Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Circulation ; 126(11): 1373-84, 2012 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-22865892

RESUMEN

BACKGROUND: Selective inhibitors of cyclooxygenase (COX)-2 increase the risk of myocardial infarction and thrombotic events, but the responsible mechanisms are not fully understood. METHODS AND RESULTS: We found that ferric chloride-induced arterial thrombus formation was significantly greater in COX-2 knockout compared with wild-type mice. Cross-transfusion experiments excluded the likelihood that COX-2 knockout platelets, despite enhanced aggregation responses to collagen and thrombin, are responsible for increased arterial thrombus formation in COX-2 knockout mice. Importantly, we observed that COX-2 deletion decreased prostacyclin synthase and production and peroxisome proliferator-activated receptor- and sirtuin-1 (SIRT1) expression, with consequent increased upregulation of tissue factor (TF), the primary initiator of blood coagulation. Treatment of wild-type mice with a prostacyclin receptor antagonist or a peroxisome proliferator-activated receptor-δ antagonist, which predisposes to arterial thrombosis, decreased SIRT1 expression and increased TF activity. Conversely, exogenous prostacyclin or peroxisome proliferator-activated receptor-δ agonist completely reversed the thrombotic phenotype in COX-2 knockout mice, restoring normal SIRT1 levels and reducing TF activity. Furthermore, inhibition of SIRT1 increased TF expression and activity and promoted generation of occlusive thrombi in wild-type mice, whereas SIRT1 activation was sufficient to decrease abnormal TF activity and prothrombotic status in COX-2 knockout mice. CONCLUSIONS: Modulation of SIRT1 and hence TF by prostacyclin/peroxisome proliferator-activated receptor-δ pathways not only represents a new mechanism in controlling arterial thrombus formation but also might be a useful target for therapeutic intervention in the atherothrombotic complications associated with COX-2 inhibitors.


Asunto(s)
Trombosis de las Arterias Carótidas/epidemiología , Trombosis de las Arterias Carótidas/metabolismo , Ciclooxigenasa 2/metabolismo , Epoprostenol/metabolismo , Sirtuina 1/metabolismo , Tromboplastina/antagonistas & inhibidores , Animales , Plaquetas/fisiología , Trombosis de las Arterias Carótidas/inducido químicamente , Cloruros/efectos adversos , Ciclooxigenasa 2/deficiencia , Ciclooxigenasa 2/genética , Compuestos Férricos/efectos adversos , Incidencia , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , PPAR delta/agonistas , PPAR delta/antagonistas & inhibidores , Receptores de Epoprostenol/agonistas , Receptores de Epoprostenol/antagonistas & inhibidores , Factores de Riesgo , Transducción de Señal , Tromboplastina/metabolismo
2.
Prostaglandins Other Lipid Mediat ; 106: 99-105, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23624019

RESUMEN

The cyclooxygenase/prostaglandin (COX/PG) signaling pathway is of central importance in inflammation and neoplasia. COX inhibitors are widely used for analgesia and also have demonstrated activity for cancer prophylaxis. However, cardiovascular toxicity associated with this drug class diminishes their clinical utility and motivates the development of safer approaches both for pain relief and cancer prevention. The terminal synthase microsomal PGE synthase-1 (mPGES-1) has attracted considerable attention as a potential target. Overexpression of mPGES-1 has been observed in both colorectal and breast cancers, and gene knockout and overexpression approaches have established a role for mPGES-1 in gastrointestinal carcinogenesis. Here we evaluate the contribution of mPGES-1 to mammary tumorigenesis using a gene knockout approach. Mice deficient in mPGES-1 were crossed with a strain in which breast cancer is driven by overexpression of human epidermal growth factor receptor 2 (HER2/neu). Loss of mPGES-1 was associated with a substantial reduction in intramammary PGE2 levels, aromatase activity, and angiogenesis in mammary glands from HER2/neu transgenic mice. Consistent with these findings, we observed a significant reduction in multiplicity of tumors ≥1mm in diameter, suggesting that mPGES-1 contributes to mammary tumor growth. Our data identify mPGES-1 as a potential anti-breast cancer target.


Asunto(s)
Eliminación de Gen , Oxidorreductasas Intramoleculares/deficiencia , Oxidorreductasas Intramoleculares/genética , Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/patología , Neovascularización Patológica/genética , Animales , Aromatasa/metabolismo , Carcinogénesis/genética , Línea Celular Tumoral , Proliferación Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Glándulas Mamarias Animales/metabolismo , Neoplasias Mamarias Experimentales/enzimología , Neoplasias Mamarias Experimentales/genética , Ratones , Ratones Transgénicos , Microvasos/metabolismo , Prostaglandina-E Sintasas , Receptor ErbB-2/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
3.
BMC Cancer ; 12: 500, 2012 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-23110523

RESUMEN

BACKGROUND: We have recently reported that the expression of peptidylarginine deiminase 2 (PADI2) is regulated by EGF in mammary cancer cells and appears to play a role in the proliferation of normal mammary epithelium; however, the role of PADI2 in the pathogenesis of human breast cancer has yet to be investigated. Thus, the goals of this study were to examine whether PADI2 plays a role in mammary tumor progression, and whether the inhibition of PADI activity has anti-tumor effects. METHODS: RNA-seq data from a collection of 57 breast cancer cell lines was queried for PADI2 levels, and correlations with known subtype and HER2/ERBB2 status were evaluated. To examine PADI2 expression levels during breast cancer progression, the cell lines from the MCF10AT model were used. The efficacy of the PADI inhibitor, Cl-amidine, was tested in vitro using MCF10DCIS cells grown in 2D-monolayers and 3D-spheroids, and in vivo using MCF10DCIS tumor xenografts. Treated MCF10DCIS cells were examined by flow-cytometry to determine the extent of apoptosis and by RT2 Profiler PCR Cell Cycle Array to detect alterations in cell cycle associated genes. RESULTS: We show by RNA-seq that PADI2 mRNA expression is highly correlated with HER2/ERBB2 (p = 2.2 × 106) in luminal breast cancer cell lines. Using the MCF10AT model of breast cancer progression, we then demonstrate that PADI2 expression increases during the transition of normal mammary epithelium to fully malignant breast carcinomas, with a strong peak of PADI2 expression and activity being observed in the MCF10DCIS cell line, which models human comedo-DCIS lesions. Next, we show that a PADI inhibitor, Cl-amidine, strongly suppresses the growth of MCF10DCIS monolayers and tumor spheroids in culture. We then carried out preclinical studies in nude (nu/nu) mice and found that Cl-amidine also suppressed the growth of xenografted MCF10DCIS tumors by more than 3-fold. Lastly, we performed cell cycle array analysis of Cl-amidine treated and control MCF10DCIS cells, and found that the PADI inhibitor strongly affects the expression of several cell cycle genes implicated in tumor progression, including p21, GADD45α, and Ki67. CONCLUSION: Together, these results suggest that PADI2 may function as an important new biomarker for HER2/ERBB2+ tumors and that Cl-amidine represents a new candidate for breast cancer therapy.


Asunto(s)
Biomarcadores de Tumor/fisiología , Neoplasias de la Mama/enzimología , Hidrolasas/fisiología , Proteínas de Neoplasias/fisiología , Animales , Antineoplásicos/farmacología , Biomarcadores de Tumor/antagonistas & inhibidores , Línea Celular Tumoral , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Inhibidores Enzimáticos/farmacología , Femenino , Perfilación de la Expresión Génica , Humanos , Hidrolasas/antagonistas & inhibidores , Ratones , Ratones Desnudos , Proteínas de Neoplasias/antagonistas & inhibidores , Arginina Deiminasa Proteína-Tipo 2 , Desiminasas de la Arginina Proteica
4.
Cancer Res ; 66(10): 5504-11, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16707480

RESUMEN

Cytochrome P450 aromatase (aromatase), a product of the CYP19 gene, catalyzes the synthesis of estrogens from androgens. Given the significance of estrogen synthesis in hormone-dependent breast carcinogenesis, it is important to elucidate the mechanisms that regulate CYP19 expression. The main objective of this study was to define the interrelationship between HER-2/neu, cyclooxygenase-2 (COX-2), and aromatase in mammary tissue. Mammary aromatase activity and prostaglandin E(2) (PGE(2)) levels were increased in mice with mammary-targeted expression of a COX-2 transgene. In vitro, overexpressing COX-2 caused both increased PGE(2) production and aromatase activity, effects that were suppressed by celecoxib, a selective COX-2 inhibitor. Previously, we found that overexpression of HER-2/neu was associated with increased levels of COX-2 in human breast cancers. Here, we show that overexpression of HER-2/neu is also associated with increased aromatase activity. These results suggested the possibility that COX-2 was the functional intermediate linking HER-2/neu and aromatase. Consistent with this idea, COX-2 deficiency led to a gene dose-dependent reduction in mammary aromatase activity in a HER-2/neu transgenic mouse model. Complementary in vitro studies showed that HER-2/neu-mediated induction of PGE(2) synthesis and aromatase activity were suppressed by inhibiting COX-2. Collectively, our data indicate that COX-2 is the functional intermediate linking HER-2/neu and aromatase and suggest that inhibitors of PGE(2) synthesis will suppress estrogen biosynthesis in breast tissue.


Asunto(s)
Aromatasa/metabolismo , Ciclooxigenasa 2/metabolismo , Glándulas Mamarias Animales/metabolismo , Receptor ErbB-2/metabolismo , Animales , Inhibidores de la Aromatasa/farmacología , Celecoxib , Ciclooxigenasa 2/biosíntesis , Ciclooxigenasa 2/genética , Inhibidores de la Ciclooxigenasa 2/farmacología , Dinoprostona/biosíntesis , Dinoprostona/metabolismo , Humanos , Glándulas Mamarias Animales/enzimología , Ratones , Ratones Transgénicos , Células 3T3 NIH , Pirazoles/farmacología , Receptor ErbB-2/biosíntesis , Receptor ErbB-2/deficiencia , Receptor ErbB-2/genética , Sulfonamidas/farmacología
5.
Breast Cancer Res ; 9(4): 210, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17640394

RESUMEN

Many human cancers exhibit elevated prostaglandin (PG) levels due to upregulation of cyclooxygenase-2 (COX-2), a key enzyme in eicosanoid biosynthesis. COX-2 over-expression has been observed in about 40% of cases of invasive breast carcinoma and at a higher frequency in preinvasive ductal carcinoma in situ tumors, Extensive pharmacologic and genetic evidence implicates COX enzymes in neoplasia. Epidemiologic analyses demonstrate a protective effect of COX-inhibiting nonsteroidal anti-inflammatory drugs with respect to human cancer. Complementary experimental studies have established that both conventional nonsteroidal anti-inflammatory drugs and selective COX-2 inhibitors suppress mammary tumor formation in rodent breast cancer models. Furthermore, knocking out Cox-2 reduces mammary tumorigenesis and angiogenesis, and, conversely, transgenic COX-2 over-expression induces tumor formation. The utility of COX/PG signaling as a target for chemoprevention has been established by randomized controlled clinical trials. However, these studies also identified increased cardiovascular risk associated with use of selective COX-2 inhibitors. Thus, current efforts are directed toward identifying safer approaches to antagonizing COX/PG signaling for cancer prevention and treatment, with a particular focus on PGE2 regulation and signaling, because PGE2 is a key pro-tumorigenic prostanoid.


Asunto(s)
Neoplasias de la Mama/enzimología , Ciclooxigenasa 2/fisiología , Prostaglandinas/fisiología , Transducción de Señal/fisiología , Neoplasias de la Mama/prevención & control , Inhibidores de la Ciclooxigenasa 2/uso terapéutico , Humanos , Inflamación
6.
Cancer Res ; 65(21): 10113-9, 2005 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-16267038

RESUMEN

The inducible prostaglandin synthase cyclooxygenase-2 (Cox-2) is overexpressed in approximately 40% of human breast cancers and at higher frequencies in preinvasive ductal carcinoma in situ (DCIS). Cox-2 expression is particularly associated with overexpression of human epidermal growth factor receptor 2 (HER2/neu). To definitively interrogate the role of Cox-2 in mammary neoplasia, we have used a genetic approach, crossing Cox-2-deficient mice with a HER2/neu transgenic strain, MMTV/NDL. At 20 weeks of age, mammary glands from virgin MMTV/NDL females contained multiple focal tumors, or mammary intraepithelial neoplasias, which histologically resembled human DCIS. Mammary tumor multiplicity and prostaglandin E2 (PGE2) levels were significantly decreased in Cox-2 heterozygous and knockout animals relative to Cox-2 wild-type controls. Notably, the proportion of larger tumors was decreased in Cox-2-deficient mice. HER2/neu-induced mammary hyperplasia was also substantially reduced in Cox-2 null mice. Additionally, mammary glands from Cox-2 knockout mice exhibited a striking reduction in vascularization, and expression of proangiogenic genes was correspondingly reduced. Decreased vascularization was observed both in dysplastic and normal-appearing regions of Cox-2-null mammary glands. Our data provide the first genetic evidence that Cox-2 contributes to HER2/neu-induced mammary tumorigenesis. This finding may help to explain the reduced risk of breast cancer associated with regular use of nonsteroidal anti-inflammatory drugs.


Asunto(s)
Ciclooxigenasa 2/deficiencia , Neoplasias Mamarias Experimentales/genética , Receptor ErbB-2/genética , Animales , Ciclooxigenasa 2/genética , Femenino , Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/enzimología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Patológica/enzimología , Neovascularización Patológica/genética
8.
Cell Signal ; 29: 41-51, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27511963

RESUMEN

Recent evidence suggests that mammary cells expressing R-spondin receptor and Wnt pathway regulator Lgr5, regarded as a stem cell marker in multiple tissues, might represent mammary stem cells (MaSCs). Whether L gr5 marks a multipotent subpopulation of Lin-CD24low/medCD49fhigh MaSCs remains controversial. To some extent the differing results reflect different assays used to assess properties of stemness, including lineage tracing in vivo, mammosphere culture, and mammary fat pad transplantation assays. To address this issue directly, we isolated Lgr5+ cells from mammary glands of Lgr5-lacZ mice and established organoids based on principles adapted from studies of Wnt-driven Lgr5+ cell populations in other organs. Mammary organoids were grown from single Lgr5+ mammary cells in Matrigel, the substratum of choice for intestinal organoids, and in a growth factor cocktail containing EGF, Wnt3a and R-spondin, designed to optimally activate the endogenous Wnt signaling program of stem cells. Colonies derived from single Lgr5+ cells manifest at least four distinct cell populations: Lgr5+ and Lgr5- basal cells and c-Kit+ and c-Kit- luminal cells that spontaneously organize into a ductal structure with basal cells around the periphery and luminal cells lining an interior cavity, reminiscent of normal mammary duct structure. Lgr5+ cell-derived organoids were sustainable during prolonged passaging. In contrast, although Lgr5- cells expand into primary colonies, colony-forming efficiency immediately dissipated upon passaging. Furthermore, reproductive hormones induce epithelial cell proliferation resulting in marked increases in lumen diameter accompanied by squamous transdifferentiation. We propose this estrogen-responsive, self-organizing duct-like structure derived from single murine Lgr5+ mammary cells represents a "mini-breast" organoid.


Asunto(s)
Estrógenos/farmacología , Glándulas Mamarias Animales/metabolismo , Organoides/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Ensayo de Unidades Formadoras de Colonias , Femenino , Glándulas Mamarias Animales/citología , Ratones Endogámicos C57BL , Organoides/citología , Organoides/efectos de los fármacos
10.
Cancer Res ; 63(8): 1906-13, 2003 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-12702582

RESUMEN

Wnt1, initially identified as a mammary oncogene, can activate transcription via beta-catenin/TCF complexes. Twist, a transcription factor of the basic helix-loop-helix class, has also been suggested to have oncogenic properties. The aim of this study was to determine whether Twist is regulated by Wnt1 and might thus be a novel mediator of Wnt signaling. We found that Twist was up-regulated in C57MG and HC11 murine mammary epithelial cells in response to Wnt1 expression. Additionally, we detected Twist expression in normal mammary gland and found elevated Twist expression in approximately 70% of mammary tumors from Wnt1 transgenic mice. A murine Twist promoter fragment was shown to be responsive to beta-catenin, and its activity was enhanced by coexpression of c-jun and Ets factors of the PEA3 family. Both PEA3 factors and c-jun were highly expressed in tumors from Wnt1 transgenic mice and may therefore contribute to the increased Twist expression observed in these tumors. To evaluate functional consequences of Twist induction, we examined the effect of Twist on mammary cell differentiation. Strikingly, overexpression of either Wnt1 or Twist in HC11 mammary epithelial cells completely suppressed induction of the milk protein beta-casein in response to lactogenic hormones. Additionally, Wnt1, but not Twist, partially abrogated induction of WDNM1, another marker of lactogenic differentiation. Taken together, our data indicate that Twist expression is regulated by Wnt/beta-catenin signaling and that both Wnt1 and Twist can function as inhibitors of lactogenic differentiation, an effect that could contribute to mammary tumorigenesis.


Asunto(s)
Glándulas Mamarias Animales/fisiología , Neoplasias Mamarias Experimentales/genética , Proteínas Nucleares/genética , Proteínas Proto-Oncogénicas/genética , Proteínas de Pez Cebra , Animales , Secuencia de Bases , Diferenciación Celular/fisiología , Proteínas del Citoesqueleto/antagonistas & inhibidores , Proteínas del Citoesqueleto/biosíntesis , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/fisiología , Femenino , Regulación Neoplásica de la Expresión Génica , Masculino , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/metabolismo , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Proteínas Nucleares/biosíntesis , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas c-jun/fisiología , Transactivadores/antagonistas & inhibidores , Transactivadores/biosíntesis , Transactivadores/genética , Transactivadores/fisiología , Factores de Transcripción/fisiología , Proteína 1 Relacionada con Twist , Regulación hacia Arriba , Proteínas Wnt , Proteína Wnt1 , beta Catenina
11.
Cancer Res ; 62(19): 5405-7, 2002 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-12359744

RESUMEN

Cyclooxygenase 2 (HER-2) (Cox-2), an inducible form of Cox, is overexpressed in HER-2/neu-positive human breast cancers. The aim of this study was to determine whether celecoxib, a selective Cox-2 inhibitor, protected against HER-2/neu-induced experimental breast cancer. Cox-2 protein was detected in breast carcinomas from mouse mammary tumor virus (MMTV)/neu mice. Treatment with celecoxib (500 ppm) significantly reduced the incidence of mammary tumors in MMTV/neu mice (P = 0.003) and caused about a 50% reduction in mammary prostaglandin E2 (PGE2) levels. Because mammary glands from MMTV/neu mice expressed all four PGE2 receptor subtypes, we speculate that signaling through PGE2 receptors is important for mammary tumorigenesis. These results strengthen the rationale for developing clinical trials to determine whether selective Cox-2 inhibitors possess anticancer properties in humans at risk for breast cancer.


Asunto(s)
Anticarcinógenos/farmacología , Inhibidores de la Ciclooxigenasa/farmacología , Isoenzimas/antagonistas & inhibidores , Neoplasias Mamarias Experimentales/prevención & control , Receptor ErbB-2/antagonistas & inhibidores , Sulfonamidas/farmacología , Animales , Celecoxib , Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa 2 , Femenino , Humanos , Isoenzimas/biosíntesis , Neoplasias Mamarias Experimentales/enzimología , Neoplasias Mamarias Experimentales/genética , Virus del Tumor Mamario del Ratón/genética , Proteínas de la Membrana , Ratones , Ratones Transgénicos , Prostaglandina-Endoperóxido Sintasas/biosíntesis , Pirazoles , Receptor ErbB-2/biosíntesis , Receptor ErbB-2/genética
12.
Clin Cancer Res ; 22(9): 2283-9, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26712688

RESUMEN

PURPOSE: Obesity, insulin resistance, and elevated levels of circulating proinflammatory mediators are associated with poorer prognosis in early-stage breast cancer. To investigate whether white adipose tissue (WAT) inflammation represents a potential unifying mechanism, we examined the relationship between breast WAT inflammation and the metabolic syndrome and its prognostic importance. EXPERIMENTAL DESIGN: WAT inflammation was defined by the presence of dead/dying adipocytes surrounded by macrophages forming crown-like structures (CLS) of the breast. Two independent groups were examined in cross-sectional (cohort 1) and retrospective (cohort 2) studies. Cohort 1 included 100 women undergoing mastectomy for breast cancer risk reduction (n = 10) or treatment (n = 90). Metabolic syndrome-associated circulating factors were compared by CLS-B status. The association between CLS of the breast and the metabolic syndrome was validated in cohort 2, which included 127 women who developed metastatic breast cancer. Distant recurrence-free survival (dRFS) was compared by CLS-B status. RESULTS: In cohorts 1 and 2, breast WAT inflammation was detected in 52 of 100 (52%) and 52 of 127 (41%) patients, respectively. Patients with breast WAT inflammation had elevated insulin, glucose, leptin, triglycerides, C-reactive protein, and IL6 and lower high-density lipoprotein cholesterol and adiponectin (P < 0.05) in cohort 1. In cohort 2, breast WAT inflammation was associated with hyperlipidemia, hypertension, and diabetes (P < 0.05). Compared with patients without breast WAT inflammation, the adjusted HR for dRFS was 1.83 (95% CI, 1.07-3.13) for patients with inflammation. CONCLUSIONS: WAT inflammation, a clinically occult process, helps to explain the relationship between metabolic syndrome and worse breast cancer prognosis. Clin Cancer Res; 22(9); 2283-9. ©2015 AACR.


Asunto(s)
Tejido Adiposo Blanco/patología , Neoplasias de la Mama/patología , Inflamación/patología , Adipocitos/metabolismo , Adipocitos/patología , Tejido Adiposo Blanco/metabolismo , Adulto , Anciano , Mama/metabolismo , Mama/patología , Neoplasias de la Mama/etiología , Neoplasias de la Mama/metabolismo , Proteína C-Reactiva/metabolismo , Estudios Transversales , Femenino , Glucosa/metabolismo , Humanos , Inflamación/etiología , Inflamación/metabolismo , Insulina/metabolismo , Interleucina-6/metabolismo , Leptina/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Síndrome Metabólico/metabolismo , Síndrome Metabólico/patología , Persona de Mediana Edad , Obesidad/complicaciones , Obesidad/metabolismo , Obesidad/patología , Estudios Retrospectivos , Triglicéridos/metabolismo
13.
Oncogene ; 23(28): 4873-84, 2004 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-15064719

RESUMEN

Secreted signaling proteins of the Wnt family are known to regulate a diverse range of developmental processes, and their signaling pathway through beta-catenin is frequently activated in cancer. The identification of both Frizzled and LRP5/6 (LRP: low-density lipoprotein receptor-related protein) proteins as components of cell-surface receptors for Wnt proteins has raised questions about their individual functions. We have investigated this issue through a structure-function analysis of Frizzled and LRP proteins that have been implicated in Wnt1 signaling. Consistent with other reports, we find that LRP6/Arrow proteins deleted for their extracellular domain are able to activate the Wnt/beta-catenin signaling pathway. Importantly, our results demonstrate that this signaling from LRP6/Arrow derivatives can occur in a Frizzled- and ligand-independent manner. Furthermore, we show that the PPSP motifs within the intracellular domain of LRP6 are required for signaling. In contrast to results with LRP6, overexpression of Frizzled proteins did not activate the pathway. Based on evidence of ligand binding to both Frizzled and LRP6, current models suggest that both proteins are components of a Wnt receptor complex that signals to beta-catenin. In light of these models, our data imply that LRP5/6/Arrow proteins constitute the distal signal-initiating component of these receptors. The results also support the notion that LRP5/6 are candidate oncogenes.


Asunto(s)
Proteínas del Citoesqueleto/fisiología , Proteínas/fisiología , Receptores de LDL/fisiología , Transactivadores/fisiología , Animales , Secuencia de Bases , Línea Celular , Cartilla de ADN , Drosophila melanogaster , Receptores Frizzled , Genes Reporteros , Humanos , Proteínas Relacionadas con Receptor de LDL , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad , Luciferasas/análisis , Oncogenes , Receptores de LDL/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Eliminación de Secuencia , Transducción de Señal/fisiología , Transfección , beta Catenina
14.
Cancer Biol Ther ; 3(1): 36-41, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14739782

RESUMEN

Secreted signaling factors of the Wnt protein family regulate many cellular processes, including cell fate decisions and cell proliferation, and aberrant Wnt signaling is associated with tumorigenesis. Many Wnt proteins act via a signaling pathway that results in stabilization of beta-catenin and consequent transcriptional activation of specific target genes. Mutations in beta-catenin or other Wnt pathway components, which result in beta-catenin accumulation, are found in a wide range of human cancers. In contrast, such mutations have been found only rarely in breast cancer. Nevertheless there is strong evidence of stabilization of beta-catenin protein in a majority of human breast tumors. Moreover, studies in mouse model systems clearly demonstrate that activated Wnt signaling leads to mammary tumorigenesis. This review summarizes the current evidence implicating Wnt/beta-catenin signaling in breast cancer and discusses several possible mechanisms by which the pathway may become activated.


Asunto(s)
Neoplasias de la Mama/genética , Proteínas Proto-Oncogénicas/genética , Animales , Proteínas del Citoesqueleto/genética , Femenino , Humanos , Neoplasias Mamarias Experimentales/fisiopatología , Ratones , Transducción de Señal , Transactivadores/genética , Proteínas Wnt , beta Catenina
15.
Semin Oncol ; 29(3 Suppl 11): 111-9, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12138405

RESUMEN

Cyclooxygenase-2 (COX-2) is being intensively evaluated as a pharmacologic target for both the prevention and treatment of cancer. Aberrant COX-2 expression was initially described in colorectal cancers and has now been detected in many human tumors, including breast cancers. Strikingly, forced expression of COX-2 in murine mammary gland drives tumor formation. Moreover, knocking out COX-2 protects against the formation of intestinal and skin tumors in animal cancer models. Consistent with these findings, selective COX-2 inhibitors possess anticancer properties. For example, selective COX-2 inhibitors reduce the formation and growth of experimental breast and colon cancers. Importantly, selective COX-2 inhibitors do not inhibit platelet function and cause fewer gastrointestinal side effects (peptic ulcer disease) than traditional nonsteroidal anti-inflammatory drugs. Clinical trials are warranted to define the role of selective COX-2 inhibitors in the prevention and treatment of cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Inhibidores de la Ciclooxigenasa/uso terapéutico , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Neoplasias de la Mama/fisiopatología , Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa/farmacología , Femenino , Humanos , Isoenzimas/metabolismo , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Proteínas de la Membrana , Neoplasias/fisiopatología , Prostaglandina-Endoperóxido Sintasas/metabolismo , Roedores
17.
Clin Cancer Res ; 19(22): 6074-83, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23958744

RESUMEN

The increasing rate of obesity worldwide is predicted to be associated with a surge in diseases. Notably, obesity has been linked to approximately 20% of cancer cases in the United States; obesity is associated with both increased risk and worse outcomes after diagnosis. Altered levels of circulating factors are strongly implicated, including insulin, insulin-like growth factor 1, leptin, adiponectin, and interleukin-6 (IL-6). In addition, increasing attention has focused on the consequences of local adipose inflammation. Inflammatory foci characterized by crown-like structures consisting of dead adipocytes encircled by macrophages occur in white adipose depots, including the breast tissue, of most overweight and obese women. Saturated fatty acids, released as a consequence of obesity-associated lipolysis, induce macrophage activation via Toll-like receptor 4, thereby stimulating NF-κB signaling. This, in turn, activates transcription of proinflammatory genes including COX-2, IL-6, IL-1ß, and TNFα. Elevated levels of proinflammatory mediators cause both local and systemic effects. Of particular relevance with regard to breast cancer is increased transcription of the CYP19 gene encoding aromatase, the rate-limiting enzyme for estrogen synthesis. Notably, this obesity-inflammation-aromatase axis provides a plausible explanation for increased rates of postmenopausal, hormone receptor-positive breast cancer associated with obesity and hence may offer targets for interventions to attenuate risk or improve prognosis. Potential approaches include weight reduction, exercise, and suppression of obesity-driven signaling pathways using pharmaceutical or dietary agents. A key future goal is to identify biomarkers that accurately report adipose inflammation, both for identification of at-risk individuals and to assess the efficacy of interventions. Clin Cancer Res; 19(22); 6074-83. ©2013 AACR.


Asunto(s)
Tejido Adiposo/inmunología , Neoplasias de la Mama/inmunología , Inflamación/inmunología , Macrófagos/inmunología , Obesidad/inmunología , Adipocitos/inmunología , Adipocitos/patología , Aromatasa/biosíntesis , Aromatasa/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Ciclooxigenasa 2/biosíntesis , Ciclooxigenasa 2/genética , Femenino , Humanos , Interleucina-1beta/biosíntesis , Interleucina-1beta/genética , Interleucina-6/biosíntesis , Interleucina-6/genética , Activación de Macrófagos/inmunología , FN-kappa B/inmunología , FN-kappa B/metabolismo , Receptor Toll-Like 4/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética
18.
PLoS One ; 7(3): e33421, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22457761

RESUMEN

Wnt/ß-catenin signaling is strongly implicated in neoplasia, but the role of this pathway in human breast cancer has been controversial. Here, we examined Wnt/ß-catenin pathway activation as a function of breast cancer progression, and tested for a relationship with HER2/neu expression, using a human tissue microarray comprising benign breast tissues, ductal carcinoma in situ (DCIS), and invasive carcinomas. Cores were scored for membranous ß-catenin, a key functional component of adherens junctions, and for nucleocytoplasmic ß-catenin, a hallmark of Wnt/ß-catenin pathway activation. Only 82% of benign samples exhibited membrane-associated ß-catenin, indicating a finite frequency of false-negative staining. The frequency of membrane positivity was similar in DCIS samples, but was significantly reduced in carcinomas (45%, P<0.001), consistent with loss of adherens junctions during acquisition of invasiveness. Negative membrane status in cancers correlated with higher grade (P = 0.04) and estrogen receptor-negative status (P = 0.03), both indices of poor prognosis. Unexpectedly, a substantial frequency of nucleocytoplasmic ß-catenin was observed in benign breast tissues (36%), similar to that in carcinomas (35%). Positive-staining basal nuclei observed in benign breast may identify putative stem cells. An increased frequency of nucleocytoplasmic ß-catenin was observed in DCIS tumors (56%), suggesting that pathway activation may be an early event in human breast neoplasia. A correlation was observed between HER2/neu expression and nucleocytoplasmic ß-catenin in node-positive carcinomas (P = 0.02). Furthermore, cytoplasmic ß-catenin was detected in HER2/neu-induced mouse mammary tumors. The Axin2(NLSlacZ) mouse strain, a previously validated reporter of mammary Wnt/ß-catenin signaling, was utilized to define in vivo transcriptional consequences of HER2/neu-induced ß-catenin accumulation. Discrete hyperplastic foci observed in mammary glands from bigenic MMTV/neu, Axin2(NLSlacZ) mice, highlighted by robust ß-catenin/TCF signaling, likely represent the earliest stage of mammary intraepithelial neoplasia in MMTV/neu mice. Our study thus provides provocative evidence for Wnt/ß-catenin signaling as an early, HER2/neu-inducible event in breast neoplasia.


Asunto(s)
Neoplasias de la Mama/metabolismo , Expresión Génica , Genes erbB-2 , Transducción de Señal , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Anciano , Animales , Femenino , Humanos , Inmunohistoquímica , Ratones , Persona de Mediana Edad , Análisis de Matrices Tisulares
19.
Cancer Prev Res (Phila) ; 5(10): 1183-94, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22787115

RESUMEN

Estrogen synthesis is catalyzed by cytochrome P450 aromatase, which is encoded by the CYP19 gene. In obese postmenopausal women, increased aromatase activity in white adipose tissue is believed to contribute to hormone-dependent breast cancer. Prostaglandin E(2) (PGE(2)) stimulates the cAMP→protein kinase A (PKA) pathway leading to increased CYP19 transcription and elevated aromatase activity in inflamed white adipose tissue. 15-hydroxyprostaglandin dehydrogenase (15-PGDH) plays a major role in the catabolism of PGE(2). Here, we investigated the mechanism by which pioglitazone, a ligand of the nuclear receptor PPARγ suppressed aromatase expression. Treatment of human preadipocytes with pioglitazone suppressed Snail, a repressive transcription factor, resulting in elevated levels of 15-PGDH and reduced levels of PGE(2) in the culture medium. Pioglitazone also inhibited cAMP→PKA signaling leading to reduced interaction between phosphorylated cAMP responsive element-binding protein, p300, and CYP19 I.3/II promoter. BRCA1, a repressor of CYP19 transcription, was induced by pioglitazone. Consistent with these in vitro findings, treatment of mice with pioglitazone activated PPARγ, induced 15-PGDH and BRCA1 while suppressing aromatase levels in the mammary gland. Collectively, these results indicate that the activation of PPARγ induces BRCA1 and suppresses the PGE(2)→cAMP→PKA axis leading to reduced levels of aromatase. PPARγ agonists may have a role in reducing the risk of hormone-dependent breast cancer in obese postmenopausal women.


Asunto(s)
Aromatasa/genética , Proteína BRCA1/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Hipoglucemiantes/farmacología , PPAR gamma/agonistas , Tiazolidinedionas/farmacología , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Animales , Aromatasa/química , Aromatasa/metabolismo , Proteína BRCA1/genética , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Células Cultivadas , Inmunoprecipitación de Cromatina , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Dinoprostona/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Femenino , Regulación Enzimológica de la Expresión Génica , Humanos , Hidroxiprostaglandina Deshidrogenasas/genética , Ratones , Pioglitazona , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
20.
Cancer Prev Res (Phila) ; 4(8): 1149-57, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21816844

RESUMEN

Preventing breast cancer is possible with selective estrogen receptor (ER) modulators and aromatase inhibitors, which reduce the risk of invasive disease by up to 65% (up to 73% for ER-positive and no effect for ER-negative cancer) and the risk of preinvasive disease [ductal carcinoma in situ (DCIS)] by up to 50%. Clearly, approaches for preventing ER-negative, and increased prevention of ER-positive breast cancers would benefit public health. A growing body of work (including recent preclinical and clinical data) support targeting the HER family [epidermal growth factor receptor (EGFR), or human epidermal growth factor receptor (HER) 1 or ErbB1) and HER2, HER3, and HER4] for preventing ER-negative and possibly ER-positive breast cancer. Preclinical studies of HER family-targeting drugs in mammary neoplasia show suppression of (i) ER-negative tumors in HER2-overexpressing mouse strains, (ii) ER-negative tumors in mutant Brca1/p53(+/-) mice, and (iii) ER-positive tumors in the methylnitrosourea (MNU) rat model; tumors arising in both the MNU and mutant Brca1/p53(+/-) models lack HER2 overexpression. Clinical trials include a recent placebo-controlled phase IIb presurgical trial of the dual EGFR HER2 inhibitor lapatinib that suppressed growth of breast premalignancy [including atypical ductal hyperplasia (ADH) and DCIS] and invasive cancer in patients with early-stage, HER2-overexpressing or -amplified breast cancer. These results suggest that lapatinib can clinically suppress the progression of ADH and DCIS to invasive breast cancer, an effect previously observed in a mouse model of HER2-overexpressing, ER-negative mammary cancer. The preclinical and clinical signals provide a compelling rationale for testing HER-targeting drugs for breast cancer prevention in women at moderate-to-high risk, leading perhaps to combinations that prevent ER-negative and ER-positive breast cancer.


Asunto(s)
Neoplasias de la Mama/prevención & control , Receptores ErbB/metabolismo , Receptor ErbB-2/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Ensayos Clínicos como Asunto , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Transgénicos , Modelos Biológicos , Placebos , Receptores de Estrógenos/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA