Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Toxicol Appl Pharmacol ; 481: 116753, 2023 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-37951547

RESUMEN

Exposure to nickel, an environmental respiratory toxicant, is associated with lung diseases including asthma, pulmonary fibrosis, bronchitis and cancers. Our previous studies have shown that a majority of the nickel-induced transcriptional changes are persistent and do not reverse even after the termination of exposure. This suggested transcriptional memory, wherein the cell 'remembers' past nickel exposure. Transcriptional memory, due to which the cells respond more robustly to a previously encountered stimulus has been identified in a number of organisms. Therefore, transcriptional memory has been described as an adaptive mechanism. However, transcriptional memory caused by environmental toxicant exposures has not been well investigated. Moreover, how the transcriptional memory caused by an environmental toxicant might influence the outcome of exposure to a second toxicant has not been explored. In this study, we investigated whether nickel-induced transcriptional memory influences the outcome of the cell's response to a second respiratory toxicant, nicotine. Nicotine, an addictive compound in tobacco, is associated with the development of chronic lung diseases including chronic obstructive pulmonary disease (COPD) and pulmonary fibrosis. Our results show that nicotine exposure upregulated a subset of genes only in the cells previously exposed to nickel. Furthermore, our analyses indicate robust activation of interferon (IFN) signaling in these cells. IFN signaling is a driver of inflammation, which is associated with many chronic lung diseases. Therefore, our results suggest that nicotine exposure of lung cells that retain the transcriptional memory of previous nickel exposure could result in increased susceptibility to developing chronic inflammatory lung diseases.


Asunto(s)
Níquel , Fibrosis Pulmonar , Humanos , Níquel/toxicidad , Nicotina/toxicidad , Fibrosis Pulmonar/patología , Pulmón/patología , Células Epiteliales , Interferones
2.
Cell ; 132(5): 887-98, 2008 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-18329373

RESUMEN

The positioning of nucleosomes with respect to DNA plays an important role in regulating transcription. However, nucleosome mapping has been performed for only limited genomic regions in humans. We have generated genome-wide maps of nucleosome positions in both resting and activated human CD4+ T cells by direct sequencing of nucleosome ends using the Solexa high-throughput sequencing technique. We find that nucleosome phasing relative to the transcription start sites is directly correlated to RNA polymerase II (Pol II) binding. Furthermore, the first nucleosome downstream of a start site exhibits differential positioning in active and silent genes. TCR signaling induces extensive nucleosome reorganization in promoters and enhancers to allow transcriptional activation or repression. Our results suggest that H2A.Z-containing and modified nucleosomes are preferentially lost from the -1 nucleosome position. Our data provide a comprehensive view of the nucleosome landscape and its dynamic regulation in the human genome.


Asunto(s)
Genoma Humano , Nucleosomas/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Cromosomas , Elementos de Facilitación Genéticos , Histonas/metabolismo , Humanos , Activación de Linfocitos , Regiones Promotoras Genéticas , ARN Polimerasa II/metabolismo , Análisis de Secuencia de ADN/métodos , Sitio de Iniciación de la Transcripción
3.
Semin Cancer Biol ; 76: 99-109, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34058338

RESUMEN

Nickel compounds are environmental toxicants, prevalent in the atmosphere due to their widespread use in several industrial processes, extensive consumption of nickel containing products, as well as burning of fossil fuels. Exposure to nickel is associated with a multitude of chronic inflammatory lung diseases including asthma, chronic obstructive pulmonary disease (COPD) and pulmonary fibrosis. In addition, nickel exposure is implicated in the development of nasal and lung cancers. Interestingly, a common pathogenic mechanism underlying the development of diseases associated with nickel exposure is epithelial-mesenchymal transition (EMT). EMT is a process by which the epithelial cells lose their junctions and polarity and acquire mesenchymal traits, including increased ability to migrate and invade. EMT is a normal and essential physiological process involved in differentiation, development and wound healing. However, EMT also contributes to a number of pathological conditions, including fibrosis, cancer and metastasis. Growing evidence suggest that EMT induction could be an important outcome of nickel exposure. In this review, we discuss the role of EMT in nickel-induced lung diseases and the mechanisms associated with EMT induction by nickel exposure.


Asunto(s)
Transición Epitelial-Mesenquimal/efectos de los fármacos , Enfermedades Pulmonares/inducido químicamente , Enfermedades Pulmonares/patología , Níquel/efectos adversos , Animales , Humanos
4.
Mol Carcinog ; 61(1): 99-110, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34727382

RESUMEN

E-cadherin plays a central role in the stability of epithelial tissues by facilitating cell-cell adhesion. Loss of E-cadherin expression is a hallmark of epithelial-mesenchymal transition (EMT), a major event in the pathogenesis of several lung diseases. Our earlier studies showed that nickel, a ubiquitous environmental toxicant, induced EMT by persistently downregulating E-cadherin expression in human lung epithelial cells and that the EMT remained irreversible postexposure. However, the molecular basis of persistent E-cadherin downregulation by nickel exposure is not understood. Here, our studies show that the binding of transcription factor Sp1 to the promoter of E-cadherin encoding gene, CDH1, is essential for its expression. Nickel exposure caused a loss of Sp1 binding at the CDH1 promoter, resulting in its downregulation and EMT induction. Loss of Sp1 binding at the CDH1 promoter was associated with an increase in the binding of ZEB1 adjacent to the Sp1 binding site. ZEB1, an EMT master regulator persistently upregulated by nickel exposure, is a negative regulator of CDH1. CRISPR-Cas9-mediated knockout of ZEB1 restored Sp1 binding at the CDH1 promoter. Furthermore, ZEB1 knockout rescued E-cadherin expression and re-established the epithelial phenotype. Since EMT is associated with a number of nickel-exposure-associated chronic inflammatory lung diseases including asthma, fibrosis and cancer and metastasis, our findings provide new insights into the mechanisms associated with nickel pathogenesis.


Asunto(s)
Antígenos CD/genética , Cadherinas/genética , Pulmón/citología , Níquel/toxicidad , Factor de Transcripción Sp1/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética , Línea Celular , Regulación hacia Abajo , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal , Técnicas de Inactivación de Genes , Humanos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Células MCF-7 , Regiones Promotoras Genéticas
5.
Toxicol Appl Pharmacol ; 457: 116317, 2022 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-36400264

RESUMEN

Nickel (Ni), a heavy metal is prevalent in the atmosphere due to both natural and anthropogenic activities. Ni is a carcinogen implicated in the development of lung and nasal cancers in humans. Furthermore, Ni exposure is associated with a number of chronic lung diseases in humans including asthma, chronic bronchitis, emphysema, pulmonary fibrosis, pulmonary edema and chronic obstructive pulmonary disease (COPD). While Ni compounds are weak mutagens, a number of studies have demonstrated the potential of Ni to alter the epigenome, suggesting epigenomic dysregulation as an important underlying cause for its pathogenicity. In the eukaryotic nucleus, the DNA is organized in a three-dimensional (3D) space through assembly of higher order chromatin structures. Such an organization is critically important for transcription and other biological activities. Accumulating evidence suggests that by negatively affecting various cellular regulatory processes, Ni could potentially affect chromatin organization. In this review, we discuss the role of Ni in altering the chromatin architecture, which potentially plays a major role in Ni pathogenicity.

6.
Toxicol Appl Pharmacol ; 457: 116314, 2022 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-36368423

RESUMEN

Reactive oxygen species (ROS) are by-products of metabolism of oxygen and they play an important role in normal homeostasis and cell signaling, as well as in the initiation of diseases including cancer when their production is upregulated. Thus, it is imperative to understand the cellular and molecular basis by which ROS impact on various biological and pathological processes. In this report, we show that human keratinocyte cell line (HaCaT) treated with hydrogen peroxide displayed an increased activity of AhR, leading to enhanced expression of its downstream targets including cytochrome P450 genes. Intriguingly, preincubation of the complete culture medium with hydrogen peroxide accelerated AhR activation and its downstream signaling. Subsequent mass spectrometric analysis reveals that the oxidant elicits the production of oxindole, a tryptophan catabolic product. We further demonstrate that 2-oxindole (a major form of oxindole) is capable of activating AhR, strongly suggesting that ROS may exert a significant impact on AhR signaling. Consistent with this, we also observe that hexavalent chromium [Cr(VI)], a heavy metal known to generate ROS in vivo, enhances AhR protein levels, as well as stimulates expression of CYP1A2 in an AhR-dependent manner. Significantly, we show that hydrogen peroxide and 2-oxindole induce expression of IDO1 and PD-L1, two immune checkpoint proteins. Given the role of IDO1 and PD-L1 in mediating T cell activity and/or differentiation, we postulate that ROS in the tumor microenvironment may play a crucial role in immune suppression via perturbing AhR signaling.

7.
Mol Cell ; 53(4): 672-81, 2014 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-24486021

RESUMEN

Eukaryotic chromosomes are partitioned into topologically associating domains (TADs) that are demarcated by distinct insulator-binding proteins (IBPs) in Drosophila. Whether IBPs regulate specific long-range contacts and how this may impact gene expression remains unclear. Here we identify "indirect peaks" of multiple IBPs that represent their distant sites of interactions through long-range contacts. Indirect peaks depend on protein-protein interactions among multiple IBPs and their common cofactors, including CP190, as confirmed by high-resolution analyses of long-range contacts. Mutant IBPs unable to interact with CP190 impair long-range contacts as well as the expression of hundreds of distant genes that are specifically flanked by indirect peaks. Regulation of distant genes strongly correlates with RNAPII pausing, highlighting how this key transcriptional stage may trap insulator-based long-range interactions. Our data illustrate how indirect peaks may decipher gene regulatory networks through specific long-range interactions.


Asunto(s)
Inmunoprecipitación de Cromatina/métodos , Regulación de la Expresión Génica , Elementos Aisladores/fisiología , ARN Polimerasa II/metabolismo , Animales , Sitios de Unión , Factor de Unión a CCCTC , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Proteínas del Ojo/metabolismo , Redes Reguladoras de Genes , Mutación , Regiones Promotoras Genéticas , Unión Proteica , Mapeo de Interacción de Proteínas , Interferencia de ARN , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo
8.
Toxicol Appl Pharmacol ; 373: 1-9, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-30998937

RESUMEN

Cadmium (Cd) is a known human lung carcinogen. In addition, Cd exposure is associated with several lung diseases including emphysema, chronic obstructive pulmonary disease (COPD), asthma and fibrosis. Although earlier studies have identified several processes dysregulated by Cd exposure, the underlying mechanisms remain unclear. Here, we examined the transcriptome of lung epithelial cells exposed to Cd to understand the molecular basis of Cd-induced diseases. Computational analysis of the transcriptome predicted a significant number of Cd-upregulated genes to be targets of miR-30 family miRNAs. Experimental validation showed downregulation of all the miR-30 family members in Cd exposed cells. We found SNAIL, an EMT master regulator, to be the most upregulated among the miR-30 targets. Furthermore, we found decrease in the levels of epithelial marker E- cadherin (CDH1) and increase in the levels of mesenchymal markers, ZEB1 and vimentin. This suggested induction of EMT in Cd exposed cells. Luciferase reporter assays showed that miR-30 repressed SNAIL by directly targeting its 3' UTR. Over expression of miR-30e and transfection of miR-30e mimics reduced Cd-induced SNAIL upregulation. Our results suggest that miR-30 negatively regulates SNAIL in lung epithelial cells and that Cd-induced downregulation of miR-30 relieves this repression, resulting in SNAIL upregulation and EMT induction. EMT plays a major role in many diseases associated with Cd exposure including fibrosis, COPD, and cancer and metastasis. Therefore, our identification of miR-30 downregulation in Cd exposed cells and the consequent activation of SNAIL provides important mechanistic insights into lung diseases associated with Cd exposure.


Asunto(s)
Cloruro de Cadmio/toxicidad , Células Epiteliales/efectos de los fármacos , Pulmón/efectos de los fármacos , MicroARNs/metabolismo , Factores de Transcripción de la Familia Snail/metabolismo , Regiones no Traducidas 3' , Antígenos CD/genética , Antígenos CD/metabolismo , Sitios de Unión , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular , Regulación hacia Abajo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Humanos , Pulmón/metabolismo , Pulmón/patología , MicroARNs/genética , Transducción de Señal , Factores de Transcripción de la Familia Snail/genética , Transcriptoma , Regulación hacia Arriba , Vimentina/genética , Vimentina/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo
9.
EMBO J ; 33(14): 1599-613, 2014 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-24916307

RESUMEN

Chromosomal domains in Drosophila are marked by the insulator-binding proteins (IBPs) dCTCF/Beaf32 and cofactors that participate in regulating long-range interactions. Chromosomal borders are further enriched in specific histone modifications, yet the role of histone modifiers and nucleosome dynamics in this context remains largely unknown. Here, we show that IBP depletion impairs nucleosome dynamics specifically at the promoters and coding sequence of genes flanked by IBP binding sites. Biochemical purification identifies the H3K36 histone methyltransferase NSD/dMes-4 as a novel IBP cofactor, which specifically co-regulates the chromatin accessibility of hundreds of genes flanked by dCTCF/Beaf32. NSD/dMes-4 presets chromatin before the recruitment of transcriptional activators including DREF that triggers Set2/Hypb-dependent H3K36 trimethylation, nucleosome positioning, and RNA splicing. Our results unveil a model for how IBPs regulate nucleosome dynamics and gene expression through NSD/dMes-4, which may regulate H3K27me3 spreading. Our data uncover how IBPs dynamically regulate chromatin organization depending on distinct cofactors.


Asunto(s)
Cromatina/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas del Ojo/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Elementos Aisladores/genética , Modelos Biológicos , Nucleosomas/fisiología , Animales , Western Blotting , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Proteínas del Ojo/genética , N-Metiltransferasa de Histona-Lisina/genética , Histonas/metabolismo , Análisis por Micromatrices , Datos de Secuencia Molecular , Análisis de Componente Principal , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ARN , Técnicas del Sistema de Dos Híbridos
10.
Mol Carcinog ; 57(6): 794-806, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29528143

RESUMEN

Nickel (Ni) is an environmental and occupational carcinogen, and exposure to Ni is associated with lung and nasal cancers in humans. Furthermore, Ni exposure is implicated in several lung diseases including chronic inflammatory airway diseases, asthma, and fibrosis. However, the mutagenic potential of Ni is low and does not correlate with its potent toxicity and carcinogenicity. Therefore, mechanisms underlying Ni exposure-associated diseases remain poorly understood. Since the health risks of environmental exposures often continue post exposure, understanding the exposure effects that persist after the termination of exposure could provide mechanistic insights into diseases. By examining the persistent effects of Ni exposure, we report that Ni induces epithelial-mesenchymal transition (EMT) and that the mesenchymal phenotype remains irreversible even after the termination of exposure. Ni-induced EMT was dependent on the irreversible upregulation of ZEB1, an EMT master regulator, via resolution of its promoter bivalency. ZEB1, upon activation, downregulated its repressors as well as the cell-cell adhesion molecule, E-cadherin, resulting in the cells undergoing EMT and switching to persistent mesenchymal status. ZEB1 depletion in cells exposed to Ni attenuated Ni-induced EMT. Moreover, Ni exposure did not induce EMT in ZEB1-depleted cells. Activation of EMT, during which the epithelial cells lose cell-cell adhesion and become migratory and invasive, plays a major role in asthma, fibrosis, and cancer and metastasis, lung diseases associated with Ni exposure. Therefore, our finding of irreversible epigenetic activation of ZEB1 by Ni exposure and the acquisition of persistent mesenchymal phenotype would have important implications in understanding Ni-induced diseases.


Asunto(s)
Epigénesis Genética/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Níquel/farmacología , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética , Hipoxia de la Célula , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal/genética , Perfilación de la Expresión Génica , Humanos , Fenotipo , Interferencia de ARN , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo
11.
Proc Natl Acad Sci U S A ; 111(40): 14631-6, 2014 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-25246589

RESUMEN

Investigations into the genomic landscape of histone modifications in heterochromatic regions have revealed histone H3 lysine 9 dimethylation (H3K9me2) to be important for differentiation and maintaining cell identity. H3K9me2 is associated with gene silencing and is organized into large repressive domains that exist in close proximity to active genes, indicating the importance of maintenance of proper domain structure. Here we show that nickel, a nonmutagenic environmental carcinogen, disrupted H3K9me2 domains, resulting in the spreading of H3K9me2 into active regions, which was associated with gene silencing. We found weak CCCTC-binding factor (CTCF)-binding sites and reduced CTCF binding at the Ni-disrupted H3K9me2 domain boundaries, suggesting a loss of CTCF-mediated insulation function as a potential reason for domain disruption and spreading. We furthermore show that euchromatin islands, local regions of active chromatin within large H3K9me2 domains, can protect genes from H3K9me2-spreading-associated gene silencing. These results have major implications in understanding H3K9me2 dynamics and the consequences of chromatin domain disruption during pathogenesis.


Asunto(s)
Cromatina/metabolismo , Epigénesis Genética/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Níquel/farmacología , Acetilación , Secuencia de Aminoácidos , Sitios de Unión/genética , Western Blotting , Bronquios/citología , Factor de Unión a CCCTC , Línea Celular , Cromatina/genética , Epigénesis Genética/genética , Células Epiteliales/metabolismo , Perfilación de la Expresión Génica , Genoma Humano/genética , Histonas/metabolismo , Humanos , Lisina/metabolismo , Metilación , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , Interferencia de ARN , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
12.
J Cell Physiol ; 231(7): 1611-20, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26588041

RESUMEN

Oxygen levels range from 2% to 9% in vivo. Atmospheric O2 levels (21%) are known to induce cell proliferation defects and cellular senescence in primary cell cultures. However, the mechanistic basis of the deleterious effects of higher O2 levels is not fully understood. On the other hand, immortalized cells including cancer cell lines, which evade cellular senescence are normally cultured at 21% O2 and the effects of higher O2 on these cells are understudied. Here, we addressed this problem by culturing immortalized human bronchial epithelial (BEAS-2B) cells at ambient atmospheric, 21% O2 and lower, 10% O2. Our results show increased inflammatory response at 21% O2 but not at 10% O2. We found higher RelA binding at the NF-κB1/RelA target gene promoters as well as upregulation of several pro-inflammatory cytokines in cells cultured at 21% O2. RelA knockdown prevented the upregulation of the pro-inflammatory cytokines at 21% O2, suggesting NF-κB1/RelA as a major mediator of inflammatory response in cells cultured at 21% O2. Interestingly, unlike the 21% O2 cultured cells, exposure of 10% O2 cultured cells to H2O2 did not elicit inflammatory response, suggesting increased ability to tolerate oxidative stress in cells cultured at lower O2 levels.


Asunto(s)
Inflamación/metabolismo , Pulmón/metabolismo , Oxígeno/metabolismo , Factor de Transcripción ReIA/genética , Proliferación Celular , Senescencia Celular , Citocinas/genética , Citocinas/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Técnicas de Silenciamiento del Gen , Humanos , Inflamación/patología , Pulmón/patología , Regiones Promotoras Genéticas , Factor de Transcripción ReIA/metabolismo
13.
Nucleic Acids Res ; 40(8): 3364-77, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22210859

RESUMEN

The TET family of FE(II) and 2-oxoglutarate-dependent enzymes (Tet1/2/3) promote DNA demethylation by converting 5-methylcytosine to 5-hydroxymethylcytosine (5hmC), which they further oxidize into 5-formylcytosine and 5-carboxylcytosine. Tet1 is robustly expressed in mouse embryonic stem cells (mESCs) and has been implicated in mESC maintenance. Here we demonstrate that, unlike genetic deletion, RNAi-mediated depletion of Tet1 in mESCs led to a significant reduction in 5hmC and loss of mESC identity. The differentiation phenotype due to Tet1 depletion positively correlated with the extent of 5hmC loss. Meta-analyses of genomic data sets suggested interaction between Tet1 and leukemia inhibitory factor (LIF) signaling. LIF signaling is known to promote self-renewal and pluripotency in mESCs partly by opposing MAPK/ERK-mediated differentiation. Withdrawal of LIF leads to differentiation of mESCs. We discovered that Tet1 depletion impaired LIF-dependent Stat3-mediated gene activation by affecting Stat3's ability to bind to its target sites on chromatin. Nanog overexpression or inhibition of MAPK/ERK signaling, both known to maintain mESCs in the absence of LIF, rescued Tet1 depletion, further supporting the dependence of LIF/Stat3 signaling on Tet1. These data support the conclusion that analysis of mESCs in the hours/days immediately following efficient Tet1 depletion reveals Tet1's normal physiological role in maintaining the pluripotent state that may be subject to homeostatic compensation in genetic models.


Asunto(s)
Citosina/análogos & derivados , Proteínas de Unión al ADN/fisiología , Células Madre Embrionarias/enzimología , Factor Inhibidor de Leucemia/metabolismo , Proteínas Proto-Oncogénicas/fisiología , Factor de Transcripción STAT3/metabolismo , 5-Metilcitosina/análogos & derivados , Animales , Células Cultivadas , Citosina/metabolismo , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Proteína Homeótica Nanog , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Interferencia de ARN , Transducción de Señal , ADN Metiltransferasa 3B
14.
Toxicol Sci ; 196(2): 170-186, 2023 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-37707797

RESUMEN

The aryl hydrocarbon receptor (AhR) is an inducible transcription factor whose ligands include the potent environmental contaminant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Ligand-activated AhR binds to DNA at dioxin response elements (DREs) containing the core motif 5'-GCGTG-3'. However, AhR binding is highly tissue specific. Most DREs in accessible chromatin are not bound by TCDD-activated AhR, and DREs accessible in multiple tissues can be bound in some and unbound in others. As such, AhR functions similarly to many nuclear receptors. Given that AhR possesses a strong core motif, it is suited for a motif-centered analysis of its binding. We developed interpretable machine learning models predicting the AhR binding status of DREs in MCF-7, GM17212, and HepG2 cells, as well as primary human hepatocytes. Cross-tissue models predicting transcription factor (TF)-DNA binding generally perform poorly. However, reasons for the low performance remain unexplored. By interpreting the results of individual within-tissue models and by examining the features leading to low cross-tissue performance, we identified sequence and chromatin context patterns correlated with AhR binding. We conclude that AhR binding is driven by a complex interplay of tissue-agnostic DRE flanking DNA sequence and tissue-specific local chromatin context. Additionally, we demonstrate that interpretable machine learning models can provide novel and experimentally testable mechanistic insights into DNA binding by inducible TFs.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Aprendizaje Automático , Receptores de Hidrocarburo de Aril , Humanos , Genoma Humano , Especificidad de Órganos , Receptores de Hidrocarburo de Aril/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo
15.
Genome Res ; 19(10): 1742-51, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19713549

RESUMEN

Chromatin modifications have been implicated in the regulation of gene expression. While association of certain modifications with expressed or silent genes has been established, it remains unclear how changes in chromatin environment relate to changes in gene expression. In this article, we used ChIP-seq (chromatin immunoprecipitation with massively parallel sequencing) to analyze the genome-wide changes in chromatin modifications during activation of total human CD4(+) T cells by T-cell receptor (TCR) signaling. Surprisingly, we found that the chromatin modification patterns at many induced and silenced genes are relatively stable during the short-term activation of resting T cells. Active chromatin modifications were already in place for a majority of inducible protein-coding genes, even while the genes were silent in resting cells. Similarly, genes that were silenced upon T-cell activation retained positive chromatin modifications even after being silenced. To investigate if these observations are also valid for miRNA-coding genes, we systematically identified promoters for known miRNA genes using epigenetic marks and profiled their expression patterns using deep sequencing. We found that chromatin modifications can poise miRNA-coding genes as well. Our data suggest that miRNA- and protein-coding genes share similar mechanisms of regulation by chromatin modifications, which poise inducible genes for activation in response to environmental stimuli.


Asunto(s)
Cromatina/fisiología , Expresión Génica , MicroARNs/genética , Proteínas/genética , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/fisiología , Proliferación Celular , Células Cultivadas , Cromatina/genética , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina/fisiología , Humanos , Activación de Linfocitos/genética , Sistemas de Lectura Abierta/genética , Regiones Promotoras Genéticas/genética
16.
Cancers (Basel) ; 14(23)2022 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-36497298

RESUMEN

Chronic environmental exposure to toxic metal(loid)s significantly contributes to human cancer development and progression. It is estimated that approximately 90% of cancer deaths are a result of metastasis of malignant cells, which is initiated by epithelial-mesenchymal transition (EMT) during early carcinogenesis. EMT is regulated by many families of genes and microRNAs (miRNAs) that control signaling pathways for cell survival, death, and/or differentiation. Recent mechanistic studies have shown that toxic metal(loid)s alter the expression of miRNAs responsible for regulating the expression of genes involved in EMT. Altered miRNA expressions have the potential to be biomarkers for predicting survival and responses to treatment in cancers. Significantly, miRNAs can be developed as therapeutic targets for cancer patients in the clinic. In this mini review, we summarize key findings from recent studies that highlight chemical-miRNA-gene interactions leading to the perturbation of EMT after exposure to toxic metal(loid)s including arsenic, cadmium, nickel, and chromium.

17.
Nucleic Acids Res ; 36(16): 5221-31, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18684996

RESUMEN

ChIP-Seq, which combines chromatin immunoprecipitation (ChIP) with ultra high-throughput massively parallel sequencing, is increasingly being used for mapping protein-DNA interactions in-vivo on a genome scale. Typically, short sequence reads from ChIP-Seq are mapped to a reference genome for further analysis. Although genomic regions enriched with mapped reads could be inferred as approximate binding regions, short read lengths (approximately 25-50 nt) pose challenges for determining the exact binding sites within these regions. Here, we present SISSRs (Site Identification from Short Sequence Reads), a novel algorithm for precise identification of binding sites from short reads generated from ChIP-Seq experiments. The sensitivity and specificity of SISSRs are demonstrated by applying it on ChIP-Seq data for three widely studied and well-characterized human transcription factors: CTCF (CCCTC-binding factor), NRSF (neuron-restrictive silencer factor) and STAT1 (signal transducer and activator of transcription protein 1). We identified 26 814, 5813 and 73 956 binding sites for CTCF, NRSF and STAT1 proteins, respectively, which is 32, 299 and 78% more than that inferred previously for the respective proteins. Motif analysis revealed that an overwhelming majority of the identified binding sites contained the previously established consensus binding sequence for the respective proteins, thus attesting for SISSRs' accuracy. SISSRs' sensitivity and precision facilitated further analyses of ChIP-Seq data revealing interesting insights, which we believe will serve as guidance for designing ChIP-Seq experiments to map in vivo protein-DNA interactions. We also show that tag densities at the binding sites are a good indicator of protein-DNA binding affinity, which could be used to distinguish and characterize strong and weak binding sites. Using tag density as an indicator of DNA-binding affinity, we have identified core residues within the NRSF and CTCF binding sites that are critical for a stronger DNA binding.


Asunto(s)
Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/metabolismo , Genómica/métodos , Elementos Reguladores de la Transcripción , Factores de Transcripción/metabolismo , Algoritmos , Secuencia de Bases , Sitios de Unión , Factor de Unión a CCCTC , Células HeLa , Humanos , Células Jurkat , Datos de Secuencia Molecular , Proteínas Represoras/metabolismo , Factor de Transcripción STAT1/metabolismo
18.
Mutat Res Rev Mutat Res ; 780: 61-68, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31395350

RESUMEN

CCCTC-binding factor (CTCF) is a highly conserved, ubiquitously expressed zinc finger protein. CTCF is a multifunctional protein, associated with a number of vital cellular processes such as transcriptional activation, repression, insulation, imprinting and genome organization. Emerging evidence indicates that CTCF is also involved in DNA damage response. In this review, we focus on the newly identified role of CTCF in facilitating DNA double-strand break repair. Due to the large number of cellular processes in which CTCF is involved, factors that functionally affect CTCF could have serious implications on genomic stability. It is becoming increasingly clear that exposure to environmental toxicants could have adverse effects on CTCF functions. Here we discuss the various ways that environmental toxicants could impact CTCF functions and the potential consequences on DNA damage response.


Asunto(s)
Factor de Unión a CCCTC/genética , Daño del ADN/genética , Reparación del ADN/genética , Animales , Roturas del ADN de Doble Cadena , Genoma/genética , Inestabilidad Genómica/genética , Humanos
19.
Epigenetics Chromatin ; 12(1): 75, 2019 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-31856895

RESUMEN

BACKGROUND: Nickel is an occupational and environmental toxicant associated with a number of diseases in humans including pulmonary fibrosis, bronchitis and lung and nasal cancers. Our earlier studies showed that the nickel-exposure-induced genome-wide transcriptional changes, which persist even after the termination of exposure may underlie nickel pathogenesis. However, the mechanisms that drive nickel-induced persistent changes to the transcriptome remain elusive. RESULTS: To elucidate the mechanisms that underlie nickel-induced long-term transcriptional changes, in this study, we examined the transcriptome and the epigenome of human lung epithelial cells during nickel exposure and after the termination of exposure. We identified two categories of persistently differentially expressed genes: (i) the genes that were differentially expressed during nickel exposure; and (ii) the genes that were differentially expressed only after the termination of exposure. Interestingly, > 85% of the nickel-induced gene expression changes occurred only after the termination of exposure. We also found extensive genome-wide alterations to the activating histone modification, H3K4me3, after the termination of nickel exposure, which coincided with the post-exposure gene expression changes. In addition, we found significant post-exposure alterations to the repressive histone modification, H3K27me3. CONCLUSION: Our results suggest that while modest first wave of transcriptional changes occurred during nickel exposure, extensive transcriptional changes occurred during a second wave of transcription for which removal of nickel ions was essential. By uncovering a new category of transcriptional and epigenetic changes, which occur only after the termination of exposure, this study provides a novel understanding of the long-term deleterious consequences of nickel exposure on human health.


Asunto(s)
Epigénesis Genética/efectos de los fármacos , Níquel/toxicidad , Línea Celular , Metilación de ADN/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Histonas/metabolismo , Humanos , Regulación hacia Arriba/efectos de los fármacos
20.
FEBS Lett ; 582(2): 391-7, 2008 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-18177740

RESUMEN

The interferon inducible transmembrane (IFITM) proteins mediate several cellular processes such as homotypic cell adhesion functions of interferons (IFNs) and cellular anti-proliferative activities. We show that the BAF complex-mediated induction of IFITM3 is dependent on binding of the transcriptional enhancer factor 1 (TEF-1/TEAD1) to the M-CAT like elements of its promoter. TEF-1 knock-down reduced the BAF complex-mediated activation of IFITM3 promoter. In the absence of the BAF complex, TEF-1 is repressive to IFITM3 expression. The regulation of IFITM3 by TEF-1 demonstrates that TEF-1 dependent regulation is more widespread than its previously established role in the expression of muscle specific genes.


Asunto(s)
ADN Helicasas/fisiología , Proteínas de Unión al ADN/fisiología , Regulación de la Expresión Génica/fisiología , Proteínas de la Membrana/genética , Proteínas Nucleares/fisiología , Proteínas de Unión al ARN/genética , Factores de Transcripción/fisiología , Secuencia de Aminoácidos , Secuencia de Bases , Línea Celular , Ensayo de Cambio de Movilidad Electroforética , Humanos , Proteínas de la Membrana/química , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Interferencia de ARN , Proteínas de Unión al ARN/química , Homología de Secuencia de Ácido Nucleico , Factores de Transcripción de Dominio TEA
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA