Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
2.
Ann Oncol ; 30(1): 115-123, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30423024

RESUMEN

Background: Invasive lobular carcinoma (ILC) as a disease entity distinct from invasive ductal carcinoma (IDC) has merited focused studies of the genomic landscape, but those to date are largely limited to the assessment of early-stage cancers. Given that genomic alterations develop as acquired resistance to endocrine therapy, studies on refractory ILC are needed. Patients and methods: Tissue from 336 primary-enriched, breast-biopsied ILC and 485 estrogen receptor (ER)-positive IDC and metastatic biopsy specimens from 180 ILC and 191 ER-positive IDC patients was assayed with hybrid-capture-based comprehensive genomic profiling for short variant, indel, copy number variants, and rearrangements in up to 395 cancer-related genes. Results: Whereas ESR1 alterations are enriched in the metastases of both ILC and IDC compared with breast specimens, NF1 alterations are enriched only in ILC metastases (mILC). NF1 alterations are predominantly under loss of heterozygosity (11/14, 79%), are mutually exclusive with ESR1 mutations [odds ratio = 0.24, P < 0.027] and are frequently polyclonal in ctDNA assays. Assessment of paired specimens shows that NF1 alterations arise in the setting of acquired resistance. An in vitro model of CDH1 mutated ER-positive breast cancer demonstrates that NF1 knockdown confers a growth advantage in the presence of 4-hydroxy tamoxifen. Our study further identified a significant increase in tumor mutational burden (TMB) in mILCs relative to breast ILCs or metastatic IDCs (8.9% >20 mutations/mb; P < 0.001). Most TMB-high mILCs harbor an APOBEC trinucleotide signature (14/16; 88%). Conclusions: This study identifies alteration of NF1 as enriched specifically in mILC. Mutual exclusivity with ESR1 alterations, polyclonality in relapsed ctDNA, and de novo acquisition suggest a role for NF1 loss in endocrine therapy resistance. Since NF1 loss leads to RAS/RAF kinase activation, patients may benefit from a matched inhibitor. Moreover, for an independent subset of mILC, TMB was elevated relative to breast ILC, suggesting possible benefit from immune checkpoint inhibitors.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/secundario , Carcinoma Lobular/secundario , Resistencia a Antineoplásicos/genética , Neurofibromina 1/genética , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos Hormonales/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Carcinoma Ductal de Mama/tratamiento farmacológico , Carcinoma Ductal de Mama/genética , Carcinoma Lobular/tratamiento farmacológico , Carcinoma Lobular/genética , Femenino , Estudios de Seguimiento , Humanos , Persona de Mediana Edad , Metástasis de la Neoplasia , Pronóstico , Receptor ErbB-2/metabolismo , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo
3.
Ann Oncol ; 29(4): 872-880, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29360925

RESUMEN

Background: Estrogen receptor-positive (ER-positive) metastatic breast cancer is often intractable due to endocrine therapy resistance. Although ESR1 promoter switching events have been associated with endocrine-therapy resistance, recurrent ESR1 fusion proteins have yet to be identified in advanced breast cancer. Patients and methods: To identify genomic structural rearrangements (REs) including gene fusions in acquired resistance, we undertook a multimodal sequencing effort in three breast cancer patient cohorts: (i) mate-pair and/or RNAseq in 6 patient-matched primary-metastatic tumors and 51 metastases, (ii) high coverage (>500×) comprehensive genomic profiling of 287-395 cancer-related genes across 9542 solid tumors (5216 from metastatic disease), and (iii) ultra-high coverage (>5000×) genomic profiling of 62 cancer-related genes in 254 ctDNA samples. In addition to traditional gene fusion detection methods (i.e. discordant reads, split reads), ESR1 REs were detected from targeted sequencing data by applying a novel algorithm (copyshift) that identifies major copy number shifts at rearrangement hotspots. Results: We identify 88 ESR1 REs across 83 unique patients with direct confirmation of 9 ESR1 fusion proteins (including 2 via immunoblot). ESR1 REs are highly enriched in ER-positive, metastatic disease and co-occur with known ESR1 missense alterations, suggestive of polyclonal resistance. Importantly, all fusions result from a breakpoint in or near ESR1 intron 6 and therefore lack an intact ligand binding domain (LBD). In vitro characterization of three fusions reveals ligand-independence and hyperactivity dependent upon the 3' partner gene. Our lower-bound estimate of ESR1 fusions is at least 1% of metastatic solid breast cancers, the prevalence in ctDNA is at least 10× enriched. We postulate this enrichment may represent secondary resistance to more aggressive endocrine therapies applied to patients with ESR1 LBD missense alterations. Conclusions: Collectively, these data indicate that N-terminal ESR1 fusions involving exons 6-7 are a recurrent driver of endocrine therapy resistance and are impervious to ER-targeted therapies.


Asunto(s)
Antineoplásicos Hormonales/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Resistencia a Antineoplásicos/genética , Receptor alfa de Estrógeno/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Neoplasias de la Mama/patología , Receptor alfa de Estrógeno/genética , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Mutación , Metástasis de la Neoplasia , Proteínas Recombinantes de Fusión/genética
4.
Breast Cancer Res Treat ; 166(1): 277-287, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28730340

RESUMEN

PURPOSE: A number of studies have tested the hypothesis that breast cancer patients with low-activity CYP2D6 genotypes achieve inferior benefit from tamoxifen treatment, putatively due to lack of metabolic activation to endoxifen. Studies have provided conflicting data, and meta-analyses suggest a small but significant increase in cancer recurrence, necessitating additional studies to allow for accurate effect assessment. We conducted a retrospective pharmacogenomic analysis of a prospectively collected community-based cohort of patients with estrogen receptor-positive breast cancer to test for associations between low-activity CYP2D6 genotype and disease outcome in 500 patients treated with adjuvant tamoxifen monotherapy and 500 who did not receive any systemic adjuvant therapy. METHODS: Tumor-derived DNA was genotyped for common, functionally consequential CYP2D6 polymorphisms (*2, *3, *4, *6, *10, *41, and copy number variants) and assigned a CYP2D6 activity score (AS) ranging from none (0) to full (2). Patients with poor metabolizer (AS = 0) phenotype were compared to patients with AS > 0 and in secondary analyses AS was analyzed quantitatively. Clinical outcome of interest was recurrence free survival (RFS) and analyses using long-rank test were adjusted for relevant clinical covariates (nodal status, tumor size, etc.). RESULTS: CYP2D6 AS was not associated with RFS in tamoxifen treated patients in univariate analyses (p > 0.2). In adjusted analyses, increasing AS was associated with inferior RFS (Hazard ratio 1.43, 95% confidence interval 1.00-2.04, p = 0.05). In patients that did not receive tamoxifen treatment, increasing CYP2D6 AS, and AS > 0, were associated with superior RFS (each p = 0.0015). CONCLUSIONS: This population-based study does not support the hypothesis that patients with diminished CYP2D6 activity achieve inferior tamoxifen benefit. These contradictory findings suggest that the association between CYP2D6 genotype and tamoxifen treatment efficacy is null or near null, and unlikely to be useful in clinical practice.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/mortalidad , Citocromo P-450 CYP2D6/genética , Genotipo , Polimorfismo Genético , Adulto , Anciano , Alelos , Antineoplásicos Hormonales/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/epidemiología , Femenino , Humanos , Estimación de Kaplan-Meier , Persona de Mediana Edad , Metástasis de la Neoplasia , Estadificación de Neoplasias , Variantes Farmacogenómicas , Pronóstico , Análisis de Supervivencia , Tamoxifeno/uso terapéutico , Resultado del Tratamiento
5.
Mol Cell Endocrinol ; 448: 21-27, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28286232

RESUMEN

Steroid receptor coactivator-1 (SRC-1), a well-studied coactivator of estrogen receptor (ER), is known to play an important and functional role in the development and maintenance of bone tissue. Previous reports suggest SRC-1 maintains bone mineral density primarily through its interaction with ER. Here we demonstrate that SRC-1 can also affect bone development independent of estrogen signaling as ovariectomized SRC-1 knockout (SRC-1 KO) mouse had decreased bone mineral density. To identify estrogen-independent SRC-1 target genes in osteoblastogenesis, we undertook an integrated analysis utilizing ChIP-Seq and mRNA microarray in transformed osteoblast-like U2OS-ERα cells. We identified critical osteoblast differentiation genes regulated by SRC-1, but not by estrogen including alkaline phosphatase and osteocalcin. Ex vivo primary culture of osteoblasts from SRC-1 wild-type and KO mice confirmed the role of SRC-1 in osteoblastogenesis, associated with altered ALPL levels. Together, these data indicate that SRC-1 can impact osteoblast function in an ER-independent manner.


Asunto(s)
Estrógenos/farmacología , Coactivador 1 de Receptor Nuclear/metabolismo , Osteoblastos/metabolismo , Osteogénesis , Fosfatasa Alcalina/metabolismo , Animales , Densidad Ósea/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Humanos , Ratones Noqueados , Osteoblastos/citología , Osteoblastos/efectos de los fármacos
6.
Oncogene ; 36(1): 133-145, 2017 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-27212032

RESUMEN

We have previously demonstrated that crosstalk between lysine-specific demethylase 1 (LSD1) and histone deacetylases (HDACs) facilitates breast cancer proliferation. However, the underlying mechanisms are largely unknown. Here, we report that expression of HDAC5 and LSD1 proteins were positively correlated in human breast cancer cell lines and tissue specimens of primary breast tumors. Protein expression of HDAC5 and LSD1 was significantly increased in primary breast cancer specimens in comparison with matched-normal adjacent tissues. Using HDAC5 deletion mutants and co-immunoprecipitation studies, we showed that HDAC5 physically interacted with the LSD1 complex through its domain containing nuclear localization sequence and phosphorylation sites. Although the in vitro acetylation assays revealed that HDAC5 decreased LSD1 protein acetylation, small interfering RNA (siRNA)-mediated HDAC5 knockdown did not alter the acetylation level of LSD1 in MDA-MB-231 cells. Overexpression of HDAC5 stabilized LSD1 protein and decreased the nuclear level of H3K4me1/me2 in MDA-MB-231 cells, whereas loss of HDAC5 by siRNA diminished LSD1 protein stability and demethylation activity. We further demonstrated that HDAC5 promoted the protein stability of USP28, a bona fide deubiquitinase of LSD1. Overexpression of USP28 largely reversed HDAC5-KD-induced LSD1 protein degradation, suggesting a role of HDAC5 as a positive regulator of LSD1 through upregulation of USP28 protein. Depletion of HDAC5 by shRNA hindered cellular proliferation, induced G1 cell cycle arrest, and attenuated migration and colony formation of breast cancer cells. A rescue study showed that increased growth of MDA-MB-231 cells by HDAC5 overexpression was reversed by concurrent LSD1 depletion, indicating that tumor-promoting activity of HDAC5 is an LSD1 dependent function. Moreover, overexpression of HDAC5 accelerated cellular proliferation and promoted acridine mutagen ICR191-induced transformation of MCF10A cells. Taken together, these results suggest that HDAC5 is critical in regulating LSD1 protein stability through post-translational modification, and the HDAC5-LSD1 axis has an important role in promoting breast cancer development and progression.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Histona Desacetilasas/metabolismo , Histona Demetilasas/metabolismo , Neoplasias de la Mama/genética , Línea Celular Tumoral , Proliferación Celular , Progresión de la Enfermedad , Activación Enzimática , Femenino , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Histona Desacetilasas/genética , Histona Demetilasas/genética , Humanos , Modelos Biológicos , Metástasis de la Neoplasia , Unión Proteica , Estabilidad Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Ubiquitinación
7.
Br J Cancer ; 112(9): 1461-70, 2015 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-25880007

RESUMEN

BACKGROUND: The mechanisms by which stress hormones impact triple-negative breast cancer (TNBC) etiology and treatment are unclear. We have previously shown that stress hormones, cortisol, and catecholamines induce rapid DNA damage and impact DNA repair in NIH 3T3 fibroblasts. This study investigates whether stress hormones increase DNA damage in breast cancer cells and if this impacts drug efficacy. METHODS: We first screened a panel of 39 breast cancer cell lines for expression of adrenergic and glucocorticoid receptors and examined if stress hormones induce DNA damage and alter cell cycle regulation in vitro. A TNBC xenograft model was used to assess the impact of restraint stress on tumour growth and chemosensitivity to paclitaxel. RESULTS: We found that stress hormones induced DNA damage, phosphorylation of ATR, which was accompanied by an up-regulation of the G1 cell kinase inhibitor p21 and a cell cycle halt of TNBCs in the G1 phase. p21 knockdown abrogated G1 arrest by stress hormones. We also demonstrated that stress significantly decreased efficacy of paclitaxel. CONCLUSION: We describe a novel mechanism through which stress hormones can induce drug resistance to paclitaxel, which may have profound implications for treating drug resistance in patients with TNBC.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Catecolaminas/farmacología , Daño del ADN/efectos de los fármacos , Hidrocortisona/farmacología , Paclitaxel/farmacología , Estrés Fisiológico/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/patología , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Femenino , Citometría de Flujo , Humanos , Ratones , Ratones Desnudos , Receptores de Estrógenos/metabolismo , Transducción de Señal , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Genet Mol Res ; 14(1): 585-96, 2015 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-25729994

RESUMEN

Acrocomia aculeata (Jacq.) Lodd. ex Mart. is a plant species commonly used as a foodstuff and also for treating diseases, since it contains high concentrations of antioxidant compounds and monounsaturated fatty acids. Considering its ethnopharmacological relevance, the aim of the present study was to assess the cytotoxic, genotoxic, and mutagenic effects of an oil extracted from the pulp of A. aculeata (OPAC) in rats. In addition, a chromatographic characterization of the fatty acids present in OPAC was performed. Male and female Wistar rats were treated orally with 125, 250, 500, 1000, or 2000 mg/kg/body weight OPAC. The effects of OPAC ingestion were determined by performing the comet assay and micronucleus test. The comet assay data demonstrated that OPAC did not increase the frequency or rate of DNA damage in groups treated with any of the concentrations assessed compared to that in the negative control group. In the micronucleus test, the animals treated did not exhibit any cytotoxic or mutagenic changes in peripheral blood erythrocytes. The results demonstrated that OPAC did not exhibit cytotoxic, genotoxic, or mutagenic effects in Wistar rats, thereby increasing the evidence for the safety of oil extracted from this plant.


Asunto(s)
Arecaceae/química , Mutágenos/toxicidad , Extractos Vegetales/toxicidad , Animales , Muerte Celular/efectos de los fármacos , Daño del ADN , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Femenino , Masculino , Micronúcleos con Defecto Cromosómico , Mutagénesis/efectos de los fármacos , Ratas Wistar
9.
Oncogene ; 33(25): 3235-45, 2014 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-23893242

RESUMEN

The androgen receptor (AR) is a transcription factor that employs many diverse interactions with coregulatory proteins in normal physiology and in prostate cancer (PCa). The AR mediates cellular responses in association with chromatin complexes and kinase cascades. Here we report that the nuclear matrix protein, scaffold attachment factor B1 (SAFB1), regulates AR activity and AR levels in a manner that suggests its involvement in PCa. SAFB1 mRNA expression was lower in PCa in comparison with normal prostate tissue in a majority of publicly available RNA expression data sets. SAFB1 protein levels were also reduced with disease progression in a cohort of human PCa that included metastatic tumors. SAFB1 bound to AR and was phosphorylated by the MST1 (Hippo homolog) serine-threonine kinase, previously shown to be an AR repressor, and MST1 localization to AR-dependent promoters was inhibited by SAFB1 depletion. Knockdown of SAFB1 in androgen-dependent LNCaP PCa cells increased AR and prostate-specific antigen (PSA) levels, stimulated growth of cultured cells and subcutaneous xenografts and promoted a more aggressive phenotype, consistent with a repressive AR regulatory function. SAFB1 formed a complex with the histone methyltransferase EZH2 at AR-interacting chromatin sites in association with other polycomb repressive complex 2 (PRC2) proteins. We conclude that SAFB1 acts as a novel AR co-regulator at gene loci where signals from the MST1/Hippo and EZH2 pathways converge.


Asunto(s)
Factor de Crecimiento de Hepatocito/metabolismo , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Proteínas Asociadas a Matriz Nuclear/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores Androgénicos/metabolismo , Receptores de Estrógenos/metabolismo , Animales , Línea Celular Tumoral , Proteína Potenciadora del Homólogo Zeste 2 , Regulación Neoplásica de la Expresión Génica , Factor de Crecimiento de Hepatocito/genética , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Masculino , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Ratones , Ratones Desnudos , Proteínas Asociadas a Matriz Nuclear/genética , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Regiones Promotoras Genéticas , Antígeno Prostático Específico/genética , Antígeno Prostático Específico/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/enzimología , Neoplasias de la Próstata Resistentes a la Castración/genética , Proteínas Proto-Oncogénicas/genética , Receptores Androgénicos/genética , Receptores de Estrógenos/genética , Transcripción Genética
10.
Br J Cancer ; 109(9): 2331-9, 2013 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-24084768

RESUMEN

BACKGROUND: Change in breast density may predict outcome of women receiving adjuvant hormone therapy for breast cancer. We performed a prospective clinical trial to evaluate the impact of inherited variants in genes involved in oestrogen metabolism and signalling on change in mammographic percent density (MPD) with aromatase inhibitor (AI) therapy. METHODS: Postmenopausal women with breast cancer who were initiating adjuvant AI therapy were enrolled onto a multicentre, randomised clinical trial of exemestane vs letrozole, designed to identify associations between AI-induced change in MPD and single-nucleotide polymorphisms in candidate genes. Subjects underwent unilateral craniocaudal mammography before and following 24 months of treatment. RESULTS: Of the 503 enrolled subjects, 259 had both paired mammograms at baseline and following 24 months of treatment and evaluable DNA. We observed a statistically significant decrease in mean MPD from 17.1 to 15.1% (P<0.001), more pronounced in women with baseline MPD ≥20%. No AI-specific difference in change in MPD was identified. No significant associations between change in MPD and inherited genetic variants were observed. CONCLUSION: Subjects with higher baseline MPD had a greater average decrease in MPD with AI therapy. There does not appear to be a substantial effect of inherited variants in biologically selected candidate genes.


Asunto(s)
Inhibidores de la Aromatasa/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Mama/efectos de los fármacos , Adulto , Anciano , Anciano de 80 o más Años , Androstadienos/uso terapéutico , Aromatasa/genética , Mama/metabolismo , Mama/patología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Quimioterapia Adyuvante/métodos , Estrógenos/metabolismo , Femenino , Humanos , Letrozol , Mamografía/métodos , Persona de Mediana Edad , Nitrilos/uso terapéutico , Polimorfismo de Nucleótido Simple , Posmenopausia/efectos de los fármacos , Posmenopausia/genética , Posmenopausia/metabolismo , Estudios Prospectivos , Triazoles/uso terapéutico
11.
Mol Endocrinol ; 26(2): 220-7, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22174377

RESUMEN

The coregulator steroid receptor coactivator (SRC)-1 increases transcriptional activity of the estrogen receptor (ER) in a number of tissues including bone. Mice deficient in SRC-1 are osteopenic and display skeletal resistance to estrogen treatment. SRC-1 is also known to modulate effects of selective ER modulators like tamoxifen. We hypothesized that single nucleotide polymorphisms (SNP) in SRC-1 may impact estrogen and/or tamoxifen action. Because the only nonsynonymous SNP in SRC-1 (rs1804645; P1272S) is located in an activation domain, it was examined for effects on estrogen and tamoxifen action. SRC-1 P1272S showed a decreased ability to coactivate ER compared with wild-type SRC-1 in multiple cell lines. Paradoxically, SRC-1 P1272S had an increased protein half-life. The Pro to Ser change disrupts a putative glycogen synthase 3 (GSK3)ß phosphorylation site that was confirmed by in vitro kinase assays. Finally, knockdown of GSK3ß increased SRC-1 protein levels, mimicking the loss of phosphorylation at P1272S. These findings are similar to the GSK3ß-mediated phospho-ubiquitin clock previously described for the related coregulator SRC-3. To assess the potential clinical significance of this SNP, we examined whether there was an association between SRC-1 P1272S and selective ER modulators response in bone. SRC-1 P1272S was associated with a decrease in hip and lumbar bone mineral density in women receiving tamoxifen treatment, supporting our in vitro findings for decreased ER coactivation. In summary, we have identified a functional genetic variant of SRC-1 with decreased activity, resulting, at least in part, from the loss of a GSK3ß phosphorylation site, which was also associated with decreased bone mineral density in tamoxifen-treated women.


Asunto(s)
Antineoplásicos Hormonales/efectos adversos , Glucógeno Sintasa Quinasa 3/metabolismo , Coactivador 1 de Receptor Nuclear/genética , Tamoxifeno/efectos adversos , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Antineoplásicos Hormonales/uso terapéutico , Desmineralización Ósea Patológica/inducido químicamente , Desmineralización Ósea Patológica/genética , Densidad Ósea/efectos de los fármacos , Neoplasias de la Mama/prevención & control , Línea Celular Tumoral , Ensayos Clínicos como Asunto , Femenino , Estudios de Asociación Genética , Glucógeno Sintasa Quinasa 3 beta , Humanos , Datos de Secuencia Molecular , Fosforilación , Polimorfismo de Nucleótido Simple , Procesamiento Proteico-Postraduccional , Estabilidad Proteica , Receptores de Estrógenos/agonistas , Receptores de Estrógenos/metabolismo , Análisis de Secuencia de ADN , Tamoxifeno/uso terapéutico
12.
J Pediatr Endocrinol Metab ; 22(8): 751-8, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19845126

RESUMEN

BACKGROUND: Premature thelarche is defined as breast development before 8 years of age. This is most often caused by central hormone disregulation and is accompanied by concurrent bone maturation. However, we present a case of premature thelarche with concurrent bone maturation without central hormone disregulation. Genes within the estrogen signaling pathway were examined for genetic changes which might be responsible for the clinical phenotype. PATIENT REPORT: A girl presented with breast development from 18 months of age with undetectable serum estrogens, prepubertal serum gonadotropins, advanced growth and skeletal maturation, but no increase of uterine size, thus presenting a premature thelarche variant. Serum estrogens remained below detectable limits until she entered into an unremarkable puberty at 12.1 years of age. No abnormalities or SNPs were found in the genes tested. CONCLUSION: We describe a case of premature thelarche which cannot be attributed to a central cause of abnormal hormone levels or to alterations in genes suspected for this phenotype. We conclude that other yet to be identified factors are involved in this unique case of premature thelarche.


Asunto(s)
Mama/crecimiento & desarrollo , Pubertad Precoz/diagnóstico , Adolescente , Niño , Preescolar , Análisis Mutacional de ADN , Estrógenos/sangre , Femenino , Genotipo , Humanos , Lactante , Estudios Longitudinales , Polimorfismo de Nucleótido Simple , Pubertad Precoz/sangre , Pubertad Precoz/etiología , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Transducción de Señal
13.
Pharmacogenomics J ; 9(4): 258-64, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19421167

RESUMEN

The selective estrogen receptor modulator tamoxifen is routinely used for treatment and prevention of estrogen-receptor-positive breast cancer. Studies of tamoxifen adherence suggest that over half of patients discontinue treatment before the recommended 5 years. We hypothesized that polymorphisms in CYP2D6, the enzyme responsible for tamoxifen activation, predict for tamoxifen discontinuation. Tamoxifen-treated women (n=297) were genotyped for CYP2D6 variants and assigned a 'score' based on predicted allele activities from 0 (no activity) to 2 (high activity). Correlation between CYP2D6 score and discontinuation rates at 4 months was tested. We observed a strong nonlinear correlation between higher CYP2D6 score and increased rates of discontinuation (r(2)=0.935, P=0.018). These data suggest that presence of active CYP2D6 alleles may predict for higher likelihood of tamoxifen discontinuation. Therefore, patients who may be most likely to benefit from tamoxifen may paradoxically be most likely to discontinue treatment prematurely.


Asunto(s)
Antineoplásicos Hormonales/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Citocromo P-450 CYP2D6/genética , Cooperación del Paciente , Moduladores Selectivos de los Receptores de Estrógeno/uso terapéutico , Tamoxifeno/uso terapéutico , Citocromo P-450 CYP2D6/metabolismo , Femenino , Humanos , Estudios Prospectivos , Tamoxifeno/efectos adversos , Tamoxifeno/metabolismo
14.
Endocrinology ; 144(6): 2683-94, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12746333

RESUMEN

Insulin receptor substrates (IRS) are central integrators of hormone, cytokine, and growth factor signaling. IRS proteins can be phosphorylated by a number of signaling pathways critical to normal mammary gland development. Studies in transgenic mice that overexpress IGF-I in the mammary gland suggested that IRS expression is important in the regulation of normal postlactational mammary involution. The goal of these studies was to examine IRS expression in the mouse mammary gland and determine the importance of IRS-1 to mammary development in the virgin mouse. IRS-1 and -2 show distinct patterns of protein expression in the virgin mouse mammary gland, and protein abundance is dramatically increased during pregnancy and lactation, but rapidly lost during involution. Consistent with hormone regulation, IRS-1 protein levels are reduced by ovariectomy, induced by combined treatment with estrogen and progesterone, and vary considerably throughout the estrous cycle. These changes occur without similar changes in mRNA levels, suggesting posttranscriptional control. Mammary glands from IRS-1 null mice have smaller fat pads than wild-type controls, but this reduction is proportional to the overall reduction in body size. Development of the mammary duct (terminal endbuds and branch points) is not altered by the loss of IRS-1, and pregnancy-induced proliferation is not changed. These data indicate that IRS undergo complex developmental and hormonal regulation in the mammary gland, and that IRS-1 is more likely to regulate mammary function in lactating mice than in virgin or pregnant mice.


Asunto(s)
Estrógenos/farmacología , Glándulas Mamarias Animales/fisiología , Fosfoproteínas/genética , Progesterona/farmacología , Transducción de Señal/fisiología , Tejido Adiposo/química , Tejido Adiposo/crecimiento & desarrollo , Tejido Adiposo/fisiología , Animales , Ciclo Estral/fisiología , Femenino , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Proteínas Sustrato del Receptor de Insulina , Péptidos y Proteínas de Señalización Intracelular , Glándulas Mamarias Animales/química , Glándulas Mamarias Animales/crecimiento & desarrollo , Ratones , Ratones Endogámicos , Ovariectomía , Fosfoproteínas/análisis , Embarazo , Transducción de Señal/efectos de los fármacos
15.
Endocr Relat Cancer ; 10(4): 517-36, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14713265

RESUMEN

Estrogen receptor alpha (ERalpha) has an established role in promoting breast cancer. Transcriptional activation by ERalpha is a complex and multistep process, and it is influenced by coactivator and corepressor proteins that can either positively or negatively modulate ERalpha-mediated transcriptional activity. Corepressors are proposed to provide a counterbalance to the estrogen-induced transactivation, and represent a potential mechanism employed by the cell to regulate hormonal responses. In this review, we present evidence from tissue culture, animal and clinical studies, supporting the hypothesis that corepressors are crucial regulators of ERalpha-mediated action, and that their loss could promote breast cancer development and resistance to endocrine therapy. We propose that ERalpha corepressors play an important biological role by controlling the magnitude of the estrogen response, mediating antiestrogen inhibition of ERalpha, repressing DNA-bound ERalpha in the absence of the ligand, and conferring active repression of ERalpha-downregulated genes. Different ERalpha corepressors regulate steroid receptor activity through a variety of mechanisms, including formation of multiprotein complexes that are able to affect chromatin remodeling, histone deacetylation, or basal transcription. Other mechanisms include competition with coactivators, interference with DNA binding and ERalpha homodimerization, alteration of ERalpha stability, sequestration of ERalpha in the cytoplasm, and effects on RNA processing. Most ERalpha corepressors can control the receptor's activity through more than one mechanism, and it is possible that the synergy between different pathways cooperates to fully inhibit ERalpha transcriptional activity, and create an integrated response to a variety of different cellular signaling pathways. We will discuss the role of corepressors in tumor suppression and the link they might present between ERalpha regulation and DNA repair. Finally, we will discuss major challenges in the field and speculate on the exciting findings that await us in the next few years.


Asunto(s)
Neoplasias de la Mama/metabolismo , Receptores de Estrógenos/fisiología , Proteínas Represoras/fisiología , Receptor alfa de Estrógeno , Femenino , Humanos , Neoplasias Hormono-Dependientes/genética , Neoplasias Hormono-Dependientes/metabolismo , Receptores de Estrógenos/antagonistas & inhibidores , Transcripción Genética/fisiología
16.
Cancer Res ; 61(15): 5771-7, 2001 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-11479214

RESUMEN

Estrogen can increase insulin-like growth factor-I receptor (IGF-IR) and insulin receptor substrate-1 (IRS-1) expression, two key components of IGF-I-mediated signaling. The result is sensitization of breast cancer cells to IGF-I and synergistic growth in the presence of estrogen and IGF-I. We hypothesized that loss of estrogen receptor alpha (ERalpha) would result in reduced IGF-mediated signaling and growth. To test this hypothesis, we examined IGF-I effects in MCF-7 breast cancer cell sublines that have been selected for loss of ERalpha (C4 and C4-12 cells are ERalpha-negative) by long-term estrogen withdrawal. C4 and C4-12 cells had reduced IGF-IR and IRS-1 mRNA and protein expression (compared with MCF-7 cells) that was not inducible by estrogen. Furthermore, C4 and C4-12 cells showed reduced IGF-I signaling and failed to show any growth response to either estrogen or IGF-I. To prove that loss of IGF and estrogen-mediated signaling and growth was a consequence of loss of ERalpha, we re-expressed ERalpha in C4-12 cells by stable transfection with HA-tagged ERalpha. Three independent C4-12 ERalpha-HA clones expressed a functional ERalpha that (a) was down-regulated by estrogen, (b) conferred estrogen-induction of cyclin D1 expression, and (c) caused estrogen-mediated increase in the number of cells in S phase. All of the effects were completely blocked by antiestrogens. Interestingly, ERalpha-HA expression in C4-12 cells did not restore estrogen induction of progesterone receptor expression. However, ERalpha-positive C4-12 cells now exhibited estrogen-induction of IGF-IR and IRS-1 levels and responded mitogenically to both estrogen and IGF-I. These data show that ERalpha is a critical requirement for IGF signaling, and to our knowledge this is the first report of functional ERalpha expression that confers estrogen-mediated growth of an ER-negative breast cancer cell line.


Asunto(s)
Neoplasias de la Mama/metabolismo , Estradiol/farmacología , Factor I del Crecimiento Similar a la Insulina/farmacología , Fosfoproteínas/fisiología , Receptor IGF Tipo 1/fisiología , Receptores de Estrógenos/fisiología , Transducción de Señal/fisiología , Neoplasias de la Mama/patología , División Celular/fisiología , Ciclina D1/biosíntesis , Receptor alfa de Estrógeno , Hemaglutininas/genética , Humanos , Proteínas Sustrato del Receptor de Insulina , Fosfoproteínas/biosíntesis , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Receptor IGF Tipo 1/biosíntesis , Receptores de Estrógenos/biosíntesis , Receptores de Estrógenos/genética , Transfección , Células Tumorales Cultivadas
17.
Br J Cancer ; 84(4): 493-8, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11207044

RESUMEN

We have recently discovered that the nuclear matrix protein SAFB is an oestrogen receptor corepressor. Since it has become clear that many steroid receptor cofactors play important roles in breast tumorigenesis, we investigated whether SAFB could also be involved in breast cancer. To address this question, the gene locus was examined for structural alterations in breast cancer tissue. Laser capture microdissection was used for isolating DNA from paired primary breast tumour and normal tissue specimens, and the loss of heterozygosity (LOH) at chromosome 19p13.2-3 was determined by use of microsatellite markers. LOH was detected at the marker D19S216, which colocalizes with the SAFB locus, in specimens from 29 (78.4%) of 37 informative patients. The peak LOH rate occurred at D19S216 near the SAFB locus, with LOH frequencies ranging from 21.6% to 47.2% at other markers. The finding of a very high LOH rate at the marker D19S216 strongly indicates the presence of a breast tumour-suppressor gene locus. While preliminary findings of mutations in SAFB suggest that this indeed may be a promising candidate, other potential candidate genes are located at this locus.


Asunto(s)
Neoplasias de la Mama/genética , Cromosomas Humanos Par 19/genética , Genes Supresores de Tumor/genética , Pérdida de Heterocigocidad , Antígenos Nucleares , Análisis Mutacional de ADN , ADN de Neoplasias/análisis , Femenino , Humanos , Repeticiones de Microsatélite/genética , Proteínas Nucleares/genética , Receptores de Estrógenos/genética
18.
Clin Cancer Res ; 7(12 Suppl): 4429s-4435s; discussion 4411s-4412s, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11916236

RESUMEN

Since the cloning of the estrogen receptor alpha (ERalpha) and subsequent identification of a second distinct form of ER, termed ERbeta, a large volume of research has begun to define the molecular mechanisms of ER action. However, although great progress has been made, ER action is still poorly understood. It is expected that a better understanding of ER action may lead to novel strategies and targets for breast cancer prevention and treatment. One of the early-realized functions of the ER was regulation of growth factor signaling, but the degree of interaction between these two mitogenic signaling pathways could not have been anticipated. Recent evidence suggests that the ER and the growth-factor-signaling pathways intersect and directly interact at every level of signal transduction. The resulting synergism between ER and growth factors has been documented both in normal breast development and, importantly, in breast cancer progression and antiestrogen resistance. In this review, we will highlight our current understanding of the molecular mechanisms of cross-talk between ER and growth-factor-signaling pathways.


Asunto(s)
Neoplasias de la Mama/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Receptor Cross-Talk/fisiología , Receptores de Estrógenos/metabolismo , Resistencia a Antineoplásicos , Moduladores de los Receptores de Estrógeno/uso terapéutico , Femenino , Humanos , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...