) action, promoting cell survival and mediated by its receptor (EPOR) in various solid tumors, including breast carcinoma, questions about the prognostic and therapeutic interest of this system. The expression of EPO/EPOR is steroid dependent in some tissues; however, a clear relationship of EPO/EPOR and steroid receptors in breast cancer has not been established thus far. Recently, the field of steroid receptors has expanded, including rapid effects mediated by membrane-associated receptors, regulating cell survival or apoptosis. The aim of this study was to evaluate EPO/EPOR and membrane-associated steroid receptor expression in breast carcinoma, in view of their prognostic significance, compared with other established markers [estrogen receptor (ER)-progesterone receptor (PR) status and Her2 expression] and hypoxia-induced factor 1 nuclear localization in 61 breast cancer specimens followed for

Asunto(s)
Neoplasias de la Mama/patología , Eritropoyetina/análisis , Proteínas de la Membrana/análisis , Receptor ErbB-2/análisis , Receptores Androgénicos/análisis , Receptores de Eritropoyetina/análisis , Receptores de Estrógenos/análisis , Receptores de Progesterona/análisis , Adulto , Anciano , Mama/patología , Neoplasias de la Mama/mortalidad , Núcleo Celular/patología , Transformación Celular Neoplásica/patología , Supervivencia sin Enfermedad , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular , Microscopía Fluorescente , Persona de Mediana Edad , Proteínas Mitocondriales , Proteínas de Neoplasias/análisis , Pronóstico , Estadística como Asunto
9.
Mol Oncol ; 9(9): 1744-59, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26115764

RESUMEN

Tamoxifen is the treatment of choice in estrogen receptor alpha breast cancer patients that are eligible for adjuvant endocrine therapy. However, ∼50% of ERα-positive tumors exhibit intrinsic or rapidly acquire resistance to endocrine treatment. Unfortunately, prediction of de novo resistance to endocrine therapy and/or assessment of relapse likelihood remain difficult. While several mechanisms regulating the acquisition and the maintenance of endocrine resistance have been reported, there are several aspects of this phenomenon that need to be further elucidated. Altered metabolic fate of tamoxifen within patients and emergence of tamoxifen-resistant clones, driven by evolution of the disease phenotype during treatment, appear as the most compelling hypotheses so far. In addition, tamoxifen was reported to induce pluripotency in breast cancer cell lines, in vitro. In this context, we have performed a whole transcriptome analysis of an ERα-positive (T47D) and a triple-negative breast cancer cell line (MDA-MB-231), exposed to tamoxifen for a short time frame (hours), in order to identify how early pluripotency-related effects of tamoxifen may occur. Our ultimate goal was to identify whether the transcriptional actions of tamoxifen related to induction of pluripotency are mediated through specific ER-dependent or independent mechanisms. We report that even as early as 3 hours after the exposure of breast cancer cells to tamoxifen, a subset of ERα-dependent genes associated with developmental processes and pluripotency are induced and this is accompanied by specific phenotypic changes (expression of pluripotency-related proteins). Furthermore we report an association between the increased expression of pluripotency-related genes in ERα-positive breast cancer tissues samples and disease relapse after tamoxifen therapy. Finally we describe that in a small group of ERα-positive breast cancer patients, with disease relapse after surgery and tamoxifen treatment, ALDH1A1 (a marker of pluripotency in epithelial cancers which is absent in normal breast tissue) is increased in relapsing tumors, with a concurrent modification of its intra-cellular localization. Our data could be of value in the discrimination of patients susceptible to develop tamoxifen resistance and in the selection of optimized patient-tailored therapies.


Asunto(s)
Aldehído Deshidrogenasa/genética , Antineoplásicos Hormonales/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Mama/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Tamoxifeno/farmacología , Aldehído Deshidrogenasa/análisis , Familia de Aldehído Deshidrogenasa 1 , Antineoplásicos Hormonales/uso terapéutico , Mama/metabolismo , Mama/patología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Resistencia a Antineoplásicos , Receptor alfa de Estrógeno/análisis , Femenino , Humanos , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Retinal-Deshidrogenasa , Tamoxifeno/uso terapéutico , Transcriptoma/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología
10.
PLoS One ; 8(12): e83250, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24376672

RESUMEN

Gliomas are common and lethal tumors of the central nervous system (CNS). Genetic alterations, inflammatory and angiogenic processes have been identified throughout tumor progression; however, treatment still remains palliative for most cases. Biological research on parameters influencing cell survival, invasion and tumor heterogeneity identified several cytokines interfering in CNS inflammation, oxidative stress and malignant transformation, including TNF-superfamily (TNFSF) members. In this report we performed a meta-analysis of public gene-array data on the expression of a group of TNFSF ligands (BAFF, APRIL, TWEAK) and their receptors (BAFF-R, TACI, BCMA, Fn14) in gliomas. In addition, we investigated by immunohistochemistry (IHC) the tumor cells' expression of these ligands and receptors in a series of 56 gliomas of different grade. We show that in IHC, BAFF and APRIL as well as their cognate receptors (BCMA, TACI) and Fn14 expression correlate with tumor grade. This result was not evidenced in micro-arrays meta-analysis. Finally, we detected for the first time Fn14, BAFF, BCMA and TACI in glioma-related vascular endothelium. Our data, combined with our previous report in glioma cell lines, suggest a role for these receptors and ligands in glioma biology and advance these molecules as potential markers for the classification of these tumors to the proliferative, angiogenic or stem-like molecular subtype.


Asunto(s)
Factor Activador de Células B/genética , Antígeno de Maduración de Linfocitos B/genética , Neoplasias del Sistema Nervioso Central/genética , Glioma/genética , Receptores del Factor de Necrosis Tumoral/genética , Proteína Activadora Transmembrana y Interactiva del CAML/genética , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Factores de Necrosis Tumoral/genética , Factor Activador de Células B/metabolismo , Antígeno de Maduración de Linfocitos B/metabolismo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias del Sistema Nervioso Central/metabolismo , Neoplasias del Sistema Nervioso Central/patología , Citocina TWEAK , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Regulación Neoplásica de la Expresión Génica , Glioma/metabolismo , Glioma/patología , Humanos , Inmunohistoquímica , Análisis por Micromatrices , Clasificación del Tumor , Receptores del Factor de Necrosis Tumoral/metabolismo , Receptor de TWEAK , Proteína Activadora Transmembrana y Interactiva del CAML/metabolismo , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Factores de Necrosis Tumoral/metabolismo
11.
Mol Oncol ; 7(3): 595-610, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23474223

RESUMEN

ERα17p is a peptide corresponding to the sequence P295LMIKRSKKNSLALSLT311 of the estrogen receptor alpha (ERα) and initially found to interfere with ERα-related calmodulin binding. ERα17p was subsequently found to elicit estrogenic responses in E2-deprived ERα-positive breast cancer cells, increasing proliferation and ERE-dependent gene transcription. Surprisingly, in E2-supplemented media, ERα17p-induced apoptosis and modified the actin network, influencing cell motility. Here, we report that ERα17p internalizes in breast cancer cells (T47D, MDA-MB-231, SKBR3) and induces a massive early (3 h) transcriptional activity. Remarkably, about 75% of significantly modified transcripts were also modified by E2, confirming the pro-estrogenic profile of ERα17p. The different ER spectra of the used cell lines allowed us to identify a specific ERα17p signature related to ERα as well as its variant ERα36. With respect to ERα, the peptide activates nuclear (cell cycle, cell proliferation, nucleic acid and protein synthesis) and extranuclear signaling pathways. In contrast, through ERα36, it mainly triggers inhibitory actions on inflammation. This is the first work reporting a detailed ERα36-specific transcriptional signature. In addition, we report that ERα17p-induced transcripts related to apoptosis and actin modifying effects of the peptide are independent from its estrogen receptor(s)-related actions. We discuss our findings in view of the potential use of ERα17p as a selective peptidomimetic estrogen receptor modulator (PERM).


Asunto(s)
Neoplasias de la Mama/genética , Mama/metabolismo , Receptor alfa de Estrógeno/química , Receptor alfa de Estrógeno/metabolismo , Perfilación de la Expresión Génica , Secuencia de Aminoácidos , Apoptosis , Mama/patología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Ciclo Celular , Línea Celular Tumoral , Movimiento Celular , Estradiol/metabolismo , Femenino , Humanos , Datos de Secuencia Molecular , Péptidos/química , Péptidos/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Activación Transcripcional
12.
Steroids ; 77(10): 943-50, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22138208

RESUMEN

Estrogens exert their effect through ERα and ERß intracellular transcription factors and rapid, usually membrane-initiated receptors, influencing cytosolic signaling and transcription. The nature of extranuclear estrogen elements has not been elucidated so far; classical or alternatively transcribed ER isoforms (ERα36, ERα46) anchored to the plasma membrane and GPR30 (GPER1) have been reported to exert early estrogen actions. Here, we used E2-BSA, an impermeable estradiol analog for a transcriptome analysis in four GREP1 positive breast cancer cell lines with different estrogen receptor profiles (T47D, MCF-7, MDA-MB-231 and SKBR3) in order to evaluate GPER1 transcriptional effects. Early effects of E2-BSA were assayed after 3h of incubation, in the absence/presence of ICI182,780 (ER-inhibitor) or G15 (GREP1-specific inhibitor). E2-BSA specifically modified 277-549 transcripts in the different cell lines. Two different clusters of transcripts could be identified: (1) the majority of transcripts were inhibited by both ICI182,780 and G15, suggesting an interaction of E2-BSA with a common ER-related element, or a direct ER-GPER1 interaction; (2) a small number of G15-only modified transcripts, in two cell lines (T47D and SKBR3 cells), indicative of specific GPER1-related effects. The latter transcripts were significantly related to pathways including FOXA2/FOXA3 transcription factor networks, RNA-Polymerases Transcription Regulation and lipid metabolism, while ICI/G15 inhibited transcripts affected pathways related to apoptosis, erythropoietin signaling, metabolic effects through the citric acid cycle, IL-4 and IL-5 mediated events and homologous DNA recombination. Finally, we review the current literature of GPER1 actions, in view of our results of ER-dependent and independent GPER1-modified pathways.


Asunto(s)
Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transcriptoma , Neoplasias de la Mama , Línea Celular Tumoral , Análisis por Conglomerados , Estradiol/análogos & derivados , Estradiol/farmacología , Estrógenos/farmacología , Femenino , Fulvestrant , Regulación Neoplásica de la Expresión Génica , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Isoformas de Proteínas/metabolismo , Receptores de Estrógenos/agonistas , Receptores de Estrógenos/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Albúmina Sérica Bovina/farmacología
13.
J Nutr Biochem ; 23(6): 656-66, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21782406

RESUMEN

Quercetin is a flavonol modifying a number of cell processes in different cell lines. Here, we present evidence that nonconjugated quercetin enters cells possibly via organic anion transporter polypeptides and quickly accumulates in the nucleus where it concentrates at distinct foci. Furthermore, it induces major transcriptional events with a high number of transcripts being modified over time and about 2200 transcripts being continuously influenced by the agent. The latter transcripts are related to cell cycle and adhesion, xenobiotic metabolism, immune-related factors and transcription. In addition, quercetin up-regulates the expression of estrogen receptors α and ß. The overall outcome on cell fate is reflected by an inhibition of cell proliferation, cell cycle arrest in the G1 phase and reduction of the cells' migratory potential due to actin cytoskeleton disorganization. Finally, we report that the flavonol modifies the transcription and/or activity of numerous transcription factors. In conclusion, our data support the idea that quercetin may actively accumulate in discrete cell structures and exert more than just antioxidant actions on epithelial cells by regulating mechanisms related to gene transcription.


Asunto(s)
Antioxidantes/farmacología , Células Epiteliales/efectos de los fármacos , Quercetina/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Epiteliales/metabolismo , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Fase G1/efectos de los fármacos , Células Hep G2 , Humanos , Factores de Transcripción , Activación Transcripcional , Regulación hacia Arriba
14.
Steroids ; 77(10): 928-34, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22198466

RESUMEN

Triple negative breast cancer is deprived of estrogen receptor alpha (ERα), progesterone receptor (PR) and HER-2 protein. It constitutes the most heterogeneous and aggressive group of breast carcinomas, for which identification of novel characteristics and characterization of putative targets becomes very demanding. In the present work we have assayed the expression of ERα36, a recently identified ERα variant of 36kDa, in a series of triple negative breast cancers, in relation to the clinical behavior and other clinico-pathological features of the tumors. While widely expressed within the cytoplasm in almost all tumors, we found that exclusively the membrane/submembrane expression of the receptor exhibits a correlation with patient's survival. Moreover, membrane ERα36 correlates in an inverse manner with the expression of miRNA210, a pro-angiogenic miR, with high prognostic relevance in triple negative carcinomas. A thorough transcriptomic, pharmacological-based approach in breast cancer cell lines, revealed an early (direct) transcriptional signature of the receptor activation, related to immune system processes and T-cell differentiation, RNA biosynthesis, regulation of metabolism, VEGF signaling and regulation of the cell cycle, with a down-regulation of CREB, NFκB and STATs transcription factors. Finally, ERα36 expression is not limited within breast cancer epithelial linen, but is equally identified in tumor vasculature, peritumoral fat tissue, lymphocytic infiltrate and stromal fibroblasts. In light of the above, ERα36 could represent a major counterpart in triple negative breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Carcinoma Ductal de Mama/metabolismo , Receptor alfa de Estrógeno/metabolismo , Neoplasias Hormono-Dependientes/metabolismo , Transcriptoma , Adulto , Anciano , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/mortalidad , Carcinoma Ductal de Mama/patología , Línea Celular Tumoral , Receptor alfa de Estrógeno/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Redes y Vías Metabólicas , MicroARNs/metabolismo , Persona de Mediana Edad , Neoplasias Hormono-Dependientes/mortalidad , Neoplasias Hormono-Dependientes/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Modelos de Riesgos Proporcionales , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estadísticas no Paramétricas
15.
Steroids ; 77(10): 959-67, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22406407

RESUMEN

The complexity of estrogen actions mainly relies to the presence of different identified receptors (ERα, ERß, their isoforms, and GPR30/GPER) and their discrete cellular distribution. Depending on the localization of the receptor that mediates estrogen effects, nuclear and extra-nuclear actions have been described. The latter can trigger a number of signaling events leading also to transcriptional modifications. In an attempt to clarify the nature of the receptor(s) involved in the membrane initiated effect of estrogens on gene expression, we performed a whole transcriptome analysis of breast cancer cell lines with different receptor profiles (T47D, MCF7, MDA-MB-231, SK-BR-3). A pharmacological approach was conducted with the use of estradiol (E(2)) or membrane-impermeable E(2)-BSA in the absence or presence of a specific ERα-ß or GPR30/GPER antagonist. Our results clearly show that in addition to the ERα isoforms and/or GPR30/GPER that mainly mediate the transcriptional effect of E(2)-BSA, there is a specific transcriptional signature (found in T47D and MCF-7 cells) suggesting the presence of an unidentified membrane ER element (ERx). Analysis of its signature and phenotypic verification revealed that important cell function such as apoptosis, transcriptional regulation, and growth factor signaling are associated with ERx.


Asunto(s)
Estradiol/farmacología , Estrógenos/farmacología , Receptores de Estrógenos/metabolismo , Albúmina Sérica Bovina/farmacología , Transcriptoma , Apoptosis , Neoplasias de la Mama , Línea Celular Tumoral , Movimiento Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Estradiol/análogos & derivados , Femenino , Fulvestrant , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Canales de Potasio de Dominio Poro en Tándem/genética , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Receptores de Estrógenos/antagonistas & inhibidores , Transducción de Señal
16.
Steroids ; 77(10): 979-87, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22426414

RESUMEN

Recently, we identified a peptide (ERα17p, P(295)LMIKRSKKNSLALSLT(311)) that corresponds to the 295-311 sequence of the estrogen receptor α (ERα, hinge region) and which exerts a panel of pharmacological effects in breast cancer cells. Remarkably, these effects can result from the interaction of ERα17p with the plasma membrane. Herein, we show that ERα17p adopts a ß-sheet secondary structure when in contact with anionic phospholipids and that it is engulfed within the lipid bilayer. While ERα17p increases the fluidity of membrane mimics, it weakly internalizes in living cells. In light of the above, one may evoke one important role of the 295-311 region of the ERα: the corresponding peptide could be secreted/delivered to the extracellular medium to interact with neighboring cells, both intracellularly and at the membrane level. Finally, the 295-311 region of ERα being in proximity to the cystein-447, the palmitoylation site of the ERα raises the question of its involvement in the interaction/stabilization of the protein with the membrane.


Asunto(s)
Membrana Celular/metabolismo , Receptor alfa de Estrógeno/metabolismo , Fragmentos de Péptidos/metabolismo , Secuencia de Aminoácidos , Animales , Células CHO , Rastreo Diferencial de Calorimetría , Cricetinae , Dimiristoilfosfatidilcolina/química , Endodesoxirribonucleasas , Proteínas de Escherichia coli , Receptor alfa de Estrógeno/química , Fluoresceínas/química , Cinética , Liposomas/química , Fluidez de la Membrana , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Permeabilidad , Fosfatidilgliceroles/química , Unión Proteica , Estabilidad Proteica , Estructura Secundaria de Proteína , Estándares de Referencia , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/normas , Termodinámica
17.
Endocrinology ; 153(2): 739-49, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22166983

RESUMEN

TNFα is known to be expressed in human skin, regulating immune-related responses. Here we report that human normal skin keratinocytes express the members of the TNF superfamily members A proliferation-inducing ligand (APRIL; TNFSF13), B cell-activating factor (BAFF; TNFSF13B), and their receptors, B cell maturation antigen (BCMA; TNFRSF17) and transmembrane activator, calcium-modulator, and cyclophilin ligand interactor (TACI; TNFRSF13B), in a distinct spatial pattern. Our data show a differential expression of these molecules within epidermal layers and skin appendages, whereas the BAFF-specific receptor BAFFR (TNFRSF13C) is absent. Importantly, APRIL and BCMA but not BAFF or TACI are up-regulated in inflammatory skin lesions of psoriasis and squamous cell carcinomas. To explore the functional significance of this system in the skin, we assayed these receptors and ligands in cultured primary keratinocytes and HaCaT cells. We show that both cell types express BAFF, APRIL, BCMA, and TACI. Furthermore, APRIL and/or BAFF trigger nuclear factor-κB activation and IL-6 and granulocyte macrophage colony-stimulating factor (GM-CSF) expression through functional BCMA receptors, an activation inhibited by anti-BCMA short hairpin RNA. However, BAFF and/or APRIL do not induce IL-8 or TNFα production. Our data advance BCMA as an inflammation-related TNFSFR member in keratinocytes, of potential importance in the management of inflammatory skin conditions.


Asunto(s)
Antígeno de Maduración de Linfocitos B/metabolismo , Dermatitis/metabolismo , Regulación de la Expresión Génica/fisiología , Queratinocitos/metabolismo , Factor Activador de Células B/genética , Factor Activador de Células B/metabolismo , Dermatitis/patología , Epidermis/metabolismo , Epidermis/patología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Proteína Activadora Transmembrana y Interactiva del CAML/genética , Proteína Activadora Transmembrana y Interactiva del CAML/metabolismo , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo
18.
PLoS One ; 7(3): e33609, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22438963

RESUMEN

The soluble TNF-like weak inducer of apoptosis (TWEAK, TNFSF12) binds to the fibroblast growth factor-inducible 14 receptor (FN14, TNFRSF12A) on the cell membrane and induces multiple biological responses, such as proliferation, migration, differentiation, angiogenesis and apoptosis. Previous reports show that TWEAK, which does not contain a death domain in its cytoplasmic tail, induces the apoptosis of tumor cell lines through the induction of TNFα secretion. TWEAK induces apoptosis in human keratinocytes. Our experiments clearly demonstrate that TWEAK does not induce the secretion of TNFα or TRAIL proteins. The use of specific inhibitors and the absence of procaspase-3 cleavage suggest that the apoptosis of keratinocytes follows a caspase- and cathepsin B-independent pathway. Further investigation showed that TWEAK induces a decrease in the mitochondrial membrane potential of keratinocytes. Confocal microscopy showed that TWEAK induces the cleavage and the translocation of apoptosis inducing factor (AIF) from the mitochondria to the nucleus, thus initiating caspase-independent apoptosis. Moreover, TWEAK induces FOXO3 and GADD45 expression, cdc2 phosphorylation and cdc2 and cyclinB1 degradation, resulting in the arrest of cell growth at the G2/M phase. Finally, we report that TWEAK and FN14 are normally expressed in the basal layer of the physiological epidermis and are greatly enhanced in benign (psoriasis) and malignant (squamous cell carcinoma) skin pathologies that are characterized by an inflammatory component. TWEAK might play an essential role in skin homeostasis and pathology.


Asunto(s)
Factor Inductor de la Apoptosis/metabolismo , Apoptosis/fisiología , Puntos de Control de la Fase G2 del Ciclo Celular/fisiología , Queratinocitos/citología , Queratinocitos/metabolismo , Factores de Necrosis Tumoral/metabolismo , Transporte Activo de Núcleo Celular , Proteína Quinasa CDC2 , Caspasas/metabolismo , Catepsina B/metabolismo , Línea Celular , Ciclina B/metabolismo , Ciclina B1/metabolismo , Quinasas Ciclina-Dependientes , Citocina TWEAK , Humanos , Inflamación/metabolismo , Inflamación/patología , Psoriasis/metabolismo , Psoriasis/patología , Receptores del Factor de Necrosis Tumoral/metabolismo , Piel/citología , Piel/metabolismo , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Ligando Inductor de Apoptosis Relacionado con TNF/biosíntesis , Receptor de TWEAK , Factor de Necrosis Tumoral alfa/biosíntesis
19.
Int J Dev Biol ; 55(7-9): 869-78, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22161842

RESUMEN

The present work reviews recent findings related to the action of steroidal (physiological) estrogens on normal mammary gland development and carcinogenesis, as well as effects of related environmental mediators (phyto- and xeno-estrogens), the role of which remains controversial. Orchestration by estrogen receptors (i.e. ERα and ERß) and coregulators of growth, apoptosis and differentiation of epithelial cells, directed our analysis. The bidirectional coordination between epithelium and stroma in parallel with maintenance of stemness are also investigated. The relevance of nuclear and extranuclear localization of ERs and other eventual estrogen binding sites, mediating differential actions in regard to these various topics, is critically addressed to delineate the importance of direct and indirect activation procedures and delicate feedback loops (ligand-induced or/and cross-talk activation, respectively). The inclusion of the outlined regulatory concepts in drug design programs for the prevention and treatment of breast cancer may have potent effects.


Asunto(s)
Neoplasias de la Mama/etiología , Mama/crecimiento & desarrollo , Estrógenos/farmacología , Estrógenos/fisiología , Fitoestrógenos/farmacología , Animales , Mama/efectos de los fármacos , Mama/fisiología , Neoplasias de la Mama/patología , Neoplasias de la Mama/fisiopatología , Carcinógenos Ambientales/toxicidad , Contaminantes Ambientales/toxicidad , Congéneres del Estradiol/toxicidad , Femenino , Humanos , Isoflavonas/farmacología , Glándulas Mamarias Animales/efectos de los fármacos , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/fisiología , Ratones , Modelos Biológicos , Fitoestrógenos/toxicidad , Receptor Cross-Talk , Receptores de Estrógenos/fisiología , Transducción de Señal , Células Madre/citología , Células Madre/efectos de los fármacos , Células Madre/fisiología
20.
Mol Oncol ; 5(1): 36-47, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21163714

RESUMEN

In recent years, our knowledge on estrogen receptors (ER) has been modified profoundly with the identification and the deciphering of the role of its protein effectors, as well as with the deeper insight of its molecular structure/function dynamics, characteristics associated with its nucleo-cytoplasmic-membrane shuttling properties. Also, significant progress has been made concerning its turn-over and associated final proteasomal degradation processes. These advances could lead in the near future to the design and the synthesis of novel receptor-interacting drugs. Recently, a number of receptor-related peptides acting as specific ER ligands have been identified and extensively studied with respect to their estrogenic/antiestrogenic activities. Among them, ERα17p, a synthetic analog of the P(295)-T(311) sequence of ERα, has been shown to exert pseudo-estrogenic effects by interacting in the close vicinity of its hinge region (BF3 domain). Remarkably, this sequence appears as the epicenter of a number of post-transcriptional modifications as well as of the recruitment of co-regulators, suggesting that it would play a key role in ERα functions. Here, we provide evidence that ERα17p induces apoptosis in ERα-positive (MCF-7, T47D) and -negative (MDA-MB-231, SK-BR-3) breast cancer cells by an ERα-independent membrane mechanism, triggering major pro-apoptotic signaling cascades. Finally, ERα17p induces the regression of breast ERα-negative cancer tumor xenografts, without apparent toxicity, suggesting that it could represent a new attractive tool for the development of future promising therapeutic approaches, and providing a novel insight to ER regulation of cell fate.


Asunto(s)
Apoptosis/fisiología , Neoplasias de la Mama/patología , Receptor alfa de Estrógeno/metabolismo , Péptidos/fisiología , Animales , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA