Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Genome Res ; 29(4): 521-531, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30846532

RESUMEN

Humans are frequently exposed to acrylamide, a probable human carcinogen found in commonplace sources such as most heated starchy foods or tobacco smoke. Prior evidence has shown that acrylamide causes cancer in rodents, yet epidemiological studies conducted to date are limited and, thus far, have yielded inconclusive data on association of human cancers with acrylamide exposure. In this study, we experimentally identify a novel and unique mutational signature imprinted by acrylamide through the effects of its reactive metabolite glycidamide. We next show that the glycidamide mutational signature is found in a full one-third of approximately 1600 tumor genomes corresponding to 19 human tumor types from 14 organs. The highest enrichment of the glycidamide signature was observed in the cancers of the lung (88% of the interrogated tumors), liver (73%), kidney (>70%), bile duct (57%), cervix (50%), and, to a lesser extent, additional cancer types. Overall, our study reveals an unexpectedly extensive contribution of acrylamide-associated mutagenesis to human cancers.


Asunto(s)
Acrilamidas/toxicidad , Carcinogénesis/genética , Exposición a Riesgos Ambientales , Mutágenos/toxicidad , Mutación , Neoplasias/genética , Animales , Carcinogénesis/inducido químicamente , Células Cultivadas , Compuestos Epoxi/toxicidad , Genoma Humano , Humanos , Ratones , Neoplasias/inducido químicamente , Proteína p53 Supresora de Tumor/genética
2.
Mol Cell ; 40(4): 533-47, 2010 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-21095584

RESUMEN

In a genome-wide siRNA analysis of p16(INK4a) (p16) modulators, we identify the Hedgehog (Hh) pathway component SUFU and formally demonstrate that Hh signaling promotes mitogenesis by suppression of p16. A fragment of the Hh-responsive GLI2 transcription factor directly binds and inhibits the p16 promoter and senescence is associated with the loss of nuclear GLI2. Hh components partially reside in the primary cilium (PC), and the small fraction of cells in mass culture that elaborate a PC have the lowest expression of p16. Suppression of p16 is effected by both PC-dependent and -independent routes, and ablation of p16 renders cells insensitive to an Hh inhibitor and increases PC formation. These results directly link a well-established developmental mitogenic pathway with a key tumor suppressor and contribute to the molecular understanding of replicative senescence, Hh-mediated oncogenesis, and potentially the role of p16 in aging.


Asunto(s)
Cilios/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Proteínas Hedgehog/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas Nucleares/metabolismo , Transducción de Señal , Femenino , Genoma Humano/genética , Humanos , Mapeo de Interacción de Proteínas , ARN Interferente Pequeño/metabolismo , Adulto Joven , Proteína Gli2 con Dedos de Zinc
3.
Nucleic Acids Res ; 42(3): 1606-18, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24217920

RESUMEN

p16 is a key regulator of cellular senescence, yet the drivers of this stable state of proliferative arrest are not well understood. Here, we identify 22 senescence-associated microRNAs (SA-miRNAs) in normal human mammary epithelial cells. We show that SA-miRNAs-26b, 181a, 210 and 424 function in concert to directly repress expression of Polycomb group (PcG) proteins CBX7, embryonic ectoderm development (EED), enhancer of zeste homologue 2 (EZH2) and suppressor of zeste 12 homologue (Suz12), thereby activating p16. We demonstrate the existence of a tight positive feedback loop in which SA-miRNAs activate and re-enforce the expression of other SA-miRNA members. In contrast, PcG members restrain senescence by epigenetically repressing the expression of these SA-miRNAs. Importantly, loss of p16 leads to repression of SA-miRNA expression, intimately coupling this effector of senescence to the SA-miRNA/PcG self-regulatory loop. Taken together, our findings illuminate an important regulatory axis that underpins the transition from proliferation to cellular senescence.


Asunto(s)
Senescencia Celular/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Epigénesis Genética , MicroARNs/metabolismo , Células Cultivadas , Retroalimentación Fisiológica , Fibroblastos/citología , Fibroblastos/metabolismo , Silenciador del Gen , Humanos , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/metabolismo , Adulto Joven
4.
Genome Res ; 21(12): 2026-37, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21873453

RESUMEN

Epigenetic mechanisms are important regulators of cell type-specific genes, including miRNAs. In order to identify cell type-specific miRNAs regulated by epigenetic mechanisms, we undertook a global analysis of miRNA expression and epigenetic states in three isogenic pairs of human mammary epithelial cells (HMEC) and human mammary fibroblasts (HMF), which represent two differentiated cell types typically present within a given organ, each with a distinct phenotype and a distinct epigenotype. While miRNA expression and epigenetic states showed strong interindividual concordance within a given cell type, almost 10% of the expressed miRNA showed a cell type-specific pattern of expression that was linked to the epigenetic state of their promoter. The tissue-specific miRNA genes were epigenetically repressed in nonexpressing cells by DNA methylation (38%) and H3K27me3 (58%), with only a small set of miRNAs (21%) showing a dual epigenetic repression where both DNA methylation and H3K27me3 were present at their promoters, such as MIR10A and MIR10B. Individual miRNA clusters of closely related miRNA gene families can each display cell type-specific repression by the same or complementary epigenetic mechanisms, such as the MIR200 family, and MIR205, where fibroblasts repress MIR200C/141 by DNA methylation, MIR200A/200B/429 by H3K27me3, and MIR205 by both DNA methylation and H3K27me3. Since deregulation of many of the epigenetically regulated miRNAs that we identified have been linked to disease processes such as cancer, it is predicted that compromise of the epigenetic control mechanisms is important for this process. Overall, these results highlight the importance of epigenetic regulation in the control of normal cell type-specific miRNA expression.


Asunto(s)
Mama/metabolismo , Epigénesis Genética/fisiología , Fibroblastos/metabolismo , MicroARNs/biosíntesis , Mama/citología , Línea Celular , Femenino , Fibroblastos/citología , Humanos , Especificidad de Órganos/fisiología
5.
Am J Pathol ; 183(5): 1645-1653, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24012678

RESUMEN

Both epigenetic silencing and genetic deletion of tumor suppressors contribute to the development and progression of breast cancer. SOX7 is a transcription factor important to development, and its down-regulation has been reported in tumor tissues and cell lines of prostate, colon, and lung cancers. However, the regulation of SOX7 expression and its functional role in breast cancer have not been reported. The current study demonstrates that SOX7 mRNA and protein expression are down-regulated in breast cancer tissues and cell lines compared with adjacent normal tissues and nontumorigenic cells, respectively. The SOX7 promoter is hypermethylated in breast cancer cell lines compared with nontumorigenic cells, and the inhibition of DNA methylation increases SOX7 mRNA levels. With shRNA-mediated SOX7 silencing, nontumorigenic immortal breast cells display increased proliferation, migration, and invasion and form structures that resemble that of breast cancer cells in a three-dimensional culture system. Conversely, ectopic SOX7 expression inhibits proliferation, migration, and invasion of breast cancer cells in vitro and tumor growth in vivo. Importantly, we discovered that SOX7 transcript levels positively correlated with clinical outcome of 674 breast cancer patients. Overall, our data suggest that SOX7 acts as a tumor suppressor in breast cancer. SOX7 expression is likely regulated by multiple mechanisms and potentially serves as a prognostic marker for breast cancer patients.


Asunto(s)
Neoplasias de la Mama/genética , Regulación Neoplásica de la Expresión Génica , Factores de Transcripción SOXF/genética , Proteínas Supresoras de Tumor/genética , Animales , Línea Celular Tumoral , Metilación de ADN/genética , Regulación hacia Abajo/genética , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Desnudos , Regiones Promotoras Genéticas , Factores de Transcripción SOXF/metabolismo , Resultado del Tratamiento , Proteínas Supresoras de Tumor/metabolismo
6.
Proc Natl Acad Sci U S A ; 108(8): 3264-9, 2011 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-21300877

RESUMEN

Loss of organization is a principle feature of cancers; therefore it is important to understand how normal adult multilineage tissues, such as bilayered secretory epithelia, establish and maintain their architectures. The self-organization process that drives heterogeneous mixtures of cells to form organized tissues is well studied in embryology and with mammalian cell lines that were abnormal or engineered. Here we used a micropatterning approach that confined cells to a cylindrical geometry combined with an algorithm to quantify changes of cellular distribution over time to measure the ability of different cell types to self-organize relative to each other. Using normal human mammary epithelial cells enriched into pools of the two principal lineages, luminal and myoepithelial cells, we demonstrated that bilayered organization in mammary epithelium was driven mainly by lineage-specific differential E-cadherin expression, but that P-cadherin contributed specifically to organization of the myoepithelial layer. Disruption of the actomyosin network or of adherens junction proteins resulted in either prevention of bilayer formation or loss of preformed bilayers, consistent with continual sampling of the local microenvironment by cadherins. Together these data show that self-organization is an innate and reversible property of communities of normal adult human mammary epithelial cells.


Asunto(s)
Células Epiteliales/citología , Glándulas Mamarias Humanas/citología , Actomiosina/fisiología , Uniones Adherentes/fisiología , Tipificación del Cuerpo , Cadherinas/fisiología , Linaje de la Célula , Células Cultivadas , Técnicas de Cocultivo , Humanos
7.
Genes Chromosomes Cancer ; 52(11): 1017-29, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23929738

RESUMEN

Chromosomal positions of common fragile sites differ in lymphoblasts and fibroblasts, with positions dependent on the epigenetically determined density of replication origins at these loci. Because rearrangement of fragile loci and associated loss of fragile gene products are hallmarks of cancers, we aimed to map common fragile sites in epithelial cells, from which most cancers derive. Among the five most frequently activated sites in human epithelial cells were chromosome bands 2q33 and Xq22.1, which are not among top fragile sites identified in lymphoblasts or fibroblasts. FRA16D at 16q23 was among the top three fragile sites in the human epithelial cells examined, as it is in lymphoblasts and fibroblasts, while FRA3B at 3p14.2, the top fragile locus in lymphoblasts, was not fragile in most epithelial cell lines tested. Epithelial cells exhibited varying hierarchies of fragile sites; some frequent epithelial cell fragile sites are apparently not frequently altered in epithelial cancers and sites that are frequently deleted in epithelial cancers are not necessarily among the most fragile. Since we have reported that loss of expression of the FRA3B-encoded FHIT protein causes increased replication stress-induced DNA damage, we also examined the effect of FHIT-deficiency on markers of genome instability in epithelial cells. FHIT-deficient cells exhibited increases in fragile breaks and in γH2AX and 53BP1 foci in G1 phase cells, confirming in epithelial cells that the FHIT gene and encompassing FRA3B, is a "caretaker gene" necessary for maintenance of genome stability.


Asunto(s)
Ácido Anhídrido Hidrolasas/genética , Sitios Frágiles del Cromosoma/genética , Inestabilidad Genómica , Proteínas de Neoplasias/genética , Neoplasias/genética , Animales , Línea Celular , Cromosomas Humanos Par 2/genética , Análisis Citogenético , Células Epiteliales/citología , Células Epiteliales/fisiología , Fase G1/genética , Marcadores Genéticos , Humanos , Ratones , Interferencia de ARN , Origen de Réplica , Análisis de Área Pequeña , Cromosoma X/genética
8.
Cell Signal ; 113: 110958, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37935340

RESUMEN

Microenvironment signals are potent determinants of cell fate and arbiters of tissue homeostasis, however understanding how different microenvironment factors coordinately regulate cellular phenotype has been experimentally challenging. Here we used a high-throughput microenvironment microarray comprised of 2640 unique pairwise signals to identify factors that support proliferation and maintenance of primary human mammary luminal epithelial cells. Multiple microenvironment factors that modulated luminal cell number were identified, including: HGF, NRG1, BMP2, CXCL1, TGFB1, FGF2, PDGFB, RANKL, WNT3A, SPP1, HA, VTN, and OMD. All of these factors were previously shown to modulate luminal cell numbers in painstaking mouse genetics experiments, or were shown to have a role in breast cancer, demonstrating the relevance and power of our high-dimensional approach to dissect key microenvironmental signals. RNA-sequencing of primary epithelial and stromal cell lineages identified the cell types that express these signals and the cognate receptors in vivo. Cell-based functional studies confirmed which effects from microenvironment factors were reproducible and robust to individual variation. Hepatocyte growth factor (HGF) was the factor most robust to individual variation and drove expansion of luminal cells via cKit+ progenitor cells, which expressed abundant MET receptor. Luminal cells from women who are genetically high risk for breast cancer had significantly more MET receptor and may explain the characteristic expansion of the luminal lineage in those women. In ensemble, our approach provides proof of principle that microenvironment signals that control specific cellular states can be dissected with high-dimensional cell-based approaches.


Asunto(s)
Neoplasias de la Mama , Células Epiteliales , Femenino , Humanos , Animales , Ratones , Células Epiteliales/metabolismo , Diferenciación Celular , Neoplasias de la Mama/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Microambiente Tumoral
9.
Proc Natl Acad Sci U S A ; 105(37): 14076-81, 2008 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-18780791

RESUMEN

Cellular identity and differentiation are determined by epigenetic programs. The characteristics of these programs in normal human mammary epithelium and their similarity to those in stem cells are unknown. To begin investigating these issues, we analyzed the DNA methylation and gene expression profiles of distinct subpopulations of mammary epithelial cells by using MSDK (methylation-specific digital karyotyping) and SAGE (serial analysis of gene expression). We identified discrete cell-type and differentiation state-specific DNA methylation and gene expression patterns that were maintained in a subset of breast carcinomas and correlated with clinically relevant tumor subtypes. CD44+ cells were the most hypomethylated and highly expressed several transcription factors with known stem cell function including HOXA10 and TCF3. Many of these genes were also hypomethylated in BMP4-treated compared with undifferentiated human embryonic stem (ES) cells that we analyzed by MSDK for comparison. Further highlighting the similarity of epigenetic programs of embryonic and mammary epithelial cells, genes highly expressed in CD44+ relative to more differentiated CD24+ cells were significantly enriched for Suz12 targets in ES cells. The expression of FOXC1, one of the transcription factors hypomethylated and highly expressed in CD44+ cells, induced a progenitor-like phenotype in differentiated mammary epithelial cells. These data suggest that epigenetically controlled transcription factors play a key role in regulating mammary epithelial cell phenotypes and imply similarities among epigenetic programs that define progenitor cell characteristics.


Asunto(s)
Mama/metabolismo , Metilación de ADN , Mama/citología , Recuento de Células , Forma de la Célula , Células Epiteliales/citología , Factores de Transcripción Forkhead/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Fenotipo , Células Madre/metabolismo , Especificidad por Sustrato
10.
Nat Aging ; 1(9): 838-849, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-35187501

RESUMEN

During aging in the human mammary gland, luminal epithelial cells lose lineage fidelity by expressing markers normally expressed in myoepithelial cells. We hypothesize that loss of lineage fidelity is a general manifestation of epithelia that are susceptible to cancer initiation. In the present study, we show that histologically normal breast tissue from younger women who are susceptible to breast cancer, as a result of harboring a germline mutation in BRCA1, BRCA2 or PALB2 genes, exhibits hallmarks of accelerated aging. These include proportionately increased luminal epithelial cells that acquired myoepithelial markers, decreased proportions of myoepithelial cells and a basal differentiation bias or failure of differentiation of cKit+ progenitors. High-risk luminal and myoepithelial cells are transcriptionally enriched for genes of the opposite lineage, inflammatory- and cancer-related pathways. We have identified breast-aging hallmarks that reflect a convergent biology of cancer susceptibility, regardless of the specific underlying genetic or age-dependent risk or the associated breast cancer subtype.


Asunto(s)
Neoplasias de la Mama , Glándulas Mamarias Humanas , Humanos , Femenino , Envejecimiento/genética , Mama/patología , Mutación de Línea Germinal/genética , Neoplasias de la Mama/genética , Proteína BRCA1/genética , Proteína BRCA2/genética
11.
Aging Cell ; 20(3): e13318, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33547862

RESUMEN

Senescence, a state of stable growth arrest, plays an important role in ageing and age-related diseases in vivo. Although the INK4/ARF locus is known to be essential for senescence programmes, the key regulators driving p16 and ARF transcription remain largely underexplored. Using siRNA screening for modulators of the p16/pRB and ARF/p53/p21 pathways in deeply senescent human mammary epithelial cells (DS HMECs) and fibroblasts (DS HMFs), we identified EGR2 as a novel regulator of senescence. EGR2 expression is up-regulated during senescence, and its ablation by siRNA in DS HMECs and HMFs transiently reverses the senescent phenotype. We demonstrate that EGR2 activates the ARF and p16 promoters and directly binds to both the ARF and p16 promoters. Loss of EGR2 down-regulates p16 levels and increases the pool of p16- p21- 'reversed' cells in the population. Moreover, EGR2 overexpression is sufficient to induce senescence. Our data suggest that EGR2 is a direct transcriptional activator of the p16/pRB and ARF/p53/p21 pathways in senescence and a novel marker of senescence.


Asunto(s)
Senescencia Celular , Proteína 2 de la Respuesta de Crecimiento Precoz/metabolismo , Adolescente , Adulto , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Glándulas Mamarias Humanas/citología , Unión Proteica , ARN Interferente Pequeño/metabolismo , Proteína de Retinoblastoma/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba , Adulto Joven
12.
Cancer Prev Res (Phila) ; 14(8): 779-794, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34140348

RESUMEN

A robust breast cancer prevention strategy requires risk assessment biomarkers for early detection. We show that expression of ELF5, a transcription factor critical for normal mammary development, is downregulated in mammary luminal epithelia with age. DNA methylation of the ELF5 promoter is negatively correlated with expression in an age-dependent manner. Both ELF5 methylation and gene expression were used to build biological clocks to estimate chronological ages of mammary epithelia. ELF5 clock-based estimates of biological age in luminal epithelia from average-risk women were within three years of chronological age. Biological ages of breast epithelia from BRCA1 or BRCA2 mutation carriers, who were high risk for developing breast cancer, suggested they were accelerated by two decades relative to chronological age. The ELF5 DNA methylation clock had better performance at predicting biological age in luminal epithelial cells as compared with two other epigenetic clocks based on whole tissues. We propose that the changes in ELF5 expression or ELF5-proximal DNA methylation in luminal epithelia are emergent properties of at-risk breast tissue and constitute breast-specific biological clocks. PREVENTION RELEVANCE: ELF5 expression or DNA methylation level at the ELF5 promoter region can be used as breast-specific biological clocks to identify women at higher than average risk of breast cancer.


Asunto(s)
Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/genética , Mama/metabolismo , Relojes Circadianos/genética , Proteínas de Unión al ADN/genética , Factores de Transcripción/genética , Adulto , Biomarcadores de Tumor/genética , Mama/patología , Neoplasias de la Mama/patología , Transformación Celular Neoplásica , Células Cultivadas , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Detección Precoz del Cáncer/métodos , Femenino , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad/genética , Pruebas Genéticas/métodos , Humanos , Persona de Mediana Edad , Especificidad de Órganos/genética , Regiones Promotoras Genéticas , Factores de Transcripción/metabolismo
13.
iScience ; 23(11): 101649, 2020 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-33103086

RESUMEN

The receptor tyrosine kinase AXL is associated with epithelial plasticity in several solid tumors including breast cancer and AXL-targeting agents are currently in clinical trials. We hypothesized that AXL is a driver of stemness traits in cancer by co-option of a regulatory function normally reserved for stem cells. AXL-expressing cells in human mammary epithelial ducts co-expressed markers associated with multipotency, and AXL inhibition abolished colony formation and self-maintenance activities while promoting terminal differentiation in vitro. Axl-null mice did not exhibit a strong developmental phenotype, but enrichment of Axl + cells was required for mouse mammary gland reconstitution upon transplantation, and Axl-null mice had reduced incidence of Wnt1-driven mammary tumors. An AXL-dependent gene signature is a feature of transcriptomes in basal breast cancers and reduced patient survival irrespective of subtype. Our interpretation is that AXL regulates access to epithelial plasticity programs in MaSCs and, when co-opted, maintains acquired stemness in breast cancer cells.

14.
Front Cell Dev Biol ; 7: 174, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31555644

RESUMEN

Preventing breast cancer before it is able to form is an ideal way to stop breast cancer. However, there are limited existing options for prevention of breast cancer. Changes in the breast tissue resulting from the aging process contribute to breast cancer susceptibility and progression and may therefore provide promising targets for prevention. Here, we describe new potential targets, immortalization and inflammaging, that may be useful for prevention of age-related breast cancers. We also summarize existing studies of warfarin and metformin, current drugs used for non-cancerous diseases, that also may be repurposed for breast cancer prevention.

15.
Aging (Albany NY) ; 11(5): 1510-1523, 2019 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-30875333

RESUMEN

Aging is a degenerative process in which genome instability plays a crucial role. To gain insight into the link between organismal aging and DNA repair capacity, we analyzed DNA double-strand break (DSB) resolution efficiency in human mammary epithelial cells from 12 healthy donors of young and old ages. The frequency of DSBs was measured by quantifying the number of γH2AX foci before and after 1Gy of γ-rays and it was higher in cells from aged donors (ADs) at all times analyzed. At 24 hours after irradiation, ADs retained a significantly higher frequency of residual DSBs than young donors (YDs), which had already reached values close to basal levels. The kinetics of DSB induction and disappearance showed that cells from ADs and YDs repair DSBs with similar speed, although analysis of early times after irradiation indicate that a repair defect may lie within the firing of the DNA repair machinery in AD cells. Indeed, using a mathematical model we calculated a constant factor of delay affecting aged human epithelial cells repair kinetics. This defect manifests with the accumulation of DSBs that might eventually undergo illegitimate repair, thus posing a relevant threat to the maintenance of genome integrity in older individuals.


Asunto(s)
Reparación del ADN/fisiología , Células Epiteliales/fisiología , Histonas/metabolismo , Glándulas Mamarias Humanas/citología , Adolescente , Adulto , Anciano , Neoplasias de la Mama/radioterapia , Células Cultivadas , Roturas del ADN de Doble Cadena , Femenino , Regulación de la Expresión Génica , Histonas/genética , Humanos , Persona de Mediana Edad , Adulto Joven
16.
Life Sci Space Res (Amst) ; 20: 101-112, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30797427

RESUMEN

There exists a wide degree of genetic variation within the normal human population which includes disease free individuals with heterozygote defects in major DNA repair genes. A lack of understanding of how this genetic variation impacts cellular phenotypes that inform cancer risk post heavy ion exposure poses a major limitation in developing personalized cancer risk assessment astronauts. We initiated a pilot study with Human Mammary Epithelial Cell strains (HMEC) derived from wild type, a p16 silenced derivative of wild type, and various genetic variants that were heterozygote for DNA repair genes; BRCA1, BRCA2 and ATM. Cells strains were exposed to different high and low LET radiation qualities to generate both simple and complex lesions and centrosome aberrations were examined as a surrogate marker of genomic instability and cancer susceptibility post different exposures. Our results indicate that centrosome aberration frequency is higher in the genetic variants under study. The aberration frequency increases with dose, complexity of the lesion generated by different radiation qualities and age of the individual. This increase in genomic instability correlates with elevated check-point activation post radiation exposure. These studies suggest that the influence of individual genetics on cell cycle regulation could modify the degree of early genomic instability in response to complex lesions and potentially define cancer predisposition in response to HZE exposure. These results will have significant implications in estimating cancer susceptibility in genetically variant individuals exposed to HZE particles.


Asunto(s)
Proteína BRCA1/genética , Proteína BRCA2/genética , Neoplasias de la Mama/patología , Mama/patología , Aberraciones Cromosómicas , Radiación Cósmica , Variación Genética , Mama/metabolismo , Mama/efectos de la radiación , Neoplasias de la Mama/genética , Neoplasias de la Mama/radioterapia , Células Cultivadas , Daño del ADN , Femenino , Humanos , Fenotipo , Fosfoproteínas , Proyectos Piloto
17.
J Cell Biol ; 218(8): 2492-2513, 2019 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-31270138

RESUMEN

The spatial organization of the genome is enigmatic. Direct evidence of physical contacts between chromosomes and their visualization at nanoscale resolution has been limited. We used superresolution microscopy to demonstrate that ribosomal DNA (rDNA) can form linkages between chromosomes. We observed rDNA linkages in many different human cell types and demonstrated their resolution in anaphase. rDNA linkages are coated by the transcription factor UBF and their formation depends on UBF, indicating that they regularly occur between transcriptionally active loci. Overexpression of c-Myc increases rDNA transcription and the frequency of rDNA linkages, further suggesting that their formation depends on active transcription. Linkages persist in the absence of cohesion, but inhibition of topoisomerase II prevents their resolution in anaphase. We propose that linkages are topological intertwines occurring between transcriptionally active rDNA loci spatially colocated in the same nucleolar compartment. Our findings suggest that active DNA loci engage in physical interchromosomal connections that are an integral and pervasive feature of genome organization.


Asunto(s)
Cromosomas Humanos/metabolismo , ADN Ribosómico/metabolismo , Microscopía/métodos , Anafase/efectos de los fármacos , Animales , Línea Celular , Nucléolo Celular/efectos de los fármacos , Nucléolo Celular/metabolismo , ADN-Topoisomerasas de Tipo II/metabolismo , Humanos , Células Híbridas/efectos de los fármacos , Células Híbridas/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Proteínas del Complejo de Iniciación de Transcripción Pol1/metabolismo , Poliploidía , Unión Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas c-myc/metabolismo , Telomerasa/metabolismo , Inhibidores de Topoisomerasa/farmacología
19.
Radiat Res ; 170(1): 23-32, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18582160

RESUMEN

The human breast is sensitive to radiation carcinogenesis, and genomic instability occurs early in breast cancer development. This study tests the hypothesis that ionizing radiation elicits genomic instability in finite life-span human mammary epithelial cells (HMEC) and asks whether densely ionizing radiation is a more potent inducer of instability. HMEC in a non-proliferative state were exposed to X rays or 1 GeV/nucleon iron ions followed by delayed plating. Karyotypic instability and centrosome aberrations were monitored in expanded clonal isolates. Severe karyotypic instability was common in the progeny of cells that survived X-ray or iron-ion exposure. There was a lower dose threshold for severe karyotypic instability after iron-ion exposure. More than 90% of X-irradiated colonies and >60% of iron-ion-irradiated colonies showed supernumerary centrosomes at levels above the 95% upper confidence limit of the mean for unirradiated clones. A dose response was observed for centrosome aberrations for each radiation type. There was a statistically significant association between the incidence of karyotypic instability and supernumerary centrosomes for iron-ion-exposed colonies and a weaker association for X-irradiated colonies. Thus genomic instability occurs frequently in finite life-span HMEC exposed to sparsely or densely ionizing radiation and may contribute to radiation-induced breast cancer.


Asunto(s)
Senescencia Celular/efectos de la radiación , Centrosoma/patología , Centrosoma/efectos de la radiación , Aberraciones Cromosómicas/efectos de la radiación , Células Epiteliales/efectos de la radiación , Glándulas Mamarias Humanas/efectos de la radiación , Apoptosis/efectos de la radiación , Células Cultivadas , Células Epiteliales/citología , Análisis de Elementos Finitos , Humanos , Hibridación Fluorescente in Situ , Glándulas Mamarias Humanas/citología , Radiación Ionizante
20.
Mol Cell Biol ; 25(10): 3923-33, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15870267

RESUMEN

Epigenetic inactivation of the RASSF1A tumor suppressor by CpG island methylation was frequently detected in cancer. However, the mechanisms of this aberrant DNA methylation are unknown. In the RASSF1A promoter, we characterized four Sp1 sites, which are frequently methylated in cancer. We examined the functional relationship between DNA methylation, histone modification, Sp1 binding, and RASSF1A expression in proliferating human mammary epithelial cells. With increasing passages, the transcription of RASSF1A was dramatically silenced. This inactivation was associated with deacetylation and lysine 9 trimethylation of histone H3 and an impaired binding of Sp1 at the RASSF1A promoter. In mammary epithelial cells that had overcome a stress-associated senescence barrier, a spreading of DNA methylation in the CpG island promoter was observed. When the RASSF1A-silenced cells were treated with inhibitors of DNA methyltransferase and histone deacetylase, binding of Sp1 and expression of RASSF1A reoccurred. In summary, we observed that histone H3 deacetylation and H3 lysine 9 trimethylation occur in the same time window as gene inactivation and precede DNA methylation. Our data suggest that in epithelial cells, histone inactivation may trigger de novo DNA methylation of the RASSF1A promoter and this system may serve as a model for CpG island inactivation of tumor suppressor genes.


Asunto(s)
Cromatina/genética , Cromatina/metabolismo , Metilación de ADN , Silenciador del Gen , Regiones Promotoras Genéticas/genética , Proteínas Supresoras de Tumor/genética , Línea Celular Tumoral , Senescencia Celular/genética , Islas de CpG/genética , ADN/genética , ADN/metabolismo , Humanos , Secuencias Reguladoras de Ácidos Nucleicos/genética , Factor de Transcripción Sp1/metabolismo , Sulfitos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA