Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Mol Cell ; 69(1): 36-47.e7, 2018 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-29249653

RESUMEN

Recent integrative epigenome analyses highlight the importance of functionally distinct chromatin states for accurate cell function. How these states are established and maintained is a matter of intense investigation. Here, we present evidence for DNA damage as an unexpected means to shape a protective chromatin environment at regions of recurrent replication stress (RS). Upon aberrant fork stalling, DNA damage signaling and concomitant H2AX phosphorylation coordinate the FACT-dependent deposition of macroH2A1.2, a histone variant that promotes DNA repair by homologous recombination (HR). MacroH2A1.2, in turn, facilitates the accumulation of the tumor suppressor and HR effector BRCA1 at replication forks to protect from RS-induced DNA damage. Consequently, replicating primary cells steadily accrue macroH2A1.2 at fragile regions, whereas macroH2A1.2 loss in these cells triggers DNA damage signaling-dependent senescence, a hallmark of RS. Altogether, our findings demonstrate that recurrent DNA damage contributes to the chromatin landscape to ensure the epigenomic integrity of dividing cells.


Asunto(s)
Carcinogénesis/genética , Cromatina/genética , Daño del ADN/genética , Reparación del ADN/genética , Replicación del ADN/genética , Histonas/genética , Recombinación Homóloga/genética , Proteína BRCA1/metabolismo , División Celular/genética , Células Cultivadas , Senescencia Celular/genética , Inestabilidad Genómica/fisiología , Humanos , Transducción de Señal/genética
2.
Proc Natl Acad Sci U S A ; 120(6): e2212072120, 2023 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-36724254

RESUMEN

Cancer treatments targeting DNA repair deficiencies often encounter drug resistance, possibly due to alternative metabolic pathways that counteract the most damaging effects. To identify such alternative pathways, we screened for metabolic pathways exhibiting synthetic lethality with inhibition of the DNA damage response kinase Ataxia-telangiectasia-mutated (ATM) using a metabolism-centered Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9 library. Our data revealed Kelch-like ECH-associated protein 1 (KEAP1) as a key factor involved in desensitizing cancer cells to ATM inhibition both in vitro and in vivo. Cells depleted of KEAP1 exhibited an aberrant overexpression of the cystine transporter SLC7A11, robustly accumulated cystine inducing disulfide stress, and became hypersensitive to ATM inhibition. These hallmarks were reversed in a reducing cellular environment indicating that disulfide stress was a crucial factor. In The Cancer Genome Atlas (TCGA) pan-cancer datasets, we found that ATM levels negatively correlated with KEAP1 levels across multiple solid malignancies. Together, our results unveil ATM and KEAP1 as new targetable vulnerabilities in solid tumors.


Asunto(s)
Ataxia Telangiectasia , Neoplasias Pulmonares , Humanos , Proteína 1 Asociada A ECH Tipo Kelch/genética , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Cistina/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Neoplasias Pulmonares/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo
3.
Mol Carcinog ; 63(6): 1024-1037, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38411275

RESUMEN

Homologous recombination (HR) and poly ADP-ribosylation are partially redundant pathways for the repair of DNA damage in normal and cancer cells. In cell lines that are deficient in HR, inhibition of poly (ADP-ribose) polymerase (poly (ADP-ribose) polymerase [PARP]1/2) is a proven target with several PARP inhibitors (PARPis) currently in clinical use. Resistance to PARPi often develops, usually involving genetic alterations in DNA repair signaling cascades, but also metabolic rewiring particularly in HR-proficient cells. We surmised that alterations in metabolic pathways by cancer drugs such as Olaparib might be involved in the development of resistance to drug therapy. To test this hypothesis, we conducted a metabolism-focused clustered regularly interspaced short palindromic repeats knockout screen to identify genes that undergo alterations during the treatment of tumor cells with PARPis. Of about 3000 genes in the screen, our data revealed that mitochondrial pyruvate carrier 1 (MPC1) is an essential factor in desensitizing nonsmall cell lung cancer (NSCLC) lung cancer lines to PARP inhibition. In contrast to NSCLC lung cancer cells, triple-negative breast cancer cells do not exhibit such desensitization following MPC1 loss and reprogram the tricarboxylic acid cycle and oxidative phosphorylation pathways to overcome PARPi treatment. Our findings unveil a previously unknown synergistic response between MPC1 loss and PARP inhibition in lung cancer cells.


Asunto(s)
Resistencia a Antineoplásicos , Neoplasias Pulmonares , Transportadores de Ácidos Monocarboxílicos , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Humanos , Línea Celular Tumoral , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Sistemas CRISPR-Cas , Resistencia a Antineoplásicos/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Proteínas de Transporte de Membrana Mitocondrial/genética , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Ftalazinas/farmacología , Piperazinas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología
4.
Proc Natl Acad Sci U S A ; 116(15): 7471-7476, 2019 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-30910969

RESUMEN

Phosphorylation of histone H2AX is a major contributor to efficient DNA repair. We recently reported neurobehavioral deficits in mice lacking H2AX. Here we establish that this neural failure stems from impairment of mitochondrial function and repression of the mitochondrial biogenesis gene PGC-1α. H2AX loss leads to reduced levels of the major subunits of the mitochondrial respiratory complexes in mouse embryonic fibroblasts and in the striatum, a brain region particularly vulnerable to mitochondrial damage. These defects are substantiated by disruption of the mitochondrial shape in H2AX mutant cells. Ectopic expression of PGC-1α restores mitochondrial oxidative phosphorylation complexes and mitigates cell death. H2AX knockout mice display increased neuronal death in the brain when challenged with 3-nitropronionic acid, which targets mitochondria. This study establishes a role for H2AX in mitochondrial homeostasis associated with neuroprotection.


Asunto(s)
Histonas/metabolismo , Mitocondrias/metabolismo , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Fosforilación Oxidativa , Animales , Muerte Celular , Transporte de Electrón/fisiología , Histonas/genética , Ratones , Ratones Noqueados , Mitocondrias/genética , Células-Madre Neurales/citología , Neuronas/citología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Fosforilación
5.
Proc Natl Acad Sci U S A ; 112(7): 2121-6, 2015 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-25646414

RESUMEN

Ataxia telangiectasia (A-T), a rare autosomal recessive disorder characterized by progressive cerebellar degeneration and a greatly increased incidence of cancer among other symptoms, is caused by a defective or missing ataxia telangiectasia mutated (ATM) gene. The ATM protein has roles in DNA repair and in the regulation of reactive oxygen species (ROS). Here, we provide, to our knowledge, the first evidence that NADPH oxidase 4 (NOX4) is involved in manifesting A-T disease. We showed that NOX4 expression levels are higher in A-T cells, and that ATM inhibition leads to increased NOX4 expression in normal cells. A-T cells exhibit elevated levels of oxidative DNA damage, DNA double-strand breaks and replicative senescence, all of which are partially abrogated by down-regulation of NOX4 with siRNA. Sections of degenerating cerebelli from A-T patients revealed elevated NOX4 levels. ATM-null mice exhibit A-T disease but they die from cancer before the neurological symptoms are manifested. Injecting Atm-null mice with fulvene-5, a specific inhibitor of NOX4 and NADPH oxidase 2 (NOX2), decreased their elevated cancer incidence to that of the controls. We conclude that, in A-T disease in humans and mice, NOX4 may be critical mediator and targeting it will open up new avenues for therapeutic intervention in neurodegeneration.


Asunto(s)
Ataxia Telangiectasia/enzimología , NADPH Oxidasas/metabolismo , Adulto , Animales , Ataxia Telangiectasia/patología , Daño del ADN , Replicación del ADN , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , NADPH Oxidasa 4 , Adulto Joven
6.
Biochim Biophys Acta ; 1819(7): 743-56, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22430255

RESUMEN

Chromatin is a dynamic complex of DNA and proteins that regulates the flow of information from genome to end product. The efficient recognition and faithful repair of DNA damage, particularly double-strand damage, is essential for genomic stability and cellular homeostasis. Imperfect repair of DNA double-strand breaks (DSBs) can lead to oncogenesis. The efficient repair of DSBs relies in part on the rapid formation of foci of phosphorylated histone H2AX (γ-H2AX) at each break site, and the subsequent recruitment of repair factors. These foci can be visualized with appropriate antibodies, enabling low levels of DSB damage to be measured in samples obtained from patients. Such measurements are proving useful to optimize treatments involving ionizing radiation, to assay in vivo the efficiency of various drugs to induce DNA damage, and to help diagnose patients with a variety of syndromes involving elevated levels of γ-H2AX. We will survey the state of the art of utilizing γ-H2AX in clinical settings. We will also discuss possibilities with other histone post-translational modifications. The ability to measure in vivo the responses of individual patients to particular drugs and/or radiation may help optimize treatments and improve patient care. This article is part of a Special Issue entitled: Chromatin in time and space.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Histonas/metabolismo , Neoplasias/metabolismo , Procesamiento Proteico-Postraduccional , Secuencia de Aminoácidos , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/química , Biomarcadores de Tumor/fisiología , Cromatina/metabolismo , Roturas del ADN de Doble Cadena , Relación Dosis-Respuesta en la Radiación , Histonas/química , Histonas/fisiología , Humanos , Datos de Secuencia Molecular , Neoplasias/diagnóstico , Neoplasias/terapia , Estructura Terciaria de Proteína
7.
Mutat Res ; 751(2): 77-81, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22580379

RESUMEN

The human genome is continuously exposed to such potentially deleterious agents as the highly reactive molecules known as reactive oxygen species (ROS). ROS include superoxide anions (O(2)(-)) and hydrogen peroxide (H(2)O(2)). Over the last decade, the ROS-generating NADPH oxidases (NOXs) have been recognized as one of the main sources of ROS production in numerous human cell types. In addition to regulating normal physiological redox-dependent processes, the NOXs are involved in cellular oxidative stress. In contrast to the other NOXs, the NADPH oxidase NOX4 exists in the immediate environment of the nucleus. There is accumulating evidence for the involvement of NOX4-derived ROS in genomic instability as well as in cancer and other inflammation-related diseases. We recently showed that NOX4 plays a critical role in oncogenic Ras-induced DNA damage. Here we reflect upon the growing awareness of NOX4, review its role in inducing genomic instability, and call attention to its possible role in nuclear redox-sensitive mechanisms underlying DNA-damage signaling and repair.


Asunto(s)
Daño del ADN , NADPH Oxidasas/fisiología , Especies Reactivas de Oxígeno/metabolismo , Células Cultivadas , Inestabilidad Genómica , NADPH Oxidasa 4 , Estrés Oxidativo
8.
Sci Rep ; 12(1): 3758, 2022 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-35260660

RESUMEN

Genomic stability is essential for organismal development, cellular homeostasis, and survival. The DNA double-strand breaks are particularly deleterious, creating an environment prone to cellular transformation and oncogenic activation. The histone variant H2AX is an essential component of the nucleosome responsible for initiating the early steps of the DNA repair process. H2AX maintains genomic stability by initiating a signaling cascade that collectively functions to promote DNA double-strand breaks repair. Recent advances have linked genomic stability to energetic metabolism, and alterations in metabolism were found to interfere with genome maintenance. Utilizing genome-wide transcripts profiling to identify differentially-expressed genes involved in energetic metabolism, we compared control and H2AX-deficient metastatic breast cancer cell lines, and found that H2AX loss leads to the repression of key genes regulating glycolysis, with a prominent effect on hexokinase-2 (HK2). These observations are substantiated by evidence that H2AX loss compromises glycolysis, effect which was reversed by ectopic expression of HK2. Utilizing models of experimental metastasis, we found that H2AX silencing halts progression of metastatic breast cancer cells MDA-MB-231. Most interestingly, ectopic expression of HK2 in H2AX-deficient cells restores their metastatic potential. Using multiple publicly available datasets, we found a significantly strong positive correlation between H2AX expression levels in patients with invasive breast cancer, and levels of glycolysis genes, particularly HK2. These observations are consistent with the evidence that high H2AX expression is associated with shorter distant metastasis-free survival. Our findings reveal a role for histone H2AX in controlling the metastatic ability of breast cancer cells via maintenance of HK2-driven glycolysis.


Asunto(s)
Neoplasias de la Mama , Hexoquinasa , Histonas , Neoplasias de la Mama/genética , Línea Celular Tumoral , Proliferación Celular/genética , ADN/metabolismo , Femenino , Inestabilidad Genómica , Glucólisis/genética , Hexoquinasa/genética , Histonas/metabolismo , Humanos
9.
Int Rev Cell Mol Biol ; 364: 241-265, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34507785

RESUMEN

Genomic instability and metabolic reprogramming are among the key hallmarks discriminating cancer cells from normal cells. The two phenomena contribute to the robust and evasive nature of cancer, particularly when cancer cells are exposed to chemotherapeutic agents. Genomic instability is defined as the increased frequency of mutations within the genome, while metabolic reprogramming is the alteration of metabolic pathways that cancer cells undergo to adapt to increased bioenergetic demand. An underlying source of these mutations is the aggregate product of damage to the DNA, and a defective repair pathway, both resulting in the expansion of genomic lesions prior to uncontrolled proliferation and survival of cancer cells. Exploitation of DNA damage and the subsequent DNA damage response (DDR) have aided in defining therapeutic approaches in cancer. Studies have demonstrated that targeting metabolic reprograming yields increased sensitivity to chemo- and radiotherapies. In the past decade, it has been shown that these two key features are interrelated. Metabolism impacts DNA damage and DDR via regulation of metabolite pools. Conversely, DDR affects the response of metabolic pathways to therapeutic agents. Because of the interplay between genomic instability and metabolic reprogramming, we have compiled findings which more selectively highlight the dialog between metabolism and DDR, with a particular focus on glucose metabolism and double-strand break (DSB) repair pathways. Decoding this dialog will provide significant clues for developing combination cancer therapies.


Asunto(s)
Inestabilidad Genómica , Neoplasias/genética , Neoplasias/metabolismo , Animales , Ensamble y Desensamble de Cromatina/genética , Daño del ADN/genética , Reparación del ADN/genética , Humanos , Metaboloma
10.
Mol Cell Oncol ; 7(5): 1771959, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32944631

RESUMEN

While genomic instability and mitochondrial homeostasis are integral for cancer progression, how these two hallmarks interact remains poorly understood. Here, we reflect on the dialogue between chromatin-based genomic instability and impairment of mitochondrial function and depict the importance of this interaction in cancer progression to metastasis.

11.
Nat Commun ; 9(1): 1526, 2018 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-29670103

RESUMEN

ATM drives DNA repair by phosphorylating the histone variant H2AX. While ATM mutations elicit prominent neurobehavioral phenotypes, neural roles for H2AX have been elusive. We report impaired motor learning and balance in H2AX-deficient mice. Mitigation of reactive oxygen species (ROS) with N-acetylcysteine (NAC) reverses the behavioral deficits. Mouse embryonic fibroblasts deficient for H2AX exhibit increased ROS production and failure to activate the antioxidant response pathway controlled by the transcription factor NRF2. The NRF2 targets GCLC and NQO1 are depleted in the striatum of H2AX knockouts, one of the regions most vulnerable to ROS-mediated damage. These findings establish a role for ROS in the behavioral deficits of H2AX knockout mice and reveal a physiologic function of H2AX in mediating influences of oxidative stress on NRF2-transcriptional targets and behavior.


Asunto(s)
Conducta Animal , Histonas/deficiencia , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Acetilcisteína/química , Animales , Antioxidantes/química , Cuerpo Estriado/metabolismo , Daño del ADN , Fibroblastos/metabolismo , Células HEK293 , Heterocigoto , Histonas/fisiología , Humanos , Ratones , Ratones Noqueados , Microscopía Confocal , Modelos Neurológicos , Destreza Motora , Oxidación-Reducción , Fenotipo , Fosforilación , Especies Reactivas de Oxígeno/metabolismo
12.
Sci Rep ; 7(1): 12734, 2017 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-28986581

RESUMEN

Current approaches have limitations in providing insight into the functional properties of particular nucleosomes in their native molecular environment. Here we describe a simple and powerful method involving elution of histones using intercalators or salt, to assess stability features dependent on DNA superhelicity and relying mainly on electrostatic interactions, respectively, and measurement of the fraction of histones remaining chromatin-bound in the individual nuclei using histone type- or posttranslational modification- (PTM-) specific antibodies and automated, quantitative imaging. The method has been validated in H3K4me3 ChIP-seq experiments, by the quantitative assessment of chromatin loop relaxation required for nucleosomal destabilization, and by comparative analyses of the intercalator and salt induced release from the nucleosomes of different histones. The accuracy of the assay allowed us to observe examples of strict association between nucleosome stability and PTMs across cell types, differentiation state and throughout the cell-cycle in close to native chromatin context, and resolve ambiguities regarding the destabilizing effect of H2A.X phosphorylation. The advantages of the in situ measuring scenario are demonstrated via the marked effect of DNA nicking on histone eviction that underscores the powerful potential of topological relaxation in the epigenetic regulation of DNA accessibility.


Asunto(s)
Imagenología Tridimensional , Nucleosomas/metabolismo , Animales , Automatización , Línea Celular Tumoral , Doxorrubicina/farmacología , Etidio/metabolismo , Humanos , Ratones , Nucleosomas/efectos de los fármacos , Sales (Química)/farmacología
14.
Oncotarget ; 7(20): 29023-35, 2016 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-27296552

RESUMEN

LOX regulates cancer progression in a variety of human malignancies. It is overexpressed in aggressive cancers and higher expression of LOX is associated with higher cancer mortality. Here, we report a new function of LOX in mitosis. We show that LOX co-localizes to mitotic spindles from metaphase to telophase, and p-H3(Ser10)-positive cells harbor strong LOX staining. Further, purification of mitotic spindles from synchronized cells show that LOX fails to bind to microtubules in the presence of nocodazole, whereas paclitaxel treated samples showed enrichment in LOX expression, suggesting that LOX binds to stabilized microtubules. LOX knockdown leads to G2/M phase arrest; reduced p-H3(Ser10), cyclin B1, CDK1, and Aurora B. Moreover, LOX knockdown significantly increased sensitivity of cancer cells to chemotherapeutic agents that target microtubules. Our findings suggest that LOX has a role in cancer cell mitosis and may be targeted to enhance the activity of microtubule inhibitors for cancer therapy.


Asunto(s)
Mitosis/fisiología , Neoplasias/patología , Proteína-Lisina 6-Oxidasa/metabolismo , Huso Acromático/enzimología , Línea Celular Tumoral , Humanos , Neoplasias/enzimología
15.
Cancer Med ; 5(8): 1731-41, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27339860

RESUMEN

The development of novel cancer treatments is a challenging task, partly because results from model systems often fail to predict drug efficacy in humans, and also tumors are often inaccessible for biochemical analysis, preventing effective monitoring of drug activity in vivo. Utilizing a model system, we evaluated the use of drug-induced DNA damage in surrogate tissues as indicators of drug efficacy. Samples of a commercially available melanoma skin model (Mattek MLNM-FT-A375) containing keratinocyte and fibroblast layers with melanoma nodules were subjected to various chemotherapeutic regimens for one, four, or eight days. At these times they were analyzed for DNA double-stranded breaks (γH2AX foci) and apoptosis (TUNEL). A wide range of drug responses in both tumor and normal tissues were observed and cataloged. For the melanoma, the most common drug response was apoptosis. The basal keratinocyte layer, which was the most reliable indicator of drug response in the melanoma skin model, responded with γH2AX foci formation that was abrupt and transient. The relationships between tumor and surrogate tissue drug responses are complex, indicating that while surrogate tissue drug responses may be useful clinical tools, careful control of variables such as the timing of sampling may be important in interpreting the results.


Asunto(s)
Antineoplásicos/farmacología , Melanoma/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Daño del ADN , ADN de Neoplasias/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Humanos , Queratinocitos/efectos de los fármacos , Melanoma/genética , Melanoma/patología , Mutágenos/farmacología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología
16.
Cell Cycle ; 15(18): 2398-404, 2016 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-27315462

RESUMEN

The epithelial-mesenchymal transition (EMT) is thought to be essential for cancer metastasis. While chromatin remodeling is involved in EMT, which processes contribute to this remodeling remain poorly investigated. Recently, we showed that silencing or removal of the histone variant H2A.X induced mesenchymal-like characteristics, including activation of the EMT transcription factors, Slug and Zeb1 in human colon cancer cells. Here, we provide the evidence that H2A.X loss in human non-tumorigenic breast cell line MCF10A results in a robust EMT activation, as substantiated by a genome-wide expression analysis. Cells deficient for H2A.X exhibit enhanced migration and invasion, along with an activation of a set of mesenchymal genes and a concomitant repression of epithelial genes. In the breast model, the EMT-related transcription factor Twist1 cooperates with Slug to regulate EMT upon H2A.X Loss. Of interest, H2A.X expression level tightly correlates with Twist1, and to a lesser extent with Slug in the panel of human breast cancer cell lines of the NCI-60 datasets. These new findings indicate that H2A.X is involved in the EMT processes in cells of different origins but pairing with transcription factors for EMT may be tissue specific.


Asunto(s)
Mama/patología , Transición Epitelial-Mesenquimal , Histonas/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción de la Familia Snail/metabolismo , Proteína 1 Relacionada con Twist/metabolismo , Mama/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/genética , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Modelos Biológicos
17.
Nat Commun ; 7: 10711, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26876487

RESUMEN

The epithelial-mesenchymal transition (EMT), considered essential for metastatic cancer, has been a focus of much research, but important questions remain. Here, we show that silencing or removing H2A.X, a histone H2A variant involved in cellular DNA repair and robust growth, induces mesenchymal-like characteristics including activation of EMT transcription factors, Slug and ZEB1, in HCT116 human colon cancer cells. Ectopic H2A.X re-expression partially reverses these changes, as does silencing Slug and ZEB1. In an experimental metastasis model, the HCT116 parental and H2A.X-null cells exhibit a similar metastatic behaviour, but the cells with re-expressed H2A.X are substantially more metastatic. We surmise that H2A.X re-expression leads to partial EMT reversal and increases robustness in the HCT116 cells, permitting them to both form tumours and to metastasize. In a human adenocarcinoma panel, H2A.X levels correlate inversely with Slug and ZEB1 levels. Together, these results point to H2A.X as a regulator of EMT.


Asunto(s)
Adenocarcinoma/genética , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Histonas/genética , Proteínas de Homeodominio/genética , Metástasis de la Neoplasia/genética , Factores de Transcripción/genética , Animales , Western Blotting , Sistemas CRISPR-Cas , Línea Celular Tumoral , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Variación Genética , Células HCT116 , Células HEK293 , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Transcripción de la Familia Snail , Homeobox 1 de Unión a la E-Box con Dedos de Zinc
18.
Radiat Res ; 183(3): 262-70, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25706776

RESUMEN

Human exposure to ionizing radiation from medical procedures has increased sharply in the last three decades. Recent epidemiological studies suggest a direct relationship between exposure to ionizing radiation and health problems, including cancer incidence. Therefore, minimizing the impact of radiation exposure in patients has become a priority in the development of future clinical practices. Crucial players in radiation-induced DNA damage include reactive oxygen species (ROS), but the sources of these have remained elusive. To the best of our knowledge, we show here for the first time that two members of the ROS-generating NADPH oxidase family (NOXs), NOX4 and NOX5, are involved in radiation-induced DNA damage. Depleting these two NOXs in human primary fibroblasts resulted in reduced levels of DNA damage as measured by levels of radiation-induced foci, a marker of DNA double-strand breaks (DSBs) and the comet assay coupled with increased cell survival. NOX involvement was substantiated with fulvene-5, a NOXs-specific inhibitor. Moreover, fulvene-5 mitigated radiation-induced DNA damage in human peripheral blood mononuclear cells ex vivo. Our results provide evidence that the inactivation of NOXs protects cells from radiation-induced DNA damage and cell death. These findings suggest that NOXs inhibition may be considered as a future pharmacological target to help minimize the negative effects of radiation exposure for millions of patients each year.


Asunto(s)
Ciclopentanos/administración & dosificación , Daño del ADN/genética , Proteínas de la Membrana/genética , NADPH Oxidasas/genética , Supervivencia Celular/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de la radiación , Daño del ADN/efectos de la radiación , Fibroblastos/efectos de los fármacos , Fibroblastos/efectos de la radiación , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , NADPH Oxidasa 4 , NADPH Oxidasa 5 , NADPH Oxidasas/antagonistas & inhibidores , Cultivo Primario de Células , Radiación Ionizante , Especies Reactivas de Oxígeno/metabolismo
19.
Anticancer Agents Med Chem ; 13(3): 502-14, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22931418

RESUMEN

Reactive oxygen species (ROS) form a class of molecules with both positive and negative impacts on cellular health. Negatively, ROS may react with cellular constituents including proteins, lipids, and DNA to generate an array of oxidative lesions. These lesions may compromise genome stability which is critical for long-term cellular homeostasis and healthy progeny. Paradoxically, ROS also function as strong signalling molecules that mediate various growth-related responses, so their presence is also essential for cellular metabolism. While ROS are generated in an unregulated manner by physical stresses such as exposure to ionizing radiation and biochemical malfunctions such as mitochondrial leakage, cells also contain the NADPH oxidases NOXs and DUOXs, which specifically generate ROS in a wide variety of tissues. While the NOXs/DUOXs may be involved in maintaining optimal cellular redox levels, there is also accumulating evidence that NADPH oxidases-derived ROS may elevate the risk for genomic instability and cancer. Cancer cells may produce high levels of ROS, and in some cases, the source of these ROS has been linked to NOX/DUOX deregulation as reported for prostate cancer (NOX1 and NOX5), melanoma and glioblastoma (NOX4) among others. In addition, recent studies reveal that targeting NADPH oxidases with NOXs inhibitors may impair tumor growth in vivo; indicating that these proteins may be useful targets in future clinical strategies to fight cancer. This review provides an overview of the current knowledge concerning these enzymes, their roles in cancer, and their potential as targets in future cancer therapies.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , NADPH Oxidasas/genética , Neoplasias/tratamiento farmacológico , Antineoplásicos/síntesis química , Transformación Celular Neoplásica , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Inhibidores Enzimáticos/síntesis química , Inestabilidad Genómica , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/clasificación , Isoenzimas/genética , Isoenzimas/metabolismo , Terapia Molecular Dirigida , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/clasificación , NADPH Oxidasas/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Oxidación-Reducción/efectos de los fármacos , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA