Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 149(5): 994-1007, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22608083

RESUMEN

Cancer evolves dynamically as clonal expansions supersede one another driven by shifting selective pressures, mutational processes, and disrupted cancer genes. These processes mark the genome, such that a cancer's life history is encrypted in the somatic mutations present. We developed algorithms to decipher this narrative and applied them to 21 breast cancers. Mutational processes evolve across a cancer's lifespan, with many emerging late but contributing extensive genetic variation. Subclonal diversification is prominent, and most mutations are found in just a fraction of tumor cells. Every tumor has a dominant subclonal lineage, representing more than 50% of tumor cells. Minimal expansion of these subclones occurs until many hundreds to thousands of mutations have accumulated, implying the existence of long-lived, quiescent cell lineages capable of substantial proliferation upon acquisition of enabling genomic changes. Expansion of the dominant subclone to an appreciable mass may therefore represent the final rate-limiting step in a breast cancer's development, triggering diagnosis.


Asunto(s)
Neoplasias de la Mama/genética , Transformación Celular Neoplásica , Evolución Clonal , Mutación , Algoritmos , Aberraciones Cromosómicas , Femenino , Humanos , Mutación Puntual
2.
Cell ; 149(5): 979-93, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22608084

RESUMEN

All cancers carry somatic mutations. The patterns of mutation in cancer genomes reflect the DNA damage and repair processes to which cancer cells and their precursors have been exposed. To explore these mechanisms further, we generated catalogs of somatic mutation from 21 breast cancers and applied mathematical methods to extract mutational signatures of the underlying processes. Multiple distinct single- and double-nucleotide substitution signatures were discernible. Cancers with BRCA1 or BRCA2 mutations exhibited a characteristic combination of substitution mutation signatures and a distinctive profile of deletions. Complex relationships between somatic mutation prevalence and transcription were detected. A remarkable phenomenon of localized hypermutation, termed "kataegis," was observed. Regions of kataegis differed between cancers but usually colocalized with somatic rearrangements. Base substitutions in these regions were almost exclusively of cytosine at TpC dinucleotides. The mechanisms underlying most of these mutational signatures are unknown. However, a role for the APOBEC family of cytidine deaminases is proposed.


Asunto(s)
Neoplasias de la Mama/genética , Análisis Mutacional de ADN , Estudio de Asociación del Genoma Completo , Mutación , Desaminasas APOBEC-1 , Proteína BRCA2/genética , Citidina Desaminasa/metabolismo , Femenino , Genes BRCA1 , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos
3.
Nature ; 500(7463): 415-21, 2013 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-23945592

RESUMEN

All cancers are caused by somatic mutations; however, understanding of the biological processes generating these mutations is limited. The catalogue of somatic mutations from a cancer genome bears the signatures of the mutational processes that have been operative. Here we analysed 4,938,362 mutations from 7,042 cancers and extracted more than 20 distinct mutational signatures. Some are present in many cancer types, notably a signature attributed to the APOBEC family of cytidine deaminases, whereas others are confined to a single cancer class. Certain signatures are associated with age of the patient at cancer diagnosis, known mutagenic exposures or defects in DNA maintenance, but many are of cryptic origin. In addition to these genome-wide mutational signatures, hypermutation localized to small genomic regions, 'kataegis', is found in many cancer types. The results reveal the diversity of mutational processes underlying the development of cancer, with potential implications for understanding of cancer aetiology, prevention and therapy.


Asunto(s)
Transformación Celular Neoplásica/genética , Mutagénesis/genética , Mutación/genética , Neoplasias/genética , Envejecimiento/genética , Algoritmos , Transformación Celular Neoplásica/patología , Citidina Desaminasa/genética , ADN/genética , ADN/metabolismo , Análisis Mutacional de ADN , Humanos , Modelos Genéticos , Mutagénesis Insercional/genética , Mutágenos/farmacología , Neoplasias/enzimología , Neoplasias/patología , Especificidad de Órganos , Reproducibilidad de los Resultados , Eliminación de Secuencia/genética , Transcripción Genética/genética
4.
Blood ; 123(25): 3914-24, 2014 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-24802772

RESUMEN

The histone methyltransferase EZH2 is frequently mutated in germinal center-derived diffuse large B-cell lymphoma and follicular lymphoma. To further characterize these EZH2 mutations in lymphomagenesis, we generated a mouse line where EZH2(Y641F) is expressed from a lymphocyte-specific promoter. Spleen cells isolated from the transgenic mice displayed a global increase in trimethylated H3K27, but the mice did not show an increased tendency to develop lymphoma. As EZH2 mutations often coincide with other mutations in lymphoma, we combined the expression of EZH2(Y641F) by crossing these transgenic mice with Eµ-Myc transgenic mice. We observed a dramatic acceleration of lymphoma development in this combination model of Myc and EZH2(Y641F). The lymphomas show histologic features of high-grade disease with a shift toward a more mature B-cell phenotype, increased cycling and gene expression, and epigenetic changes involving important pathways in B-cell regulation and function. Furthermore, they initiate disease in secondary recipients. In summary, EZH2(Y641F) can collaborate with Myc to accelerate lymphomagenesis demonstrating a cooperative role of EZH2 mutations in oncogenesis. This murine lymphoma model provides a new tool to study global changes in the epigenome caused by this frequent mutation and a promising model system for testing novel treatments.


Asunto(s)
Transformación Celular Neoplásica/genética , Linfoma/genética , Mutación , Complejo Represivo Polycomb 2/genética , Proteínas Proto-Oncogénicas c-myc/genética , Animales , Linfocitos B/metabolismo , Linfocitos B/patología , Western Blotting , Células de la Médula Ósea/metabolismo , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Perfilación de la Expresión Génica , Histonas/metabolismo , Humanos , Estimación de Kaplan-Meier , Linfoma/metabolismo , Linfoma/patología , Linfoma de Células B/genética , Linfoma de Células B/metabolismo , Linfoma de Células B/patología , Lisina/metabolismo , Masculino , Metilación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Complejo Represivo Polycomb 2/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Bazo/metabolismo , Bazo/patología
5.
N Engl J Med ; 366(3): 234-42, 2012 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-22187960

RESUMEN

BACKGROUND: Germline truncating mutations in DICER1, an endoribonuclease in the RNase III family that is essential for processing microRNAs, have been observed in families with the pleuropulmonary blastoma-family tumor and dysplasia syndrome. Mutation carriers are at risk for nonepithelial ovarian tumors, notably sex cord-stromal tumors. METHODS: We sequenced the whole transcriptomes or exomes of 14 nonepithelial ovarian tumors and noted closely clustered mutations in the region of DICER1 encoding the RNase IIIb domain of DICER1 in four samples. We then sequenced this region of DICER1 in additional ovarian tumors and in certain other tumors and queried the effect of the mutations on the enzymatic activity of DICER1 using in vitro RNA cleavage assays. RESULTS: DICER1 mutations in the RNase IIIb domain were found in 30 of 102 nonepithelial ovarian tumors (29%), predominantly in Sertoli-Leydig cell tumors (26 of 43, or 60%), including 4 tumors with additional germline DICER1 mutations. These mutations were restricted to codons encoding metal-binding sites within the RNase IIIb catalytic centers, which are critical for microRNA interaction and cleavage, and were somatic in all 16 samples in which germline DNA was available for testing. We also detected mutations in 1 of 14 nonseminomatous testicular germ-cell tumors, in 2 of 5 embryonal rhabdomyosarcomas, and in 1 of 266 epithelial ovarian and endometrial carcinomas. The mutant DICER1 proteins had reduced RNase IIIb activity but retained RNase IIIa activity. CONCLUSIONS: Somatic missense mutations affecting the RNase IIIb domain of DICER1 are common in nonepithelial ovarian tumors. These mutations do not obliterate DICER1 function but alter it in specific cell types, a novel mechanism through which perturbation of microRNA processing may be oncogenic. (Funded by the Terry Fox Research Institute and others.).


Asunto(s)
ARN Helicasas DEAD-box/genética , Mutación Missense , Neoplasias Ováricas/genética , Ribonucleasa III/genética , Tumor de Células de Sertoli-Leydig/genética , Carcinosarcoma/genética , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Mutación de Línea Germinal , Humanos , MicroARNs/metabolismo , Neoplasias de Células Germinales y Embrionarias/genética , Rabdomiosarcoma/genética , Análisis de Secuencia de ADN
6.
N Engl J Med ; 363(16): 1532-43, 2010 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-20942669

RESUMEN

BACKGROUND: Ovarian clear-cell and endometrioid carcinomas may arise from endometriosis, but the molecular events involved in this transformation have not been described. METHODS: We sequenced the whole transcriptomes of 18 ovarian clear-cell carcinomas and 1 ovarian clear-cell carcinoma cell line and found somatic mutations in ARID1A (the AT-rich interactive domain 1A [SWI-like] gene) in 6 of the samples. ARID1A encodes BAF250a, a key component of the SWI­SNF chromatin remodeling complex. We sequenced ARID1A in an additional 210 ovarian carcinomas and a second ovarian clear-cell carcinoma cell line and measured BAF250a expression by means of immunohistochemical analysis in an additional 455 ovarian carcinomas. RESULTS: ARID1A mutations were seen in 55 of 119 ovarian clear-cell carcinomas (46%), 10 of 33 endometrioid carcinomas (30%), and none of the 76 high-grade serous ovarian carcinomas. Seventeen carcinomas had two somatic mutations each. Loss of the BAF250a protein correlated strongly with the ovarian clear-cell carcinoma and endometrioid carcinoma subtypes and the presence of ARID1A mutations. In two patients, ARID1A mutations and loss of BAF250a expression were evident in the tumor and contiguous atypical endometriosis but not in distant endometriotic lesions. CONCLUSIONS: These data implicate ARID1A as a tumor-suppressor gene frequently disrupted in ovarian clear-cell and endometrioid carcinomas. Since ARID1A mutation and loss of BAF250a can be seen in the preneoplastic lesions, we speculate that this is an early event in the transformation of endometriosis into cancer. (Funded by the British Columbia Cancer Foundation and the Vancouver General Hospital­University of British Columbia Hospital Foundation.).


Asunto(s)
Adenocarcinoma de Células Claras/genética , Carcinoma Endometrioide/genética , Endometriosis/complicaciones , Mutación , Proteínas Nucleares/genética , Neoplasias Ováricas/genética , Factores de Transcripción/genética , Adenocarcinoma de Células Claras/metabolismo , Adenocarcinoma de Células Claras/patología , Carcinoma Endometrioide/metabolismo , Carcinoma Endometrioide/patología , Línea Celular Tumoral , Proteínas de Unión al ADN , Endometriosis/patología , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Genes Supresores de Tumor , Humanos , Proteínas Nucleares/metabolismo , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Análisis de Secuencia de ARN , Factores de Transcripción/metabolismo
7.
Blood ; 117(8): 2451-9, 2011 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-21190999

RESUMEN

Next-generation sequencing of follicular lymphoma and diffuse-large B-cell lymphoma has revealed frequent somatic, heterozygous Y641 mutations in the histone methyltransferase EZH2. Heterozygosity and the presence of equal quantities of both mutant and wild-type mRNA and expressed protein suggest a dominant mode of action. Surprisingly, B-cell lymphoma cell lines and lymphoma samples harboring heterozygous EZH2(Y641) mutations have increased levels of histone H3 Lys-27-specific trimethylation (H3K27me3). Expression of EZH2(Y641F/N) mutants in cells with EZH2(WT) resulted in an increase of H3K27me3 levels in vivo. Structural modeling of EZH2(Y641) mutants suggests a "Tyr/Phe switch" model whereby structurally neutral, nontyrosine residues at position 641 would decrease affinity for unmethylated and monomethylated H3K27 substrates and potentially favor trimethylation. We demonstrate, using in vitro enzyme assays of reconstituted PRC2 complexes, that Y641 mutations result in a decrease in monomethylation and an increase in trimethylation activity of the enzyme relative to the wild-type enzyme. This represents the first example of a disease-associated gain-of-function mutation in a histone methyltransferase, whereby somatic EZH2 Y641 mutations in lymphoma act dominantly to increase, rather than decrease, histone methylation. The dominant mode of action suggests that allele-specific EZH2 inhibitors should be a future therapeutic strategy for this disease.


Asunto(s)
Proteínas de Unión al ADN/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Linfoma de Células B Grandes Difuso/genética , Mutación Missense , Factores de Transcripción/genética , Biopsia , Catálisis , Línea Celular Tumoral , Proteína Potenciadora del Homólogo Zeste 2 , Humanos , Linfoma/genética , Metilación , Modelos Moleculares , Complejo Represivo Polycomb 2 , Especificidad por Sustrato
8.
Blood ; 118(12): 3350-8, 2011 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-21628414

RESUMEN

Processing of pre-miRNA through Dicer1 generates an miRNA duplex that consists of an miRNA and miRNA* strand. Despite the general view that miRNA*s have no functional role, we further investigated miRNA* species in 10 deep-sequencing libraries from mouse and human tissue. Comparisons of miRNA/miRNA* ratios across the miRNA sequence libraries revealed that 50% of the investigated miRNA duplexes exhibited a highly dominant strand. Conversely, 10% of miRNA duplexes showed a comparable expression of both strands, whereas the remaining 40% exhibited variable ratios across the examined libraries, as exemplified by miR-223/miR-223* in murine and human cell lines. Functional analyses revealed a regulatory role for miR-223* in myeloid progenitor cells, which implies an active role for both arms of the miR-223 duplex. This was further underscored by the demonstration that miR-223 and miR-223* targeted the insulin-like growth factor 1 receptor/phosphatidylinositol 3-kinase axis and that high miR-223* levels were associated with increased overall survival in patients with acute myeloid leukemia. Thus, we found a supporting role for miR-223* in differentiating myeloid cells in normal and leukemic cell states. The fact that the miR-223 duplex acts through both arms extends the complexity of miRNA-directed gene regulation of this myeloid key miRNA.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Leucemia Mieloide Aguda/genética , MicroARNs , Células Progenitoras Mieloides/metabolismo , Hibridación de Ácido Nucleico/métodos , Fosfatidilinositol 3-Quinasas/metabolismo , Receptores de Superficie Celular/metabolismo , Ribonucleasa III/metabolismo , Transducción de Señal , Adolescente , Adulto , Animales , Diferenciación Celular/genética , Línea Celular Tumoral , Proliferación Celular , ARN Helicasas DEAD-box/genética , ADN Complementario/análisis , ADN Complementario/biosíntesis , Genes Reporteros , Vectores Genéticos , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/mortalidad , Leucemia Mieloide Aguda/patología , Luciferasas/análisis , Masculino , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Persona de Mediana Edad , Células Progenitoras Mieloides/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosfatidilinositol 3-Quinasas/genética , Receptores de Superficie Celular/genética , Retroviridae , Ribonucleasa III/genética , Transducción de Señal/genética , Bibliotecas de Moléculas Pequeñas/análisis , Tasa de Supervivencia , Transfección
9.
Cancers (Basel) ; 15(10)2023 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-37345142

RESUMEN

CIC encodes a transcriptional repressor and MAPK signalling effector that is inactivated by loss-of-function mutations in several cancer types, consistent with a role as a tumour suppressor. Here, we used bioinformatic, genomic, and proteomic approaches to investigate CIC's interaction networks. We observed both previously identified and novel candidate interactions between CIC and SWI/SNF complex members, as well as novel interactions between CIC and cell cycle regulators and RNA processing factors. We found that CIC loss is associated with an increased frequency of mitotic defects in human cell lines and an in vivo mouse model and with dysregulated expression of mitotic regulators. We also observed aberrant splicing in CIC-deficient cell lines, predominantly at 3' and 5' untranslated regions of genes, including genes involved in MAPK signalling, DNA repair, and cell cycle regulation. Our study thus characterises the complexity of CIC's functional network and describes the effect of its loss on cell cycle regulation, mitotic integrity, and transcriptional splicing, thereby expanding our understanding of CIC's potential roles in cancer. In addition, our work exemplifies how multi-omic, network-based analyses can be used to uncover novel insights into the interconnected functions of pleiotropic genes/proteins across cellular contexts.

10.
N Engl J Med ; 360(26): 2719-29, 2009 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-19516027

RESUMEN

BACKGROUND: Granulosa-cell tumors (GCTs) are the most common type of malignant ovarian sex cord-stromal tumor (SCST). The pathogenesis of these tumors is unknown. Moreover, their histopathological diagnosis can be challenging, and there is no curative treatment beyond surgery. METHODS: We analyzed four adult-type GCTs using whole-transcriptome paired-end RNA sequencing. We identified putative GCT-specific mutations that were present in at least three of these samples but were absent from the transcriptomes of 11 epithelial ovarian tumors, published human genomes, and databases of single-nucleotide polymorphisms. We confirmed these variants by direct sequencing of complementary DNA and genomic DNA. We then analyzed additional tumors and matched normal genomic DNA, using a combination of direct sequencing, analyses of restriction-fragment-length polymorphisms, and TaqMan assays. RESULTS: All four index GCTs had a missense point mutation, 402C-->G (C134W), in FOXL2, a gene encoding a transcription factor known to be critical for granulosa-cell development. The FOXL2 mutation was present in 86 of 89 additional adult-type GCTs (97%), in 3 of 14 thecomas (21%), and in 1 of 10 juvenile-type GCTs (10%). The mutation was absent in 49 SCSTs of other types and in 329 unrelated ovarian or breast tumors. CONCLUSIONS: Whole-transcriptome sequencing of four GCTs identified a single, recurrent somatic mutation (402C-->G) in FOXL2 that was present in almost all morphologically identified adult-type GCTs. Mutant FOXL2 is a potential driver in the pathogenesis of adult-type GCTs.


Asunto(s)
Factores de Transcripción Forkhead/genética , Tumor de Células de la Granulosa/genética , Mutación Missense , Neoplasias Ováricas/genética , Secuencia de Bases , Femenino , Proteína Forkhead Box L2 , Perfilación de la Expresión Génica , Marcadores Genéticos , Genotipo , Tumor de Células de la Granulosa/diagnóstico , Tumor de Células de la Granulosa/patología , Humanos , Inmunohistoquímica , Neoplasias Ováricas/diagnóstico , Neoplasias Ováricas/patología , Mutación Puntual , Análisis de Secuencia de ARN , Polimerasa Taq
11.
BMC Genomics ; 12: 209, 2011 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-21527035

RESUMEN

BACKGROUND: Humans and mice with loss of function mutations in GPR54 (KISS1R) or kisspeptin do not progress through puberty, caused by a failure to release GnRH. The transcriptional networks regulated by these proteins in the hypothalamus have yet to be explored by genome-wide methods. RESULTS: We show here, using 1 million exon mouse arrays (Exon 1.0 Affymetrix) and quantitative polymerase chain reaction (QPCR) validation to analyse microdissected hypothalamic tissue from Gpr54 and Kiss1 knockout mice, the extent of transcriptional regulation in the hypothalamus. The sensitivity to detect important transcript differences in microdissected RNA was confirmed by the observation of counter-regulation of Kiss1 expression in Gpr54 knockouts and confirmed by immunohistochemistry (IHC). Since Gpr54 and Kiss1 knockout animals are effectively pre-pubertal with low testosterone (T) levels, we also determined which of the validated transcripts were T-responsive and which varied according to genotype alone. We observed four types of transcriptional regulation (i) genotype only dependent regulation, (ii) T only dependent regulation, (iii) genotype and T-dependent regulation with interaction between these variables, (iv) genotype and T-dependent regulation with no interaction between these variables. The results implicate for the first time several transcription factors (e.g. Npas4, Esr2), proteases (Klk1b22), and the orphan 10-transmembrane transporter TMEM144 in the biology of GPR54/kisspeptin function in the hypothalamus. We show for the neuronal activity regulated transcription factor NPAS4, that distinct protein over-expression is seen in the hypothalamus and hippocampus in Gpr54 knockout mice. This links for the first time the hypothalamic-gonadal axis with this important regulator of inhibitory synapse formation. Similarly we confirm TMEM144 up-regulation in the hypothalamus by RNA in situ hybridization and western blot. CONCLUSIONS: Taken together, global transcriptional profiling shows that loss of GPR54 and kisspeptin are not fully equivalent in the mouse hypothalamus.


Asunto(s)
Redes Reguladoras de Genes , Hipotálamo/metabolismo , Proteínas/genética , Proteínas/metabolismo , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/genética , Testosterona/metabolismo , Animales , Genotipo , Kisspeptinas , Masculino , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Kisspeptina-1 , Transcripción Genética
12.
Anal Chem ; 83(16): 6254-8, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21702506

RESUMEN

We present a method to perform absolute quantification of glycogen in human embryonic stem cells (hESCs) in situ based on the use of Raman microspectroscopy. The proposed quantification method was validated by comparison to a commonly used commercial glycogen assay kit. With Raman microspectroscopy, we could obtain the glycogen content of hESCs faster and apparently more accurately than with the kit. In addition, glycogen distributions across a colony could be obtained. Raman spectroscopy can provide reliable estimates of the in situ glycogen content in hESCs, and this approach should also be extensible to their other biochemical constituents as well as to other cell types.


Asunto(s)
Células Madre Embrionarias/química , Glucógeno/análisis , Espectrometría Raman/métodos , Células Madre Embrionarias/citología , Humanos , Juego de Reactivos para Diagnóstico
13.
Mod Pathol ; 24(1): 64-81, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20852590

RESUMEN

P-cadherin is a calcium-dependent cell-cell adhesion glycoprotein. P-cadherin expression is restricted to the myoepithelial cells in normal breast tissue, and aberrant staining has also been described in invasive tumors. Several small studies have reported P-cadherin as a marker of poor outcome in breast cancer patients but its prognostic significance in relation to other variables has not been established in a large series of breast cancers. A tissue microarray was constructed from 3992 cases of invasive breast carcinoma, and P-cadherin expression was evaluated using immunohistochemistry. Median follow-up was 12.5 years. The immunohistochemistry-based definitions of cancer subtypes were luminal (ER+ or PR+/HER2-), luminal/HER2+ (ER+ or PR+/HER2+), HER2+ (ER-/PR-/HER2+), and basal (ER-/PR-/HER2-/CK5/6+ or EGFR+). Clinical covariate and biomarker associations were assessed using contingency tables, and Pearson's χ(2) or Fisher's exact test. Survival associations were assessed using Kaplan-Meier plots, logrank and Breslow tests, and Cox proportional hazards regression analysis. P-cadherin was expressed in 34.8% (1290/3710, 50% cut point) of cases. P-cadherin staining was strongly associated with HER2+ and basal carcinoma subtypes (P<0.0005). P-cadherin-positive patients showed significantly poorer short-term (0-10 years) overall survival, disease-specific survival, distant relapse-free interval, and locoregional relapse-free interval in univariable models (P<0.05). In multivariable Cox models containing standard clinical covariates and cancer subtypes, P-cadherin did not show independent prognostic value. P-cadherin expression was positively associated with histological grade, chemotherapy, Ki-67, EGFR, CK5/6, p53, YB-1, and HER2 expression (P<0.002), and negatively associated with age at diagnosis, ER, PR, and Bcl-2 expression (P<0.0005). This study shows the value of P-cadherin as a marker of poor prognosis. The large sample size of this series clarifies contradictory findings of many smaller studies. P-cadherin positivity is associated with high-grade tumor subtypes and well-established markers of poor prognosis, and may represent a promising antibody therapeutic target.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Cadherinas/metabolismo , Carcinoma Ductal de Mama/metabolismo , Carcinoma Lobular/metabolismo , Adulto , Anciano , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/mortalidad , Carcinoma Ductal de Mama/patología , Carcinoma Lobular/mortalidad , Carcinoma Lobular/patología , Femenino , Humanos , Persona de Mediana Edad , Pronóstico , Tasa de Supervivencia , Análisis de Matrices Tisulares
14.
Blood ; 113(7): 1432-43, 2009 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-18854576

RESUMEN

MLL5 is a divergent member of the Drosophila Trithorax-related (SET) domain and plant homeodomain (PHD) domain-containing chromatin regulators that are involved in the regulation of transcriptional "memory" during differentiation. Human MLL5 is located on chromosome 7q22, which frequently is deleted in myeloid leukemias, suggesting a possible role in hemopoiesis. To address this question, we generated a loss-of-function allele (Mll5(tm1Apa)) in the murine Mll5 locus. Unlike other Mll genes, Mll5(tm1Apa) homozygous mice are viable but display defects in immunity and hematopoiesis. First, Mll5(tm1Apa) homozygous mice show increased susceptibility to spontaneous eye infections, associated with a cell-autonomous impairment of neutrophil function. Second, Mll5(tm1Apa/tm1Apa) mice exhibit a mild impairment of erythropoiesis. Third, Mll5(tm1Apa/tm1Apa) hematopoietic stem cells (HSCs) have impaired competitive repopulating capacity both under normal conditions and when subjected to self-renewal stimulation by NUP98-HOXA10. Fourth, Mll5(tm1Apa) homozygous HSCs show a dramatic sensitivity to DNA demethylation-induced differentiation (5-azadeoxycytidine). Taken together, our data show that MLL5 is involved in terminal myeloid differentiation and the regulation of HSC self-renewal by a mechanism that involves DNA methylation. These data warrant investigation of MLL5 expression levels as a predictive marker of demethylating-agent response in patients with myelodysplastic syndromes and leukemias and identify MLL5 as a key regulator of normal hematopoiesis.


Asunto(s)
Metilación de ADN/fisiología , Hematopoyesis/inmunología , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Neutrófilos/inmunología , Animales , Antimetabolitos Antineoplásicos/farmacología , Azacitidina/análogos & derivados , Azacitidina/farmacología , Infecciones Bacterianas/genética , Infecciones Bacterianas/inmunología , Blefaritis/genética , Blefaritis/inmunología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Decitabina , Genotipo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Homocigoto , Ratones , Ratones Noqueados , Neutrófilos/citología
15.
J Pathol ; 220(2): 307-15, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19921711

RESUMEN

Next-generation DNA sequencing devices have revolutionized cancer genomics by bringing whole genome resequencing of patients' tumours within practical and economic reach. We present an overview of the techniques involved and review early results from the resequencing of cancer genomes. The possible impacts of whole-genome and trancriptome resequencing in clinical cancer research and the practice of pathology are discussed.


Asunto(s)
Neoplasias/genética , Patología Molecular/tendencias , Análisis de Secuencia de ADN/métodos , ADN de Neoplasias/genética , Genoma Humano , Humanos , Mutación , Neoplasias/patología , Análisis de Secuencia de ADN/tendencias
16.
Proc Natl Acad Sci U S A ; 105(42): 16374-9, 2008 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-18922781

RESUMEN

The dsRNA-binding protein Staufen was the first RNA-binding protein proven to play a role in RNA localization in Drosophila. A mammalian homolog, Staufen1 (Stau1), has been implicated in dendritic RNA localization in neurons, translational control, and mRNA decay. However, the precise mechanisms by which it fulfills these specific roles are only partially understood. To determine its physiological functions, the murine Stau1 gene was disrupted by homologous recombination. Homozygous stau1(tm1Apa) mutant mice express a truncated Stau1 protein lacking the functional RNA-binding domain 3. The level of the truncated protein is significantly reduced. Cultured hippocampal neurons derived from stau1(tm1Apa) homozygous mice display deficits in dendritic delivery of Stau1-EYFP and beta-actin mRNA-containing ribonucleoprotein particles (RNPs). Furthermore, these neurons have a significantly reduced dendritic tree and develop fewer synapses. Homozygous stau1(tm1Apa) mutant mice are viable and show no obvious deficits in development, fertility, health, overall brain morphology, and a variety of behavioral assays, e.g., hippocampus-dependent learning. However, we did detect deficits in locomotor activity. Our data suggest that Stau1 is crucial for synapse development in vitro but not critical for normal behavioral function.


Asunto(s)
Alelos , Dendritas/metabolismo , Proteínas de Unión al ARN/metabolismo , Ribonucleoproteínas/metabolismo , Animales , Regulación de la Expresión Génica , Hipocampo/metabolismo , Homocigoto , Locomoción , Ratones , Mutación/genética , Unión Proteica , ARN/metabolismo , Proteínas de Unión al ARN/genética
17.
Nat Commun ; 12(1): 5406, 2021 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-34518533

RESUMEN

DNA methylation is aberrant in cancer, but the dynamics, regulatory role and clinical implications of such epigenetic changes are still poorly understood. Here, reduced representation bisulfite sequencing (RRBS) profiles of 1538 breast tumors and 244 normal breast tissues from the METABRIC cohort are reported, facilitating detailed analysis of DNA methylation within a rich context of genomic, transcriptional, and clinical data. Tumor methylation from immune and stromal signatures are deconvoluted leading to the discovery of a tumor replication-linked clock with genome-wide methylation loss in non-CpG island sites. Unexpectedly, methylation in most tumor CpG islands follows two replication-independent processes of gain (MG) or loss (ML) that we term epigenomic instability. Epigenomic instability is correlated with tumor grade and stage, TP53 mutations and poorer prognosis. After controlling for these global trans-acting trends, as well as for X-linked dosage compensation effects, cis-specific methylation and expression correlations are uncovered at hundreds of promoters and over a thousand distal elements. Some of these targeted known tumor suppressors and oncogenes. In conclusion, this study demonstrates that global epigenetic instability can erode cancer methylomes and expose them to localized methylation aberrations in-cis resulting in transcriptional changes seen in tumors.


Asunto(s)
Neoplasias de la Mama/genética , Metilación de ADN , Epigénesis Genética , Epigenómica/métodos , Regulación Neoplásica de la Expresión Génica , Estudios de Cohortes , Islas de CpG/genética , Replicación del ADN/genética , Femenino , Genoma Humano/genética , Inestabilidad Genómica/genética , Genómica/métodos , Humanos , Células MCF-7 , Mutación , Regiones Promotoras Genéticas/genética , Análisis de Supervivencia
18.
Cell Metab ; 1(5): 293-6, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-16054076

RESUMEN

Recent genetic evidence in humans and from mouse knockouts has linked kisspeptin-driven GPR54 signaling to the regulation of GnRH release from the hypothalamus. These molecules appear to represent a previously unsuspected control point for GnRH secretion, with important implications for the biology and pathology of the sex steroid axis.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Proteínas/metabolismo , Receptores de Neuropéptido/metabolismo , Animales , Hormonas Esteroides Gonadales/metabolismo , Humanos , Kisspeptinas , Ratones , Modelos Biológicos , Receptores Acoplados a Proteínas G , Receptores de Kisspeptina-1 , Transducción de Señal/fisiología , Proteínas Supresoras de Tumor
19.
Breast Cancer Res ; 12(3): R38, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20565980

RESUMEN

INTRODUCTION: Breast cancer is genetically and clinically a heterogeneous disease. However, the exact contribution of different cell types and oncogenic mutations to this heterogeneity are not well understood. Recently, we discovered an interaction between Wnt and integrin-linked kinase (ILK) within the signaling cascade that regulates cell growth and survival. Interestingly, mammary-specific expression of either one of these proteins has been shown to promote mammary tumorigenesis. In light of our recent findings and to investigate the potential interaction between Wnt and ILK proteins during mammary tumor formation and progression, we established a transgenic mouse model that expresses both Wnt and ILK in mammary epithelial cells. METHODS: A novel transgenic mouse model with mammary-specific expression of both Wnt1 and ILK was generated by crossing the two previously characterized mouse models, MMTV-Wnt1 and MMTV-ILK. The resulting MMTV-Wnt/ILK mice were closely monitored for tumor development and growth, as well as for the tumor onset. The molecular phenotypes of both tumors and premalignant mammary glands were investigated by using biochemical and global gene-expression analysis approaches. RESULTS: A significant acceleration in mammary tumor incidence and growth was observed in the MMTV-Wnt/ILK mice. Pre-neoplastic mammary glands also display lobuloalveolar hyperplasia and an increase in ductal epithelium proliferation. Apart from elevated expression of Wnt/ILK targets, such as beta-catenin and cyclin D1, gene-expression profiling identified the surprising activation of the FOXA1 transcription factor. Upregulation of FOXA1, which is also known as the molecular marker of differentiated mammary luminal cells, was consistent with the expansion of the enriched luminal progenitor population or CD29loCD24hiCD61+ cells in MMTV-Wnt/ILK tumors. CONCLUSIONS: These results show cooperation between Wnt1 and ILK transgenes during mammary carcinogenesis, leading to changes in a transcriptional network, which could dictate a specific breast cancer phenotype with enhanced growth dynamics. The MMTV-Wnt/ILK can be used as a model to identify further the genes downstream of the estrogen receptor-beta/FOXA1 and to investigate the mechanisms targeting the expansion of the luminal progenitor cells leading to hyperplasia and tumorigenesis.


Asunto(s)
Transformación Celular Neoplásica , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Proteínas Serina-Treonina Quinasas/fisiología , Transducción de Señal , Proteína Wnt1/fisiología , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Células Epiteliales/patología , Femenino , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Humanos , Masculino , Neoplasias Mamarias Animales/genética , Virus del Tumor Mamario del Ratón/genética , Ratones , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Cytometry A ; 73(10): 904-17, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18698634

RESUMEN

High-content microscopic screening systems are powerful tools for extracting quantitative multiparameter measures from large number of cells under numerous conditions. These systems perform well in applications that monitor the presence of objects, but lack in their ability to accurately estimate object intensities and summarize these findings due to variations in background, aberrations in illumination, and variability in staining over the image and/or sample wells. We present effective and automated methods that are applicable to analyzing intensity-based cell cycle assays under high-throughput screening conditions. We characterize the system aberration response from images of calibration beads and then enhance the detection and segmentation accuracy of traditional algorithms by preprocessing images for local background variations. We also provide a rapid, adaptive, cell-cycle partitioning algorithm to characterize each sample well based on the estimated locally and globally corrected cell intensity measures of BrdU and DAPI incorporation. We demonstrated the utility and range of our cell ploidy and probe density measurement methods in a pilot screen using a siRNA library against 779 human protein kinases. With our method, multiple image-based quantitative phenotypes can be realized from a single high-throughput image-based microtiter-plate screen.


Asunto(s)
Ciclo Celular , Citometría de Flujo , Procesamiento de Imagen Asistido por Computador/normas , Proteínas Quinasas/genética , ARN Interferente Pequeño/genética , Algoritmos , Bromodesoxiuridina/análisis , Bromodesoxiuridina/metabolismo , Calibración , Línea Celular Tumoral , Separación Celular , Silenciador del Gen , Humanos , Indoles/análisis , Indoles/metabolismo , Coloración y Etiquetado
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA