Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Hum Mol Genet ; 23(8): 2198-209, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24256810

RESUMEN

Genome-wide association studies of colorectal cancer (CRC) have identified a number of common variants associated with modest risk, including rs3802842 at chromosome 11q23.1. Several genes map to this region but rs3802842 does not map to any known transcribed or regulatory sequences. We reasoned, therefore, that rs3802842 is not the functional single-nucleotide polymorphism (SNP), but is in linkage disequilibrium (LD) with a functional SNP(s). We performed ChIP-seq for histone modifications in SW480 and HCT-116 CRC cells, and incorporated ChIP-seq and DNase I hypersensitivity data available through ENCODE within a 137-kb genomic region containing rs3802842 on 11q23.1. We identified SNP rs10891246 in LD with rs3802842 that mapped within a bidirectional promoter region of genes C11orf92 and C11orf93. Following mutagenesis to the risk allele, the promoter demonstrated lower levels of reporter gene expression. A second SNP rs7130173 was identified in LD with rs3802842 that mapped to a candidate enhancer region, which showed strong unidirectional activity in both HCT-116 and SW480 CRC cells. The risk allele of rs7130173 demonstrated reduced enhancer activity compared with the common allele, and reduced nuclear protein binding affinity in electromobility shift assays compared with the common allele suggesting differential transcription factor (TF) binding. SNPs rs10891246 and rs7130173 are on the same haplotype, and expression quantitative trait loci (eQTL) analyses of neighboring genes implicate C11orf53, C11orf92 and C11orf93 as candidate target genes. These data imply that rs10891246 and rs7130173 are functional SNPs mapping to 11q23.1 and that C11orf53, C11orf92 and C11orf93 represent novel candidate target genes involved in CRC etiology.


Asunto(s)
Mapeo Cromosómico , Cromosomas Humanos Par 11/genética , Neoplasias Colorrectales/genética , Elementos de Facilitación Genéticos/genética , Polimorfismo de Nucleótido Simple/genética , Regiones Promotoras Genéticas/genética , Inmunoprecipitación de Cromatina , Ensayo de Cambio de Movilidad Electroforética , Humanos , Luciferasas/metabolismo , Repeticiones de Microsatélite/genética , Sitios de Carácter Cuantitativo , Factores de Riesgo , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas
2.
Free Radic Biol Med ; 170: 70-84, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33450377

RESUMEN

The development of multicellular organisms involves the unpacking of a complex genetic program. Extensive characterization of discrete developmental steps has revealed the genetic program is controlled by an epigenetic state. Shifting the epigenome is a group of epigenetic enzymes that modify DNA and proteins to regulate cell type specific gene expression. While the role of these modifications in development has been established, the input(s) responsible for electing changes in the epigenetic state remains unknown. Development is also associated with dynamic changes in cellular metabolism, redox, free radical production, and oxygen availability. It has previously been postulated that these changes are causal in development by affecting gene expression. This suggests that oxygen is a morphogenic compound that impacts the removal of epigenetic marks. Likewise, metabolism and reactive oxygen species influence redox signaling through iron and glutathione to limit the availability of key epigenetic cofactors such as α-ketoglutarate, ascorbate, NAD+ and S-adenosylmethionine. Given the close relationship between these cofactors and epigenetic marks it seems likely that the two are linked. Here we describe how changing these inputs might affect the epigenetic state during development to drive gene expression. Combined, these cofactors and reactive oxygen species constitute the epigenetic landscape guiding cells along differing developmental paths.


Asunto(s)
Epigénesis Genética , Histonas , Metilación de ADN , Histonas/metabolismo , Oxidación-Reducción , Oxígeno/metabolismo
3.
Free Radic Biol Med ; 170: 2-5, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33932538

RESUMEN

An epigenetic landscape encompasses a series of dynamic interconnected mechanisms working together to fashion a diverse set of phenotypes from a singular genotype. The epigenetic plasticity observed in disease and development is facilitated by enzymes that create and remove covalent modifications to DNA and histones. Several important discoveries within the past decade have revealed that epigenetic control mechanisms are subject to redox regulation and mitochondrial-to-nuclear retrograde signaling. This has led to our current understanding that the writers and erasers of the epigenome are influenced by several levels of redox and metabolic control including the bioavailability of oxygen, nutrients, and metabolite co-factors necessary for optimal enzyme activity. Thus, these enzymes perceive a cell's redox state, metabolic status, and environmental signals to influence chromatin structure and accessibility to the transcriptional apparatus. Not only are the activities of epigenetic enzymes affected by cellular redox conditions, but also, in feedback loop fashion, genes encoding antioxidant enzymes as well as prooxidant enzymes can be altered in their expression patterns by epigenetic silencing mechanisms. The altered expression of the anti- and prooxidant genes can then contribute to the onset or progression of disease. Epigenetic regulation of gene expression by the confluence of redox biology and gene-environment interactions is an active area of research and our understanding of these links continues to evolve. Given the emergent importance of crosstalk between redox biology and epigenetic regulatory mechanisms, it is timely that this issue should explore the current state of knowledge on this topic and how changes in metabolism and redox flux can result in tectonic shifts of the epigenetic landscape.


Asunto(s)
Metilación de ADN , Epigénesis Genética , Biología , Histonas/metabolismo , Humanos , Oxidación-Reducción
4.
Clin Cancer Res ; 15(11): 3672-9, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19458056

RESUMEN

PURPOSE: Transcriptional regulation of estrogen receptor-alpha (ERalpha) involves both epigenetic mechanisms and trans-active factors, such as TFAP2C, which induces ERalpha transcription through an AP-2 regulatory region in the ERalpha promoter. Attempts to induce endogenous ERalpha expression in ERalpha-negative breast carcinomas by forced overexpression of TFAP2C have not been successful. We hypothesize that epigenetic chromatin structure alters the activity of TFAP2C at the ERalpha promoter. EXPERIMENTAL DESIGN: DNA methylation, histone acetylation, and chromatin accessibility were examined at the ERalpha promoter in a panel of breast carcinoma cell lines. TFAP2C and polymerase II binding were analyzed by chromatin immunoprecipitation. Epigenetic chromatin structure was altered using drug treatment with 5-aza-2'-deoxycytidine (AZA) and trichostatin A (TSA). RESULTS: The ERalpha promoter in the ERalpha-negative lines MDA-MB-231, MCF10A, and MCF7-5C show CpG island methylation, histone 3 lysine 9 deacetylation, and decreased chromatin accessibility compared with ERalpha-positive cell lines MCF7 and T47-D. Treatment with AZA/TSA increased chromatin accessibility at the ERalpha promoter and allowed TFAP2C to induce ERalpha expression in ERalpha-negative cells. Chromatin immunoprecipitation analysis showed that binding of TFAP2C to the ERalpha promoter is blocked in ERalpha-negative cells but that treatment with AZA/TSA enabled TFAP2C and polymerase II binding. CONCLUSION: We conclude that the activity of TFAP2C at specific target genes depends upon epigenetic chromatin structure. Furthermore, the combination of increasing chromatin accessibility and inducing TFAP2C provides a more robust activation of the ERalpha gene in ERalpha-negative breast cancer cells.


Asunto(s)
Cromatina/metabolismo , Receptor alfa de Estrógeno/genética , Regiones Promotoras Genéticas/genética , Factor de Transcripción AP-2/metabolismo , Acetilación/efectos de los fármacos , Azacitidina/análogos & derivados , Azacitidina/farmacología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Cromatina/efectos de los fármacos , Inmunoprecipitación de Cromatina , Islas de CpG/genética , Metilación de ADN/efectos de los fármacos , Decitabina , Inhibidores Enzimáticos/farmacología , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Histonas/metabolismo , Humanos , Ácidos Hidroxámicos/farmacología , Lisina/metabolismo , Unión Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción AP-2/genética
5.
Cancer Res ; 67(13): 6392-9, 2007 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-17616699

RESUMEN

Thiol antioxidants, including N-acetyl-L-cysteine (NAC), are widely used as modulators of the intracellular redox state. We investigated the hypothesis that NAC-induced reactive oxygen species (ROS) signaling perturbs cellular proliferation by regulating the cell cycle regulatory protein cyclin D1 and the ROS scavenging enzyme Mn-superoxide dismutase (MnSOD). When cultured in media containing NAC, mouse fibroblasts showed G(1) arrest with decreased cyclin D1 protein levels. The absence of a NAC-induced G(1) arrest in fibroblasts overexpressing cyclin D1 (or a nondegradable mutant of cyclin D1-T286A) indicates that cyclin D1 regulates this G(1) arrest. A delayed response to NAC exposure was an increase in both MnSOD protein and activity. NAC-induced G(1) arrest is exacerbated in MnSOD heterozygous fibroblasts. Results from electron spin resonance spectroscopy and flow cytometry measurements of dihydroethidine fluorescence showed an approximately 2-fold to 3-fold increase in the steady-state levels of superoxide (O(2)(*-)) in NAC-treated cells compared with control. Scavenging of O(2)(*-) with Tiron reversed the NAC-induced G(1) arrest. These results show that an O(2)(*-) signaling pathway regulates NAC-induced G(1) arrest by decreasing cyclin D1 protein levels and increasing MnSOD activity.


Asunto(s)
Acetilcisteína/farmacología , Ciclina D1/metabolismo , Superóxido Dismutasa/metabolismo , Superóxidos/metabolismo , Animales , Dicarbetoxidihidrocolidina/análogos & derivados , Dicarbetoxidihidrocolidina/farmacología , Espectroscopía de Resonancia por Spin del Electrón , Fibroblastos/metabolismo , Fase G1 , Humanos , Ratones , Células 3T3 NIH , Oxidación-Reducción , Especies Reactivas de Oxígeno , Transducción de Señal
6.
Free Radic Biol Med ; 45(11): 1573-80, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18845242

RESUMEN

Many breast cancer cells typically exhibit lower expression of manganese superoxide dismutase (MnSOD) compared to the normal cells from which they arise. This decrease can often be attributed to a defect in the transcription of SOD2, the gene encoding MnSOD; however, the mechanism responsible for this change remains unclear. Here, we describe how altered histone modifications and a repressive chromatin structure constitute an epigenetic process to down regulate SOD2 in human breast carcinoma cell lines. Utilizing chromatin immunoprecipitation (ChIP) we observed decreased levels of dimethyl H3K4 and acetylated H3K9 at key regulatory elements of the SOD2 gene. Consistent with these results, we show that loss of these histone modifications creates a repressive chromatin structure at SOD2. Transcription factor ChIP experiments revealed that this repressive chromatin structure influences the binding of SP-1, AP-1, and NFkappaB to SOD2 regulatory cis-elements in vivo. Lastly, we show that treatment with the histone deacetylase inhibitors trichostatin A and sodium butyrate can reactivate SOD2 expression in breast cancer cell lines. Taken together, these results indicate that epigenetic silencing of SOD2 could be facilitated by changes in histone modifications and represent one mechanism leading to the altered expression of MnSOD observed in many breast cancers.


Asunto(s)
Neoplasias de la Mama/genética , Silenciador del Gen , Histonas/metabolismo , Superóxido Dismutasa/genética , Acetilación , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Cromatina/metabolismo , Cromatina/ultraestructura , Inmunoprecipitación de Cromatina , Femenino , Código de Histonas , Inhibidores de Histona Desacetilasas , Histona Desacetilasas/metabolismo , Humanos , Ácidos Hidroxámicos/farmacología , Metilación , FN-kappa B/metabolismo , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Factor de Transcripción Sp1/metabolismo , Superóxido Dismutasa/metabolismo , Factor de Transcripción AP-1/metabolismo , Activación Transcripcional
7.
Cancer Res ; 66(3): 1605-10, 2006 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16452219

RESUMEN

It has been hypothesized that cancer cells increase glucose metabolism to protect against metabolic fluxes of hydroperoxides via glutathione-dependent peroxidases. 2-Deoxy-D-glucose, inhibits glucose metabolism and has been shown to cause cytotoxicity in cancer cells that is partially mediated by disruptions in thiol metabolism. In the current study, human breast cancer cells were continuously treated (24 hours) with 2-deoxy-D-glucose, and total glutathione content as well as the expression of the first enzyme in the glutathione synthetic pathway [glutamate cysteine ligase (GCL)] were found to be induced 2.0-fold. Inhibiting GCL activity during 2-deoxy-D-glucose exposure using l-buthionine-[S,R]-sulfoximine (BSO) significantly enhanced the cytotoxic effects of 2-deoxy-D-glucose and caused increases in endpoints indicative of oxidative stress, including % oxidized glutathione and steady-state levels of pro-oxidants as assayed using an oxidation-sensitive fluorescent probe. These results show that treatment of human breast cancer cells with 2-deoxy-d-glucose causes metabolic oxidative stress that is accompanied by increases in steady-state levels of GCL mRNA, GCL activity, and glutathione content. Furthermore, inhibition of 2-deoxy-D-glucose-mediated induction of GCL activity with BSO increases endpoints indicative of oxidative stress and sensitizes cancer cells to 2-deoxy-D-glucose-induced cytotoxicity. These results support the hypothesis that drug combinations capable of inhibiting both glucose and hydroperoxide metabolism may provide an effective biochemical strategy for sensitizing human cancer cells to metabolic oxidative stress.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/enzimología , Butionina Sulfoximina/farmacología , Desoxiglucosa/farmacología , Glutamato-Cisteína Ligasa/antagonistas & inhibidores , Antimetabolitos Antineoplásicos/farmacología , Neoplasias de la Mama/sangre , Línea Celular Tumoral , Desoxiglucosa/sangre , Sinergismo Farmacológico , Inhibidores Enzimáticos/farmacología , Glutamato-Cisteína Ligasa/genética , Glutamato-Cisteína Ligasa/metabolismo , Glutatión/metabolismo , Disulfuro de Glutatión/metabolismo , Humanos , Estrés Oxidativo , ARN Mensajero/genética , ARN Mensajero/metabolismo
8.
Free Radic Biol Med ; 43(7): 1023-36, 2007 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17761298

RESUMEN

The development of organisms requires concerted changes in gene activity. The free radical theory of development proposes that oxygen serves as a morphogen to educe development by influencing the production of metabolic oxidants such as free radicals and reactive oxygen species. One of the central tenets of this theory is that these metabolic oxidants influence development by altering the antioxidant capacity of cells by changing their production of glutathione (GSH). Here we extend on these principles by linking GSH production and oxygen sensing in the control of gene expression to establish the epigenotype of cells during development. We prescribe this novel role to GSH and oxygen during development because these metabolites influence the activity of enzymes responsible for initiating and perpetuating epigenetic control of gene expression. Increased GSH production influences epigenetic processes including DNA and histone methylation by limiting the availability of S-adenosylmethionine, the cofactor utilized during epigenetic control of gene expression by DNA and histone methyltransferases. Moreover, the recent discovery of histone demethylases that require oxygen as a cofactor directly links epigenetic processes to oxygen gradients during development.


Asunto(s)
Metilación de ADN , Epigénesis Genética , Radicales Libres , Estrés Oxidativo , Animales , Daño del ADN/genética , Humanos , Neoplasias/inducido químicamente , Neoplasias/genética , Neoplasias/metabolismo , Oxidación-Reducción
10.
Epigenetics ; 10(4): 259-73, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25774863

RESUMEN

Epigenetics, or regulation of gene expression independent of DNA sequence, is the missing link between genotype and phenotype. Epigenetic memory, mediated by histone and DNA modifications, is controlled by a set of specialized enzymes, metabolite availability, and signaling pathways. A mostly unstudied subject is how sub-toxic exposure to several xenobiotics during specific developmental stages can alter the epigenome and contribute to the development of disease phenotypes later in life. Furthermore, it has been shown that exposure to low-dose xenobiotics can also result in further epigenetic remodeling in the germ line and contribute to increase disease risk in the next generation (multigenerational and transgenerational effects). We here offer a perspective on current but still incomplete knowledge of xenobiotic-induced epigenetic alterations, and their possible transgenerational transmission. We also propose several molecular mechanisms by which the epigenetic landscape may be altered by environmental xenobiotics and hypothesize how diet and physical activity may counteract epigenetic alterations.


Asunto(s)
Ensamble y Desensamble de Cromatina , Discapacidades del Desarrollo/etiología , Exposición a Riesgos Ambientales , Xenobióticos/efectos adversos , Dieta , Exposición a Riesgos Ambientales/efectos adversos , Epigénesis Genética , Genotipo , Humanos , Fenotipo , Xenobióticos/metabolismo
11.
Antioxid Redox Signal ; 20(10): 1590-8, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23795822

RESUMEN

SIGNIFICANCE: Molecular oxygen is a Janus-faced electron acceptor for biological systems, serving as a reductant for respiration, or as the genesis for oxygen-derived free radicals that damage macromolecules. Superoxide is well known to perturb nonheme iron proteins, including Fe/S proteins such as aconitase and succinate dehydrogenase, as well as other enzymes containing labile iron such as the prolyl hydroxylase domain-containing family of enzymes; whereas hydrogen peroxide is more specific for two-electron reactions with thiols on glutathione, glutaredoxin, thioredoxin, and the peroxiredoxins. RECENT ADVANCES: Over the past two decades, familial cases of amyotrophic lateral sclerosis (ALS) have been shown to have an association with commonly altered superoxide dismutase 1 (SOD1) activity, expression, and protein structure. This has led to speculation that an altered redox balance may have a role in creating the ALS phenotype. CRITICAL ISSUES: While SOD1 alterations in familial ALS are manifold, they generally create perturbations in the flux of electrons. The nexus of SOD1 between one- and two-electron signaling processes places it at a key signaling regulatory checkpoint for governing cellular responses to physiological and environmental cues. FUTURE DIRECTIONS: The manner in which ALS-associated mutations adjust SOD1's role in controlling the flow of electrons between one- and two-electron signaling processes remains obscure. Here, we discuss the ways in which SOD1 mutations influence the form and function of copper zinc SOD, the consequences of these alterations on free radical biology, and how these alterations might influence cell signaling during the onset of ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/enzimología , Superóxido Dismutasa/metabolismo , Esclerosis Amiotrófica Lateral/genética , Animales , Humanos , Mutación Missense , Oxidación-Reducción , Estrés Oxidativo , Transducción de Señal , Superóxido Dismutasa/genética , Superóxido Dismutasa-1 , Superóxidos/metabolismo
12.
Antioxid Redox Signal ; 18(15): 1946-55, 2013 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-22946823

RESUMEN

SIGNIFICANCE: Manganese superoxide dismutase (SOD2), encoded by the nuclear gene SOD2, is a critical mitochondrial antioxidant enzyme whose activity has broad implications in health and disease. Thirty years ago, Oberley and Buettner elegantly folded SOD2 into cancer biology with the free radical theory of cancer, which was built on the observation that many human cancers had reduced SOD2 activity. In the original formulation, the loss of SOD2 in tumor cells produced a state of perpetual oxidative stress, which, in turn, drove genetic instability, leading to cancer development. RECENT ADVANCES: In the past two decades, research has established that SOD2 transcriptional activity is controlled, at least in part, via epigenetic mechanisms at different stages in the development of human cancer. These mechanisms, which include histone methylation, histone acetylation, and DNA methylation, are increasingly recognized as being aberrantly regulated in human cancer. Indeed, the epigenetic progenitor model proposed by Henikoff posits that epigenetic events are central governing agents of carcinogenesis. Important recent advances in epigenetics research have indicated that the loss of SOD activity itself may contribute to changes in epigenetic regulation, establishing a vicious cycle that drives further epigenetic instability. CRITICAL ISSUES: With these observations in mind, we propose an epigenetic revision to the free radical theory of cancer: that loss of SOD activity promotes epigenetic aberrancies, driving the epigenetic instability in tumor cells which produces broad phenotypic effects. FUTURE DIRECTIONS: The development of next-generation sequencing technologies and novel approaches in systems biology and bioinformatics promise to make testing this exciting model a reality in the near future.


Asunto(s)
Islas de CpG , Metilación de ADN , Histonas/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Humanos , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Superóxidos/metabolismo
13.
Free Radic Biol Med ; 53(11): 2178-87, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23022407

RESUMEN

Cancer arises from normal cells that acquire a series of molecular changes; however, the founding events that create the clonogens from which a tumor will arise and progress have been the subject of speculation. Through the efforts of several generations of cancer biologists it has been established that the malignant phenotype is an amalgamation of genetic and metabolic alterations. Numerous theories have suggested that either, or both, of these elements might serve as the impetus for cancer formation. Recently, the epigenetic origins of cancer have been suggested as an additional mechanism giving rise to the malignant phenotype. When the discovery that the enzymes responsible for initiating and perpetuating epigenetic events is linked to metabolism by their cofactors, a new paradigm for the origins of cancer can be created. Here, we summarize the foundation of such a paradigm on the origins of cancer, in which metabolic alterations create an epigenetic progenitor that clonally expands to become cancer. We suggest that metabolic alterations disrupt the production and availability of cofactors such as S-adenosylmethionine, α-ketoglutarate, NAD(+), and acetyl-CoA to modify the epigenotype of cells. We further speculate that redox biology can change epigenetic events through oxidation of enzymes and alterations in metabolic cofactors that affect epigenetic events such as DNA methylation. Combined, these metabolic and redox changes serve as the foundation for altering the epigenotype of normal cells and creating the epigenetic progenitor of cancer.


Asunto(s)
Epigénesis Genética , Neoplasias/metabolismo , Acetilación , Animales , Metabolismo de los Hidratos de Carbono , Epigenómica , Genoma Humano , Histonas/metabolismo , Humanos , Metilación , Neoplasias/genética , Oxidación-Reducción , Procesamiento Proteico-Postraduccional
14.
PLoS One ; 7(3): e33151, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22427974

RESUMEN

Leaf senescence is the orderly dismantling of older tissue that allows recycling of nutrients to developing portions of the plant and is accompanied by major changes in gene expression. Histone modifications correlate to levels of gene expression, and this study utilizes ChIP-seq to classify activating H3K4me3 and silencing H3K27me3 marks on a genome-wide scale for soil-grown mature and naturally senescent Arabidopsis leaves. ChIPnorm was used to normalize data sets and identify genomic regions with significant differences in the two histone methylation patterns, and the differences were correlated to changes in gene expression. Genes that showed an increase in the H3K4me3 mark in older leaves were senescence up-regulated, while genes that showed a decrease in the H3K4me3 mark in the older leaves were senescence down-regulated. For the H3K27me3 modification, genes that lost the H3K27me3 mark in older tissue were senescence up-regulated. Only a small number of genes gained the H3K27me3 mark, and these were senescence down-regulated. Approximately 50% of senescence up-regulated genes lacked the H3K4me3 mark in both mature and senescent leaf tissue. Two of these genes, SAG12 and At1g73220, display strong senescence up-regulation without the activating H3K4me3 histone modification. This study provides an initial epigenetic framework for the developmental transition into senescence.


Asunto(s)
Arabidopsis/crecimiento & desarrollo , Senescencia Celular/fisiología , Epigénesis Genética/fisiología , Regulación de la Expresión Génica de las Plantas/fisiología , Genoma de Planta/genética , Histonas/metabolismo , Hojas de la Planta/metabolismo , Arabidopsis/genética , Senescencia Celular/genética , Inmunoprecipitación de Cromatina , Metilación , Hojas de la Planta/citología , Hojas de la Planta/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
15.
Methods Mol Biol ; 767: 253-67, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21822881

RESUMEN

Chromatin immunoprecipitation (ChIP) is used to evaluate the interaction of proteins and genomic DNA. In eukaryotic cells, the DNA is highly compacted with the evolutionarily conserved histone proteins (which together with DNA form the nucleosome) and other chromosomal-associated proteins to form the chromatin structure. Chromatin structure is dynamically regulated by several mechanisms including transcription factor binding and various posttranslational modifications of the histone proteins. The chromatin structure can be affected by environmental factors, such as those that induce differentiation or promote self-renewal in stem cells. Using very specific antibodies, one can evaluate the specific amino acids within the histones and each one of these modifications is associated with a distinct DNA-templated process, including transcription. Therefore, determining the location of transcription factors and histone modifications can yield important insights into the DNA-associated activities that are occurring at that particular region of the genome at that time. ChIP followed by high-throughput DNA sequencing (ChIP-Seq) provides a means to rapidly determine the precise genomic location of transcription factor binding sites and histone modifications on a genome-wide scale. Genome-wide mapping of histone modifications and chromatin-associated proteins have already begun to reveal the mechanisms responsible for regulating the pattern of gene expression in mouse embryonic stem cells. However, similar studies in human embryonic stem cells are currently lacking due to the difficulty in obtaining the large number of purified cells typically required for ChIP and ChIP-Seq experiments. Here, we describe a detailed method for determining the locations of specific histone modifications using only one million cells.


Asunto(s)
Inmunoprecipitación de Cromatina/métodos , Epigénesis Genética , Genoma Humano/genética , Células Madre Pluripotentes/metabolismo , Análisis de Secuencia de ADN/métodos , Animales , Cromatina/aislamiento & purificación , Cromatina/metabolismo , ADN/aislamiento & purificación , ADN/metabolismo , Biblioteca de Genes , Histonas/metabolismo , Humanos , Lisina/metabolismo , Metilación , Ratones , Compuestos Orgánicos/metabolismo , Reacción en Cadena de la Polimerasa , Proteínas/metabolismo
16.
Free Radic Biol Med ; 47(2): 115-27, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19362589

RESUMEN

Cancer is a pathology that is associated with aberrant gene expression and an altered metabolism. Whereas changes in gene expression have historically been attributed to mutations, it has become apparent that epigenetic processes also play a critical role in controlling gene expression during carcinogenesis. Global changes in epigenetic processes, including DNA methylation and histone modifications, have been observed in cancer. These epigenetic alterations can aberrantly silence or activate gene expression during the formation of cancer; however, the process leading to this epigenetic switch in cancer remains unknown. Carcinogenesis is also associated with metabolic defects that increase mitochondrially derived reactive oxygen species, create an atypical redox state, and change the fundamental means by which cells produce energy. Here, we summarize the influence of these metabolic defects on epigenetic processes. Metabolic defects affect epigenetic enzymes by limiting the availability of cofactors like S-adenosylmethionine. Increased production of reactive oxygen species alters DNA methylation and histone modifications in tumor cells by oxidizing DNMTs and HMTs or through direct oxidation of nucleotide bases. Last, the Warburg effect and increased glutamine consumption in cancer influence histone acetylation and methylation by affecting the activity of sirtuins and histone demethylases.


Asunto(s)
Epigénesis Genética , Neoplasias/metabolismo , Acetilación , Animales , Metilación de ADN/fisiología , Radicales Libres/metabolismo , Glutatión/biosíntesis , Histonas/metabolismo , Humanos , Neoplasias/genética , Nucleosomas/metabolismo , Oxidación-Reducción , S-Adenosilmetionina/biosíntesis , S-Adenosilmetionina/genética
17.
J Oncol ; 2009: 780874, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20066163

RESUMEN

AP-2alpha and c-MYC are important transcription factors involved in multiple cellular processes. They each display the paradoxical capacities to stimulate both cell proliferation and apoptosis under different conditions. In the present study we found that over expression of c-MYC was associated with accumulation of reactive oxygen species (ROS) and apoptosis in human keratinocytes, both of which were significantly inhibited by co-expression of AP-2. The effects of AP-2 on c-MYC were active at several levels. First, AP-2 and c-MYC were confirmed to interact at the protein level as previously described. In addition, forced expression of AP-2 significantly decreased steady state levels of c-MYC mRNA and protein. These findings suggested that AP-2 may have a direct effect on the c-myc gene. Chromatin immunoprecipitation assays demonstrated that AP-2 proteins bound to a cluster of AP-2 binding sites located within a 2 kb upstream regulatory region of c-myc These results suggest that the negative regulation of AP-2 on c-MYC activity was achieved through binding of AP-2 protein to the c-myc gene. The effects of AP-2 on c-MYC induced ROS accumulation and apoptosis in epidermal keratinocytes are likely to play an important role in cell growth, differentiation and carcinogenesis of the skin.

18.
Nat Genet ; 40(11): 1341-7, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18836445

RESUMEN

Previously we have shown that nonsyndromic cleft lip with or without cleft palate (NSCL/P) is strongly associated with SNPs in IRF6 (interferon regulatory factor 6). Here, we use multispecies sequence comparisons to identify a common SNP (rs642961, G>A) in a newly identified IRF6 enhancer. The A allele is significantly overtransmitted (P = 1 x 10(-11)) in families with NSCL/P, in particular those with cleft lip but not cleft palate. Further, there is a dosage effect of the A allele, with a relative risk for cleft lip of 1.68 for the AG genotype and 2.40 for the AA genotype. EMSA and ChIP assays demonstrate that the risk allele disrupts the binding site of transcription factor AP-2alpha and expression analysis in the mouse localizes the enhancer activity to craniofacial and limb structures. Our findings place IRF6 and AP-2alpha in the same developmental pathway and identify a high-frequency variant in a regulatory element contributing substantially to a common, complex disorder.


Asunto(s)
Labio Leporino/genética , Elementos de Facilitación Genéticos/genética , Predisposición Genética a la Enfermedad , Factores Reguladores del Interferón/genética , Factor de Transcripción AP-2/metabolismo , Alelos , Animales , Secuencia de Bases , Sitios de Unión , Estudios de Casos y Controles , Secuencia Conservada , Haplotipos , Humanos , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Oportunidad Relativa , Fenotipo , Polimorfismo de Nucleótido Simple/genética , Unión Proteica
19.
Exp Mol Pathol ; 83(2): 277-82, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17651731

RESUMEN

Amyloid precursor protein (APP) has been implicated in squamous cell carcinoma. In this study we show that forced expression of the transcription factor activating protein 2alpha (AP-2alpha) results in significantly increased steady state levels of APP mRNA in human keratinocytes. Sequence analysis of the 5' end of the human APP gene revealed five putative binding sites for AP-2, suggesting that APP is a direct target for transactivation by AP-2. AP-2 protein bound at least 3 of these putative promoter elements in vitro as determined by electrophoretic mobility shift assay. Chromatin immunoprecipitation (ChIP) analysis showed that these binding sites were occupied by AP-2 in cells, thus indicating the relevance to AP-2 binding in vivo. We then analyzed APP and AP-2 mRNA and protein expression in squamous cell carcinoma tumor samples. Analysis of RNA extracted from human tissue showed a significant positive correlation between AP-2alpha and APP mRNA expression. Immunohistochemical staining of tumor samples also demonstrated a positive correlation which was substantiated through western blot studies. Taken together, these findings demonstrate a role for the transcription factor AP-2alpha in the regulation of APP gene expression in human keratinocytes.


Asunto(s)
Precursor de Proteína beta-Amiloide/genética , Carcinoma de Células Escamosas/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Boca/genética , Receptores de Superficie Celular/genética , Factor de Transcripción AP-2/genética , Factor de Transcripción AP-2/fisiología , Transcripción Genética , Carcinoma de Células Escamosas/patología , Línea Celular , Cromatina/genética , Cartilla de ADN , Humanos , Queratinocitos/fisiología , Neoplasias de la Boca/patología , Nexinas de Proteasas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
20.
Epigenetics ; 1(4): 163-71, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17965603

RESUMEN

Malignant breast cancer cells often exhibit lower expression and activity of manganese superoxide dismutase (MnSOD) than their normal cell counterparts; however, the mechanism(s) responsible for this change remains unclear. We examined whether SOD2, the gene encoding MnSOD, was epigenetically repressed in breast cancer cell lines by DNA methylation and histone acetylation. RT-PCR analysis of SOD2 mRNA showed the nontumorigenic breast epithelial cell line MCF-10A to have two to three fold higher expression levels than either UACC-893 or MDA-MB-435 breast carcinoma cells. Analysis of a region in the SOD2 promoter by sodium bisulfite genomic sequencing demonstrated significantly higher levels of CpG methylation in both human breast carcinoma cell lines assessed than in MCF-10A cells. CREB binding in vitro to a cognate site derived from this region was repressed by DNA methylation, and CREB binding to the 5' regulatory region of the SOD2 gene in vivo as determined by ChIP was significantly lower in breast carcinoma cells than in MCF-10A. Increased cytosine methylation was also accompanied by a significant decrease in the level of acetylated histones in the same region of the SOD2 promoter. Finally, a causal link between cytosine methylation and transcriptional repression was established by increasing MnSOD mRNA, protein and activity in breast carcinoma cells using the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine. These findings indicate that epigenetic silencing of SOD2 constitutes one mechanism leading to the decreased expression of MnSOD observed in many breast cancers.


Asunto(s)
Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Epigénesis Genética/genética , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Superóxido Dismutasa/genética , Línea Celular Tumoral , Cartilla de ADN , Femenino , Gliceraldehído-3-Fosfato Deshidrogenasas/genética , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA