Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Circ Res ; 106(3): 573-82, 2010 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-20019332

RESUMEN

RATIONALE: Mutations in the LMNA gene, which encodes the nuclear lamina proteins lamin A and lamin C, are the most common cause of familial dilated cardiomyopathy (DCM). Mechanical stress-induced apoptosis has been proposed as the mechanism underpinning DCM in lamin A/C-deficient hearts, but supporting in vivo evidence has been lacking. OBJECTIVE: Our aim was to study interventions to modify mechanical stress in heterozygous Lmna knockout (Lmna(+/-)) mice. METHODS AND RESULTS: Cardiac structure and function were evaluated before and after exercise training, thoracic aortic constriction, and carvedilol treatment. Lmna(+/-) mice develop adult-onset DCM with relatively more severe disease in males. Lmna(+/-) cardiomyocytes show altered nuclear morphology and perinuclear desmin organization, with enhanced responses to hypo-osmotic stress indicative of cytoskeletal instability. Despite these structural defects that provide a template for mechanical stress-induced damage, young Lmna(+/-) mice subjected to 6 weeks of moderate or strenuous exercise training did not show induction of apoptosis or accelerated DCM. In contrast, regular moderate exercise attenuated DCM development in male Lmna(+/-) mice. Sustained pressure overload generated by thoracic aortic constriction depressed ventricular contraction in young wild-type and Lmna(+/-) mice with no sex or genotype differences in the time-course or severity of response. Treatment of male Lmna(+/-) mice from 12 to 40 weeks with the beta-blocker, carvedilol, prevented the dilatation and contractile dysfunction that was observed in placebo-treated mice. CONCLUSIONS: These data suggest that factors other than mechanical stress-induced apoptosis contribute to DCM and provide the first demonstration that regular moderate exercise and carvedilol can modify disease progression in lamin A/C-deficient hearts.


Asunto(s)
Antagonistas Adrenérgicos beta/uso terapéutico , Carbazoles/uso terapéutico , Cardiomiopatía Dilatada/genética , Corazón/fisiopatología , Lamina Tipo A/deficiencia , Miocardio/patología , Propanolaminas/uso terapéutico , Estrés Mecánico , Animales , Aorta Torácica , Apoptosis , Cardiomiopatía Dilatada/tratamiento farmacológico , Cardiomiopatía Dilatada/patología , Cardiomiopatía Dilatada/fisiopatología , Carvedilol , Constricción , Desmina/análisis , Femenino , Genotipo , Lamina Tipo A/genética , Masculino , Ratones , Ratones Noqueados , Presión Osmótica , Condicionamiento Físico Animal , Disfunción Ventricular Izquierda/patología , Disfunción Ventricular Izquierda/fisiopatología
2.
Mol Cell Endocrinol ; 301(1-2): 89-96, 2009 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-19100308

RESUMEN

Steroid hormones and their metabolising enzymes have been studied extensively for their potential role in prostate cancer, with more recent interest in the androgen/estrogen inactivating enzyme 17beta-hydroxysteroid dehydrogenase type 4 (HSD17B4). Gene expression profiling showed HSD17B4 to be significantly overexpressed in prostate cancer compared to matched-benign epithelium. We therefore hypothesized that altered HSD17B4 expression may contribute to prostate cancer progression via altered hormone balance. In this study, HSD17B4 mRNA and protein expression were assessed by in situ hybridisation (ISH) and immunohistochemistry (IHC), respectively, in tissue arrays of prostate tissue from 172 patients treated by radical prostatectomy. Overexpression of HSD17B4 mRNA and protein was associated with prostate cancer (P<0.0001) and multivariate Cox proportional hazards analysis, adjusted for known prognostic indicators, demonstrated HSD17B4 mRNA and high protein expression were significant independent predictors of poor patient outcome as measured by time until PSA relapse (mRNA: hazards ratio [HR]=1.90, 95% confidence interval [CI]=1.15-3.12; P<0.0001; and protein: HR=2.09, 95% CI=1.31-3.33; P=0.0026). Here we provide strong evidence that both mRNA and protein overexpression of HSD17B4 is not only associated with the presence of prostate cancer, but is also a significant independent predictor of poor patient outcome.


Asunto(s)
17-Hidroxiesteroide Deshidrogenasas/metabolismo , Biomarcadores de Tumor/metabolismo , Hidroliasas/metabolismo , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/terapia , 17-Hidroxiesteroide Deshidrogenasas/genética , Anciano , Regulación Neoplásica de la Expresión Génica , Humanos , Hidroliasas/genética , Inmunohistoquímica , Hibridación in Situ , Masculino , Persona de Mediana Edad , Proteína-2 Multifuncional Peroxisomal , Modelos de Riesgos Proporcionales , Prostatectomía , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Esteroides/metabolismo , Resultado del Tratamiento
3.
Nucleic Acids Res ; 35(18): e119, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17855397

RESUMEN

CpG methylation is a key component of the epigenome architecture that is associated with changes in gene expression without a change to the DNA sequence. Since the first reports on deregulation of DNA methylation, in diseases such as cancer, and the initiation of the Human Epigenome Project, an increasing need has arisen for a detailed, high-throughput and quantitative method of analysis to discover and validate normal and aberrant DNA methylation profiles in large sample cohorts. Here we present an improved protocol using base-specific fragmentation and MALDI-TOF mass spectrometry that enables a sensitive and high-throughput method of DNA methylation analysis, quantitative to 5% methylation for each informative CpG residue. We have determined the accuracy, variability and sensitivity of the protocol, implemented critical improvements in experimental design and interpretation of the data and developed a new formula to accurately measure CpG methylation. Key innovations now permit determination of differential and allele-specific methylation, such as in cancer and imprinting. The new protocol is ideally suitable for detailed DNA methylation analysis of multiple genomic regions and large sample cohorts that is critical for comprehensive profiling of normal and diseased human epigenomes.


Asunto(s)
Alelos , Islas de CpG , Metilación de ADN , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Genómica/métodos , Humanos , Reacción en Cadena de la Polimerasa , Polimorfismo de Nucleótido Simple , Ribonucleasa Pancreática , Sulfitos/química , Moldes Genéticos , Transcripción Genética
4.
BMC Genomics ; 9: 197, 2008 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-18442417

RESUMEN

BACKGROUND: Highly fecund mouse strains provide an ideal model to understand the factors affecting maternal performance. The QSi5 inbred strain of mice was selected for high fecundity and low inter-litter interval, and is very successful at weaning large numbers of offspring when compared to other inbred strains. RESULTS: Post-natal pup weight gain was used to estimate mammary gland output and to compare the performance of QSi5 mice to CBA mice. Cumulative litter weights and individual pup weight gain was significantly higher throughout the first eight days of lactation in QSi5 mice compared to CBA mice. Morphometric analysis of mammary glands during pregnancy in QSi5 mice revealed a 150 percent greater ductal side branching compared to CBA mice (P < 0.001). Ontology and pathway classification of transcript profiles from the two strains identified an enrichment of genes involved in a number of pathways, including the MAPK, tight junction, insulin signalling and Wnt signalling. Eleven of these genes, including six genes from the MAPK signalling pathway, were identified as associated with postnatal growth. Further, positive mediators of Wnt signalling, including Wnt4, Csnk2a1 and Smad4, were over-represented in the QSi5 strain profile, while negative regulators, including Dkkl1, Ppp2r1a and Nlk, were under-represented. These findings are consistent with the role of Wnt and MAPK signalling pathway in ductal morphogenesis and lobuloalveolar development suggesting enhanced activity in QSi5 mice. A similar pattern of phenotype concordance was seen amongst 12 genes from the tight junction pathway, but a pattern did not emerge from the insulin signalling genes. Amongst a group of differentially expressed imprinted genes, two maternal imprinted genes that suppress growth induced via the IGF signalling pathway, Grb10 and Igf2r, were under-represented in QSi5 mice. Whereas Peg3 and Plagl1, both paternally imprinted genes that enhance neonatal growth, were over-represented in QSi5 mice. CONCLUSION: We propose that the combined action of at least three major signalling pathways involved in mammary gland development and milk secretion, namely Wnt, MAPK and tight junction pathways, contribute to the superior maternal performance phenotype in QSi5 mice. Additionally, favourable expression patterns of the imprinted genes Peg3, Plagl1, Grb10 and Igf2r may also contribute.


Asunto(s)
Perfilación de la Expresión Génica , Glándulas Mamarias Animales/metabolismo , Conducta Materna , Transducción de Señal , Animales , Femenino , Fertilidad , Ratones , Ratones Endogámicos CBA , Ratones Endogámicos , Embarazo , Especificidad de la Especie
5.
Adv Exp Med Biol ; 617: 445-51, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18497068

RESUMEN

Estrogen (E) plays a pivotal regulatory role in the control of cell proliferation in the normal breast and breast cancer (BC). To identify genes with likely roles in proliferation control that are regulated by E and its downstream target c-myc, we compared transcript profiles of antiestrogens-arrested cells stimulated to reinitiate cell cycle progression by E treatment or c-myc induction. Approximately 2/3 of the probe sets significantly regulated by E (adjusted p < 0.01) increased in expression. Half of the E-regulated probe sets were also regulated by c-myc. Genes involved in cell growth, cell proliferation, and cell survival were over-represented in the E-regulated geneset. Analysis of selected candidates has identified a nucleolar protein whose expression is correlated with c-myc expression in BC cell lines. These data indicate that a significant component of E-induced mitogenesis is mediated by c-myc and that selected c-myc target genes may be surrogate markers of c-myc expression in BC.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Estrógenos/farmacología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Zinc/farmacología , Neoplasias de la Mama/genética , Resistencia a Antineoplásicos , Antagonistas de Estrógenos/farmacología , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Proto-Oncogénicas c-myc/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Receptores de Estrógenos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Activación Transcripcional , Células Tumorales Cultivadas
6.
J Clin Pathol ; 60(6): 622-6, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16822880

RESUMEN

BACKGROUND: Currently, no specific immunohistochemical markers are available to differentiate primary mucinous epithelial ovarian cancer (MOC) from adenocarcinomas originating at other sites that have metastasised to the ovary, which may have an impact on patient management and prognosis. AIM: To investigate the expression of two intestinal markers, galectin 4 and meprin alpha, in mucinous carcinomas of the ovary and gastrointestinal tract. METHODS: Using immunohistochemical analysis, the expression of galectin 4 and meprin alpha was investigated in 10 MOCs and in 38 mucinous adenocarcinomas of colon, pancreas, stomach and appendix, the most common sites of origin of ovarian metastases. RESULTS: Total cytoplasmic galectin 4 expression was relatively consistent between the different carcinomas. Membranous meprin alpha expression was significantly lower in MOCs compared with gastrointestinal carcinomas. Moreover, meprin alpha expression showed greater discrimination between the ovarian and gastrointestinal carcinomas than the cytokeratins CK7 and CK20, the current standard immunohistochemical markers used to determine the tissue origin of mucinous carcinomas involving the ovaries. CONCLUSIONS: Meprin alpha is a useful additional marker in differentiating primary from secondary mucinous adenocarcinomas of the ovary.


Asunto(s)
Adenocarcinoma Mucinoso/diagnóstico , Biomarcadores de Tumor/metabolismo , Neoplasias Gastrointestinales/patología , Metaloendopeptidasas/metabolismo , Neoplasias Ováricas/diagnóstico , Adenocarcinoma/diagnóstico , Adenocarcinoma/metabolismo , Adenocarcinoma/secundario , Adenocarcinoma Mucinoso/metabolismo , Diagnóstico Diferencial , Femenino , Galectina 4/metabolismo , Humanos , Técnicas para Inmunoenzimas , Queratina-20/metabolismo , Queratina-7/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/secundario
7.
Cancer Epidemiol Biomarkers Prev ; 15(4): 711-6, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16614113

RESUMEN

Studies to elucidate dysregulated gene expression patterns in premalignant prostate lesions have identified several candidate genes with the potential to be targeted to prevent the development and progression of prostate cancer and act as biomarkers of early disease. Herein, we explored the importance of two proteins, neuropeptide Y (NPY) and macrophage inhibitory cytokine-1 (MIC-1), as biomarkers of preinvasive prostate disease and investigated the relationship of expression to biochemical recurrence following treatment for localized prostate cancer. NPY and MIC-1 protein expression was determined by immunohistochemistry on tissue microarrays containing 1,626 cores of benign, low-grade prostatic intraepithelial neoplasia (PIN), high-grade PIN (HGPIN), and prostate cancer tissue from 243 radical prostatectomy patients. Both NPY and MIC-1 showed higher proportional immunostaining in HGPIN and prostate cancer compared with benign epithelium (P < 0.0001). NPY and MIC-1 immunostaining was higher in low-grade PIN compared with other benign tissues (both P < 0.0001) and was equivalent to immunostaining in HGPIN. NPY immunostaining of prostate cancer was independently associated with relapse, after adjusting for traditional prognostic factors, as a categorical variable in 20% intervals (P = 0.0449-0.0103) and as a continuous variable (P = 0.0017). Low MIC-1 immunostaining (20% categories) was associated with pathologic stage >2C after adjusting for predictors of pathologic stage (P = 0.3894-0.0176). This is the first study to show that altered NPY and MIC-1 expression are significantly associated with prostate cancer progression and suggests that these molecules be developed further as biomarkers in the management of prostate disease.


Asunto(s)
Citocinas/genética , Neuropéptido Y/genética , Neoplasia Intraepitelial Prostática/genética , Neoplasias de la Próstata/metabolismo , Progresión de la Enfermedad , Expresión Génica , Factor 15 de Diferenciación de Crecimiento , Humanos , Inmunohistoquímica , Masculino , Pronóstico , Modelos de Riesgos Proporcionales , Neoplasias de la Próstata/patología
8.
J Steroid Biochem Mol Biol ; 102(1-5): 147-55, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17052904

RESUMEN

The oncoprotein c-Myc is frequently overexpressed in breast cancer and ectopic expression in breast cancer cell lines attenuates responses to antiestrogen treatment. Here, we review preliminary data aimed at further elucidating a potential role for c-Myc in clinical endocrine resistance in breast cancer. Immunohistochemical and semi-quantitative PCR revealed that c-Myc protein and c-myc mRNA were frequently overexpressed in both ER-positive and ER-negative breast carcinoma. Furthermore, both constitutive and inducible c-Myc overexpression in MCF-7 breast cancer cell lines markedly reduced their sensitivity to the growth inhibitory effects of the pure antiestrogen ICI 182,780. In order to identify potential downstream targets of c-Myc that mediate this effect, Affymetrix microarrays were employed to examine the patterns of gene expression shared by MCF-7 cells stimulated by estrogen, or by induction of c-Myc. Approximately 50% of estrogen target genes identified 6h after treatment were also regulated by c-Myc. One novel target, EMU4, was transcriptionally regulated by c-Myc. In addition, there was a strong correlation between c-myc and EMU4 mRNA expression in a battery of breast cancer cell lines. These data confirm that c-Myc overexpression is a common event in breast cancer, and that this is associated with resistance to antiestrogens in vitro. Furthermore, the development of an experimental paradigm for the discovery of c-Myc and estrogen target genes associated with endocrine resistance provides a framework for the discovery and validation of genes involved in estrogen signalling, and c-Myc-mediated-antiestrogen resistance.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Resistencia a Antineoplásicos , Antagonistas de Estrógenos/farmacología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Estrógenos/farmacología , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Proto-Oncogénicas c-myc/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Receptores de Estrógenos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
9.
Mol Endocrinol ; 19(7): 1868-83, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15705664

RESUMEN

The secretory activation stage of mammary gland development occurs after parturition and converts inactive lobuloalveoli to active milk secretion. This process is triggered by progestin withdrawal and depends upon augmented prolactin (Prl) signaling. Little is known about the Prl-induced transcriptional changes that occur in the mammary gland to drive this process. To examine changes in the mammary transcriptome responsible for secretory activation, we have used transcript profiling of three mouse models that exhibit failure of secretory activation: knockout of galanin (a regulator of pituitary Prl production and a mammary cell autonomous modulator of Prl action); treatment with S179D Prl (a phosphoprolactin mimic); and knockout of a single Prl receptor allele. A significant reduction in expression was observed in genes belonging to 46 gene ontologies including those representing milk proteins, metabolism, lipid, cholesterol and fatty acid biosynthetic enzymes, immune response, and key transcription factors. A set of 35 genes, commonly regulated in all three models, was identified and their role in lactogenesis was validated by examining their expression in response to Prl stimulation or signal transducer and activator of transcription 5 knockdown in the HC11 mouse mammary cell culture model. The transcript profiles provided by these experiments identify 35 key genes (many for the first time) involved in the secretory activation phase of mammary gland development, show that S179D acts as an antagonist of Prl action, and provide insight into the partial penetrance of failed lactation in Prl receptor heterozygous females.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Lactancia/genética , Glándulas Mamarias Animales/crecimiento & desarrollo , Proteínas de la Leche/metabolismo , Transactivadores/metabolismo , Transcripción Genética , Alelos , Animales , Células Cultivadas , Proteínas de Unión al ADN/antagonistas & inhibidores , Femenino , Galanina/genética , Perfilación de la Expresión Génica , Lípidos/biosíntesis , Glándulas Mamarias Animales/anatomía & histología , Glándulas Mamarias Animales/metabolismo , Ratones , Ratones Noqueados , Proteínas de la Leche/antagonistas & inhibidores , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosforilación , Prolactina/genética , Prolactina/metabolismo , Prolactina/farmacología , Biosíntesis de Proteínas/genética , Factor de Transcripción STAT5 , Transactivadores/antagonistas & inhibidores
10.
Cancer Res ; 63(14): 4196-203, 2003 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-12874026

RESUMEN

Current models of prostate cancer classification are poor at distinguishing between tumors that have similar histopathological features but vary in clinical course and outcome. Here, we applied classical survival analysis to genome-wide gene expression profiles of prostate cancers and preoperative prostate-specific antigen (PSA) levels from each patient, to identify prognostic markers of disease relapse that provide additional predictive value relative to PSA concentration. Three of approximately 200 probesets showing strongest correlation with relapse were identified as the gene for the putative calcium channel protein, trp-p8, with loss of trp-p8 mRNA expression associated with a significantly shorter time to PSA relapse-free survival. We observed subsequently that trp-p8 is lost in the transition to androgen independence in a prostate cancer xenograft model and in prostate cancer tissue from patients treated preoperatively with antiandrogen therapy, suggesting that trp-p8 is androgen regulated, and its loss may be associated with more advanced disease. The identification of trp-p8 and other proteins implicated in the phosphatidylinositol signal transduction pathway that are associated with prostate cancer outcome, both here and in other published work, suggests an integral role for this pathway in prostate carcinogenesis. Thus, our findings demonstrate that multivariable survival analysis can be applied to gene expression profiles of prostate cancers with censored follow-up data and used to identify molecular markers of prostate cancer relapse with strong predictive power and relevance to the etiology of this disease.


Asunto(s)
Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Animales , Análisis por Conglomerados , Perfilación de la Expresión Génica , Humanos , Masculino , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Pronóstico , Neoplasias de la Próstata/metabolismo , Análisis de Supervivencia , Trasplante Heterólogo
11.
Clin Cancer Res ; 10(13): 4427-36, 2004 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-15240533

RESUMEN

PURPOSE: A better understanding of the molecular pathways underlying the development of epithelial ovarian cancer (EOC) is critical to identify ovarian tumor markers for use in diagnostic or therapeutic applications. The aims of this study were to integrate the results from 14 transcript profiling studies of EOC to identify novel biomarkers and to examine their expression in early and late stages of the disease. EXPERIMENTAL DESIGN: A database incorporating genes identified as being highly up-regulated in each study was constructed. Candidate tumor markers were selected from genes that overlapped between studies and by evidence of surface membrane or secreted expression. The expression patterns of three integral membrane proteins, discoidin domain receptor 1 (DDR1), claudin 3 (CLDN3), and epithelial cell adhesion molecule, all of which are involved in cell adhesion, were evaluated in a cohort of 158 primary EOC using immunohistochemistry. RESULTS: We confirmed that these genes are highly overexpressed in all histological subtypes of EOC compared with normal ovarian surface epithelium, identifying DDR1 and CLDN3 as new biomarkers of EOC. Furthermore, we determined that these genes are also expressed in ovarian epithelial inclusion cysts, a site of metaplastic changes within the normal ovary, in borderline tumors and in low-grade and stage cancer. A trend toward an association between low CLDN3 expression and poor patient outcome was also observed. CONCLUSIONS: These results suggest that up-regulation of DDR1, CLDN3, and epithelial cell adhesion molecule are early events in the development of EOC and have potential application in the early detection of disease.


Asunto(s)
Antígenos de Neoplasias/biosíntesis , Moléculas de Adhesión Celular/biosíntesis , Proteínas de la Membrana/biosíntesis , Neoplasias Ováricas/metabolismo , Ovario/metabolismo , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Biomarcadores de Tumor , Adhesión Celular , Línea Celular Tumoral , Membrana Celular/metabolismo , Claudina-3 , Estudios de Cohortes , ADN Complementario/metabolismo , Bases de Datos como Asunto , Receptor con Dominio Discoidina 1 , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Molécula de Adhesión Celular Epitelial , Epitelio/metabolismo , Femenino , Marcadores Genéticos , Humanos , Inmunohistoquímica , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/metabolismo , Factores de Tiempo , Resultado del Tratamiento , Regulación hacia Arriba
12.
PLoS One ; 4(3): e4710, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19270750

RESUMEN

BACKGROUND: Breast cancers lacking the estrogen receptor (ER) can be distinguished from other breast cancers on the basis of poor prognosis, high grade, distinctive histopathology and unique molecular signatures. These features further distinguish estrogen receptor negative (ER-) tumor subtypes, but targeted therapy is currently limited to tumors over-expressing the ErbB2 receptor. METHODOLOGY/PRINCIPAL FINDINGS: To uncover the pathways against which future therapies could be developed we undertook a meta-analysis of gene expression from five large microarray datasets relative to ER status. A measure of association with ER status was calculated for every Affymetrix HG-U133A probe set and the pathways that distinguished ER- tumors were defined by testing for enrichment of biologically defined gene sets using Gene Set Enrichment Analysis (GSEA). As expected, the expression of the direct transcriptional targets of the ER was muted in ER- tumors, but the expression of genes indirectly regulated by estrogen was enhanced. We also observed enrichment of independent MYC- and E2F-driven transcriptional programs. We used a cell model of estrogen and MYC action to define the interaction between estrogen and MYC transcriptional activity in breast cancer. We found that the basal subgroup of ER- breast cancer showed a strong MYC transcriptional response that reproduced the indirect estrogen response seen in estrogen receptor positive (ER+) breast cancer cells. CONCLUSIONS/SIGNIFICANCE: Increased transcriptional activity of MYC is a characteristic of basal breast cancers where it mimics a large part of an estrogen response in the absence of the ER, suggesting a mechanism by which these cancers achieve estrogen-independence and providing a potential therapeutic target for this poor prognosis sub group of breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Carcinoma Basocelular/genética , Factores de Transcripción E2F/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/fisiología , Proteínas Proto-Oncogénicas c-myc/genética , Receptores de Estrógenos/genética , Algoritmos , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Carcinoma Basocelular/metabolismo , Diagnóstico por Computador , Femenino , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Elementos Reguladores de la Transcripción
13.
Genes Dev ; 22(5): 581-6, 2008 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-18316476

RESUMEN

Hormonal cues regulate mammary development, but the consequent transcriptional changes and cell fate decisions are largely undefined. We show that knockout of the prolactin-regulated Ets transcription factor Elf5 prevented formation of the secretory epithelium during pregnancy. Conversely, overexpression of Elf5 in an inducible transgenic model caused alveolar differentiation and milk secretion in virgin mice, disrupting ductal morphogenesis. CD61+ luminal progenitor cells accumulated in Elf5-deficient mammary glands and were diminished in glands with Elf5 overexpression. Thus Elf5 specifies the differentiation of CD61+ progenitors to establish the secretory alveolar lineage during pregnancy, providing a link between prolactin, transcriptional events, and alveolar development.


Asunto(s)
Diferenciación Celular/genética , Proteínas de Unión al ADN/metabolismo , Lactancia/genética , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/metabolismo , Morfogénesis/genética , Células Madre/metabolismo , Factores de Transcripción/metabolismo , Animales , Linaje de la Célula/genética , Proteínas de Unión al ADN/genética , Epitelio/crecimiento & desarrollo , Epitelio/metabolismo , Femenino , Integrina beta3/análisis , Glándulas Mamarias Animales/citología , Ratones , Ratones Transgénicos , Embarazo , Células Madre/citología , Factores de Transcripción/genética
14.
PLoS One ; 3(8): e2987, 2008 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-18714337

RESUMEN

BACKGROUND: Estrogen is a pivotal regulator of cell proliferation in the normal breast and breast cancer. Endocrine therapies targeting the estrogen receptor are effective in breast cancer, but their success is limited by intrinsic and acquired resistance. METHODOLOGY/PRINCIPAL FINDINGS: With the goal of gaining mechanistic insights into estrogen action and endocrine resistance, we classified estrogen-regulated genes by function, and determined the relationship between functionally-related genesets and the response to tamoxifen in breast cancer patients. Estrogen-responsive genes were identified by transcript profiling of MCF-7 breast cancer cells. Pathway analysis based on functional annotation of these estrogen-regulated genes identified gene signatures with known or predicted roles in cell cycle control, cell growth (i.e. ribosome biogenesis and protein synthesis), cell death/survival signaling and transcriptional regulation. Since inducible expression of c-Myc in antiestrogen-arrested cells can recapitulate many of the effects of estrogen on molecular endpoints related to cell cycle progression, the estrogen-regulated genes that were also targets of c-Myc were identified using cells inducibly expressing c-Myc. Selected genes classified as estrogen and c-Myc targets displayed similar levels of regulation by estrogen and c-Myc and were not estrogen-regulated in the presence of siMyc. Genes regulated by c-Myc accounted for 50% of all acutely estrogen-regulated genes but comprised 85% (110/129 genes) in the cell growth signature. siRNA-mediated inhibition of c-Myc induction impaired estrogen regulation of ribosome biogenesis and protein synthesis, consistent with the prediction that estrogen regulates cell growth principally via c-Myc. The 'cell cycle', 'cell growth' and 'cell death' gene signatures each identified patients with an attenuated response in a cohort of 246 tamoxifen-treated patients. In multivariate analysis the cell death signature was predictive independent of the cell cycle and cell growth signatures. CONCLUSIONS/SIGNIFICANCE: These functionally-based gene signatures can stratify patients treated with tamoxifen into groups with differing outcome, and potentially identify distinct mechanisms of tamoxifen resistance.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Estrógenos/fisiología , Perfilación de la Expresión Génica , Genes myc , Proteínas Proto-Oncogénicas c-myc/fisiología , Tamoxifeno/uso terapéutico , Antineoplásicos Hormonales/uso terapéutico , Neoplasias de la Mama/patología , Ciclo Celular , Muerte Celular , Línea Celular Tumoral , ADN de Neoplasias/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Análisis de Regresión , Transcripción Genética/efectos de los fármacos , Resultado del Tratamiento
15.
Am J Physiol Endocrinol Metab ; 293(1): E364-71, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17456639

RESUMEN

Muscle is a target of growth hormone (GH) action and a major contributor to whole body metabolism. Little is known about how GH regulates metabolic processes in muscle or the extent to which muscle contributes to changes in whole body substrate metabolism during GH treatment. To identify GH-responsive genes that regulate substrate metabolism in muscle, we studied six hypopituitary men who underwent whole body metabolic measurement and skeletal muscle biopsies before and after 2 wk of GH treatment (0.5 mg/day). Transcript profiles of four subjects were analyzed using Affymetrix GeneChips. Serum insulin-like growth factor I (IGF-I) and procollagens I and III were measured by RIA. GH increased serum IGF-I and procollagens I and III, enhanced whole body lipid oxidation, reduced carbohydrate oxidation, and stimulated protein synthesis. It induced gene expression of IGF-I and collagens in muscle. GH reduced expression of several enzymes regulating lipid oxidation and energy production. It reduced calpain 3, increased ribosomal protein L38 expression, and displayed mixed effects on genes encoding myofibrillar proteins. It increased expression of circadian gene CLOCK, and reduced that of PERIOD. In summary, GH exerted concordant effects on muscle expression and blood levels of IGF-I and collagens. It induced changes in genes regulating protein metabolism in parallel with a whole body anabolic effect. The discordance between muscle gene expression profiles and metabolic responses suggests that muscle is unlikely to contribute to GH-induced stimulation of whole body energy and lipid metabolism. GH may regulate circadian function in skeletal muscle by modulating circadian gene expression with possible metabolic consequences.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Hormona de Crecimiento Humana/farmacología , Hipopituitarismo/genética , Hipopituitarismo/metabolismo , Redes y Vías Metabólicas/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Análisis de Secuencia por Matrices de Oligonucleótidos , Adulto , Anciano , Colágeno/genética , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Perfilación de la Expresión Génica , Terapia de Reemplazo de Hormonas , Hormona de Crecimiento Humana/uso terapéutico , Humanos , Hipopituitarismo/tratamiento farmacológico , Factor I del Crecimiento Similar a la Insulina/genética , Masculino , Redes y Vías Metabólicas/genética , Persona de Mediana Edad , Músculo Esquelético/metabolismo
16.
J Infect Dis ; 188(8): 1205-8, 2003 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-14551891

RESUMEN

We sought to determine whether variants of the human alpha-1-antitrypsin (AAT) gene, also known as "PI," or "SERPINA1," are associated with human immunodeficiency virus (HIV) infection in 2 African-based populations from HIV-pandemic sub-Saharan Africa. Eleven commonly occurring African-associated polymorphic markers in the coding and intronic regions of the AAT gene were analyzed via denaturing gradient gel electrophoresis. A significant association between HIV-1 infection and the presence of an allelic variant was observed in the case of the M2 and A332A haplotypes, thus presenting AAT as a potentially novel HIV-1 susceptibility locus.


Asunto(s)
Marcadores Genéticos , Predisposición Genética a la Enfermedad , Infecciones por VIH/genética , Polimorfismo Genético , alfa 1-Antitripsina/genética , Adolescente , Adulto , África del Sur del Sahara , Pueblo Asiatico , Población Negra , Electroforesis en Gel de Poliacrilamida , Femenino , Frecuencia de los Genes , Variación Genética , Seronegatividad para VIH/genética , VIH-1 , Haplotipos , Humanos , Masculino , Persona de Mediana Edad , Población Blanca
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda