Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Rev Mol Cell Biol ; 22(2): 75-95, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33328614

RESUMEN

Cellular senescence, first described in vitro in 1961, has become a focus for biotech companies that target it to ameliorate a variety of human conditions. Eminently characterized by a permanent proliferation arrest, cellular senescence occurs in response to endogenous and exogenous stresses, including telomere dysfunction, oncogene activation and persistent DNA damage. Cellular senescence can also be a controlled programme occurring in diverse biological processes, including embryonic development. Senescent cell extrinsic activities, broadly related to the activation of a senescence-associated secretory phenotype, amplify the impact of cell-intrinsic proliferative arrest and contribute to impaired tissue regeneration, chronic age-associated diseases and organismal ageing. This Review discusses the mechanisms and modulators of cellular senescence establishment and induction of a senescence-associated secretory phenotype, and provides an overview of cellular senescence as an emerging opportunity to intervene through senolytic and senomorphic therapies in ageing and ageing-associated diseases.


Asunto(s)
Envejecimiento , Senescencia Celular , Telómero , Investigación Biomédica Traslacional , Animales , Proliferación Celular , Daño del ADN , Humanos , Fenotipo
3.
Trends Genet ; 37(4): 337-354, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33020022

RESUMEN

Subcellular compartmentalization contributes to the organization of a plethora of molecular events occurring within cells. This can be achieved in membraneless organelles generated through liquid-liquid phase separation (LLPS), a demixing process that separates and concentrates cellular reactions. RNA is often a critical factor in mediating LLPS. Recent evidence indicates that DNA damage response foci are membraneless structures formed via LLPS and modulated by noncoding transcripts synthesized at DNA damage sites. Neurodegeneration is often associated with DNA damage, and dysfunctional LLPS events can lead to the formation of toxic aggregates. In this review, we discuss those gene products involved in neurodegeneration that undergo LLPS and their involvement in the DNA damage response.


Asunto(s)
Daño del ADN/genética , Degeneración Nerviosa/genética , Orgánulos/genética , Transcripción Genética , Humanos , Extracción Líquido-Líquido , Degeneración Nerviosa/patología , Orgánulos/química , Transición de Fase
4.
Nat Mater ; 22(5): 644-655, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36581770

RESUMEN

The process in which locally confined epithelial malignancies progressively evolve into invasive cancers is often promoted by unjamming, a phase transition from a solid-like to a liquid-like state, which occurs in various tissues. Whether this tissue-level mechanical transition impacts phenotypes during carcinoma progression remains unclear. Here we report that the large fluctuations in cell density that accompany unjamming result in repeated mechanical deformations of cells and nuclei. This triggers a cellular mechano-protective mechanism involving an increase in nuclear size and rigidity, heterochromatin redistribution and remodelling of the perinuclear actin architecture into actin rings. The chronic strains and stresses associated with unjamming together with the reduction of Lamin B1 levels eventually result in DNA damage and nuclear envelope ruptures, with the release of cytosolic DNA that activates a cGAS-STING (cyclic GMP-AMP synthase-signalling adaptor stimulator of interferon genes)-dependent cytosolic DNA response gene program. This mechanically driven transcriptional rewiring ultimately alters the cell state, with the emergence of malignant traits, including epithelial-to-mesenchymal plasticity phenotypes and chemoresistance in invasive breast carcinoma.


Asunto(s)
Actinas , Neoplasias , ADN , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Citosol/metabolismo , Transducción de Señal
5.
EMBO Rep ; 23(2): e53658, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-34854526

RESUMEN

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes the coronavirus disease 2019 (COVID-19), known to be more common in the elderly, who also show more severe symptoms and are at higher risk of hospitalization and death. Here, we show that the expression of the angiotensin converting enzyme 2 (ACE2), the SARS-CoV-2 cell receptor, increases during aging in mouse and human lungs. ACE2 expression increases upon telomere shortening or dysfunction in both cultured mammalian cells and in vivo in mice. This increase is controlled at the transcriptional level, and Ace2 promoter activity is DNA damage response (DDR)-dependent. Both pharmacological global DDR inhibition of ATM kinase activity and selective telomeric DDR inhibition by the use of antisense oligonucleotides prevent Ace2 upregulation following telomere damage in cultured cells and in mice. We propose that during aging telomere dysfunction due to telomeric shortening or damage triggers DDR activation and this causes the upregulation of ACE2, the SARS-CoV-2 cell receptor, thus contributing to make the elderly more susceptible to the infection.


Asunto(s)
Envejecimiento , Enzima Convertidora de Angiotensina 2/genética , COVID-19 , Daño del ADN , Telómero , Anciano , Envejecimiento/genética , Animales , Humanos , Ratones , SARS-CoV-2 , Telómero/genética
6.
J Cell Sci ; 134(6)2021 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-33558311

RESUMEN

The DNA damage response (DDR) is the signaling cascade that recognizes DNA double-strand breaks (DSBs) and promotes their resolution via the DNA repair pathways of non-homologous end joining (NHEJ) or homologous recombination (HR). We and others have shown that DDR activation requires DROSHA; however, whether DROSHA exerts its functions by associating with damage sites, what controls its recruitment, and how DROSHA influences DNA repair remains poorly understood. Here, we show that DROSHA associates with DSBs independently of transcription. Neither H2AX, nor ATM or DNA-PK kinase activities are required for recruitment of DROSHA to break sites. Rather, DROSHA interacts with RAD50, and inhibition of the MRN complex by mirin treatment abolishes this interaction. MRN complex inactivation by RAD50 knockdown or mirin treatment prevents DROSHA recruitment to DSBs and, as a consequence, also prevents 53BP1 (also known as TP53BP1) recruitment. During DNA repair, DROSHA inactivation reduces NHEJ and boosts HR frequency. Indeed, DROSHA knockdown also increases the association of downstream HR factors such as RAD51 to DNA ends. Overall, our results demonstrate that DROSHA is recruited at DSBs by the MRN complex and directs DNA repair towards NHEJ.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , Daño del ADN/genética , Reparación del ADN/genética , Recombinación Homóloga
7.
Cell ; 133(6): 1006-18, 2008 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-18555777

RESUMEN

Cells enter senescence, a state of stable proliferative arrest, in response to a variety of cellular stresses, including telomere erosion, DNA damage, and oncogenic signaling, which acts as a barrier against malignant transformation in vivo. To identify genes controlling senescence, we conducted an unbiased screen for small hairpin RNAs that extend the life span of primary human fibroblasts. Here, we report that knocking down the chemokine receptor CXCR2 (IL8RB) alleviates both replicative and oncogene-induced senescence (OIS) and diminishes the DNA-damage response. Conversely, ectopic expression of CXCR2 results in premature senescence via a p53-dependent mechanism. Cells undergoing OIS secrete multiple CXCR2-binding chemokines in a program that is regulated by the NF-kappaB and C/EBPbeta transcription factors and coordinately induce CXCR2 expression. CXCR2 upregulation is also observed in preneoplastic lesions in vivo. These results suggest that senescent cells activate a self-amplifying secretory network in which CXCR2-binding chemokines reinforce growth arrest.


Asunto(s)
Senescencia Celular , Receptores de Interleucina-8B/genética , Receptores de Interleucina-8B/metabolismo , Transducción de Señal , Adenocarcinoma/metabolismo , Animales , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Línea Celular , Línea Celular Tumoral , Quimiocinas/metabolismo , Daño del ADN , Regulación hacia Abajo , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Ligandos , Neoplasias Pulmonares/metabolismo , Ratones , FN-kappa B/metabolismo , Lesiones Precancerosas/metabolismo , Interferencia de ARN , Receptores de Interleucina-8A/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
8.
Annu Rev Genomics Hum Genet ; 18: 87-113, 2017 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-28859573

RESUMEN

Until recently, DNA damage arising from physiological DNA metabolism was considered a detrimental by-product for cells. However, an increasing amount of evidence has shown that DNA damage could have a positive role in transcription activation. In particular, DNA damage has been detected in transcriptional elements following different stimuli. These physiological DNA breaks are thought to be instrumental for the correct expression of genomic loci through different mechanisms. In this regard, although a plethora of methods are available to precisely map transcribed regions and transcription start sites, commonly used techniques for mapping DNA breaks lack sufficient resolution and sensitivity to draw a robust correlation between DNA damage generation and transcription. Recently, however, several methods have been developed to map DNA damage at single-nucleotide resolution, thus providing a new set of tools to correlate DNA damage and transcription. Here, we review how DNA damage can positively regulate transcription initiation, the current techniques for mapping DNA breaks at high resolution, and how these techniques can benefit future studies of DNA damage and transcription.


Asunto(s)
Daño del ADN , Reparación del ADN , Pruebas de Mutagenicidad/métodos , Análisis de Secuencia de ADN/métodos , Transcripción Genética , Animales , ADN/metabolismo , Eucariontes/genética , Regulación de la Expresión Génica , Humanos
9.
Chem Rev ; 118(8): 4365-4403, 2018 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-29600857

RESUMEN

Coding for proteins has been considered the main function of RNA since the "central dogma" of biology was proposed. The discovery of noncoding transcripts shed light on additional roles of RNA, ranging from the support of polypeptide synthesis, to the assembly of subnuclear structures, to gene expression modulation. Cellular RNA has therefore been recognized as a central player in often unanticipated biological processes, including genomic stability. This ever-expanding list of functions inspired us to think of RNA as a "smart" phone, which has replaced the older obsolete "cellular" phone. In this review, we summarize the last two decades of advances in research on the interface between RNA biology and genome stability. We start with an account of the emergence of noncoding RNA, and then we discuss the involvement of RNA in DNA damage signaling and repair, telomere maintenance, and genomic rearrangements. We continue with the depiction of single-molecule RNA detection techniques, and we conclude by illustrating the possibilities of RNA modulation in hopes of creating or improving new therapies. The widespread biological functions of RNA have made this molecule a reoccurring theme in basic and translational research, warranting it the transcendence from classically studied "cellular" RNA to "smart" RNA.


Asunto(s)
Inestabilidad Genómica , ARN no Traducido/genética , Roturas del ADN de Doble Cadena , Daño del ADN , Regulación de la Expresión Génica , Humanos , Interferencia de ARN , Proteínas de Unión al ARN/metabolismo , Transcripción Genética
10.
Int J Mol Sci ; 21(24)2020 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-33371452

RESUMEN

Because of their intrinsic characteristics, telomeres are genomic loci that pose significant problems during the replication of the genome. In particular, it has been observed that telomeres that are maintained in cancer cells by the alternative mechanism of the lengthening of telomeres (ALT) harbor higher levels of replicative stress compared with telomerase-positive cancer cells. R-loops are three-stranded structures formed by a DNA:RNA hybrid and a displaced ssDNA. Emerging evidence suggests that controlling the levels of R-loops at ALT telomeres is critical for telomere maintenance. In fact, on the one hand, they favor telomere recombination, but on the other, they are a source of detrimental replicative stress. DRIP (DNA:RNA immunoprecipitation) is the main technique used for the detection of R-loops, and it is based on the use of the S9.6 antibody, which recognizes preferentially DNA:RNA hybrids in a sequence-independent manner. The detection of DNA:RNA hybrids in repetitive sequences such as telomeres requires some additional precautions as a result of their repetitive nature. Here, we share an optimized protocol for the detection of telomeric DNA:RNA hybrids, and we demonstrate its application in an ALT and in a telomerase-positive cell line. We demonstrate that ALT telomeres bear higher levels of DNA:RNA hybrids, and we propose this method as a reliable way to detect them in telomeres.


Asunto(s)
ADN/análisis , Reacción en Cadena de la Polimerasa/métodos , ARN/análisis , Telómero/genética , ADN/genética , Células HeLa , Humanos , ARN/genética , Telomerasa/metabolismo , Homeostasis del Telómero
11.
EMBO J ; 34(10): 1371-84, 2015 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-25820263

RESUMEN

The causal role of aneuploidy in cancer initiation remains under debate since mutations of euploidy-controlling genes reduce cell fitness but aneuploidy strongly associates with human cancers. Telomerase activation allows immortal growth by stabilizing telomere length, but its role in aneuploidy survival has not been characterized. Here, we analyze the response of primary human cells and murine hematopoietic stem cells (HSCs) to aneuploidy induction and the role of telomeres and the telomerase in this process. The study shows that aneuploidy induces replication stress at telomeres leading to telomeric DNA damage and p53 activation. This results in p53/Rb-dependent, premature senescence of human fibroblast, and in the depletion of hematopoietic cells in telomerase-deficient mice. Endogenous telomerase expression in HSCs and enforced expression of telomerase in human fibroblasts are sufficient to abrogate aneuploidy-induced replication stress at telomeres and the consequent induction of premature senescence and hematopoietic cell depletion. Together, these results identify telomerase as an aneuploidy survival factor in mammalian cells based on its capacity to alleviate telomere replication stress in response to aneuploidy induction.


Asunto(s)
Aneuploidia , Telomerasa/metabolismo , Telómero/metabolismo , Animales , Senescencia Celular/genética , Senescencia Celular/fisiología , Replicación del ADN/genética , Replicación del ADN/fisiología , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones , Telomerasa/genética , Telómero/genética , Proteína p53 Supresora de Tumor/metabolismo
13.
Nat Rev Mol Cell Biol ; 8(9): 729-40, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17667954

RESUMEN

Cells continually experience stress and damage from exogenous and endogenous sources, and their responses range from complete recovery to cell death. Proliferating cells can initiate an additional response by adopting a state of permanent cell-cycle arrest that is termed cellular senescence. Understanding the causes and consequences of cellular senescence has provided novel insights into how cells react to stress, especially genotoxic stress, and how this cellular response can affect complex organismal processes such as the development of cancer and ageing.


Asunto(s)
Envejecimiento , Senescencia Celular/fisiología , Neoplasias/etiología , Animales , Ciclo Celular , Daño del ADN , Humanos
14.
J Cell Sci ; 129(7): 1468-76, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26906421

RESUMEN

The DNA damage response (DDR) plays a central role in preserving genome integrity. Recently, we reported that the endoribonucleases DICER and DROSHA contribute to DDR activation by generating small non-coding RNAs, termed DNA damage response RNA (DDRNA), carrying the sequence of the damaged locus. It is presently unclear whether DDRNAs act by promoting the primary recognition of DNA lesions or the secondary recruitment of DDR factors into cytologically detectable foci and consequent signal amplification. Here, we demonstrate that DICER and DROSHA are dispensable for primary recruitment of the DDR sensor NBS1 to DNA damage sites. Instead, the accumulation of the DDR mediators MDC1 and 53BP1 (also known as TP53BP1), markers of secondary recruitment, is reduced in DICER- or DROSHA-inactivated cells. In addition, NBS1 (also known as NBN) primary recruitment is resistant to RNA degradation, consistent with the notion that RNA is dispensable for primary recognition of DNA lesions. We propose that DICER, DROSHA and DDRNAs act in the response to DNA damage after primary recognition of DNA lesions and, together with γH2AX, are essential for enabling the secondary recruitment of DDR factors and fuel the amplification of DDR signaling.


Asunto(s)
ARN Helicasas DEAD-box/genética , Daño del ADN/genética , Reparación del ADN/inmunología , Histonas/metabolismo , Ribonucleasa III/genética , Proteínas Adaptadoras Transductoras de Señales , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Reparación del ADN/genética , Humanos , Proteínas Nucleares/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , Ribonucleasa Pancreática/metabolismo , Transactivadores/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo
15.
Nature ; 488(7410): 231-5, 2012 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-22722852

RESUMEN

Non-coding RNAs (ncRNAs) are involved in an increasingly recognized number of cellular events. Some ncRNAs are processed by DICER and DROSHA RNases to give rise to small double-stranded RNAs involved in RNA interference (RNAi). The DNA-damage response (DDR) is a signalling pathway that originates from a DNA lesion and arrests cell proliferation3. So far, DICER and DROSHA RNA products have not been reported to control DDR activation. Here we show, in human, mouse and zebrafish, that DICER and DROSHA, but not downstream elements of the RNAi pathway, are necessary to activate the DDR upon exogenous DNA damage and oncogene-induced genotoxic stress, as studied by DDR foci formation and by checkpoint assays. DDR foci are sensitive to RNase A treatment, and DICER- and DROSHA-dependent RNA products are required to restore DDR foci in RNase-A-treated cells. Through RNA deep sequencing and the study of DDR activation at a single inducible DNA double-strand break, we demonstrate that DDR foci formation requires site-specific DICER- and DROSHA-dependent small RNAs, named DDRNAs, which act in a MRE11­RAD50­NBS1-complex-dependent manner (MRE11 also known as MRE11A; NBS1 also known as NBN). DDRNAs, either chemically synthesized or in vitro generated by DICER cleavage, are sufficient to restore the DDR in RNase-A-treated cells, also in the absence of other cellular RNAs. Our results describe an unanticipated direct role of a novel class of ncRNAs in the control of DDR activation at sites of DNA damage.


Asunto(s)
Daño del ADN/genética , ARN no Traducido/genética , Ribonucleasa III/genética , Pez Cebra/genética , Animales , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Roturas del ADN de Doble Cadena , Enzimas Reparadoras del ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Activación Enzimática , Células HEK293 , Células HeLa , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Ratones , Proteínas Nucleares/metabolismo , Interferencia de ARN , ARN no Traducido/biosíntesis , Ribonucleasa Pancreática/metabolismo , Análisis de Secuencia de ARN , Especificidad por Sustrato/genética
16.
EMBO Rep ; 16(2): 221-31, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25527408

RESUMEN

Eukaryotic cells respond to DNA double-strand breaks (DSBs) by activating a checkpoint that depends on the protein kinases Tel1/ATM and Mec1/ATR. Mec1/ATR is activated by RPA-coated single-stranded DNA (ssDNA), which arises upon nucleolytic degradation (resection) of the DSB. Emerging evidences indicate that RNA-processing factors play critical, yet poorly understood, roles in genomic stability. Here, we provide evidence that the Saccharomyces cerevisiae RNA decay factors Xrn1, Rrp6 and Trf4 regulate Mec1/ATR activation by promoting generation of RPA-coated ssDNA. The lack of Xrn1 inhibits ssDNA generation at the DSB by preventing the loading of the MRX complex. By contrast, DSB resection is not affected in the absence of Rrp6 or Trf4, but their lack impairs the recruitment of RPA, and therefore of Mec1, to the DSB. Rrp6 and Trf4 inactivation affects neither Rad51/Rad52 association nor DSB repair by homologous recombination (HR), suggesting that full Mec1 activation requires higher amount of RPA-coated ssDNA than HR-mediated repair. Noteworthy, deep transcriptome analyses do not identify common misregulated gene expression that could explain the observed phenotypes. Our results provide a novel link between RNA processing and genome stability.


Asunto(s)
ADN de Cadena Simple/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína de Replicación A/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN/genética , Reparación del ADN/fisiología , ADN de Cadena Simple/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Exorribonucleasas/genética , Exorribonucleasas/metabolismo , Complejo Multienzimático de Ribonucleasas del Exosoma/genética , Complejo Multienzimático de Ribonucleasas del Exosoma/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Serina-Treonina Quinasas/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
17.
EMBO J ; 31(13): 2839-51, 2012 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-22569128

RESUMEN

In normal human somatic cells, telomere dysfunction causes cellular senescence, a stable proliferative arrest with tumour suppressing properties. Whether telomere dysfunction-induced senescence (TDIS) suppresses cancer growth in humans, however, is unknown. Here, we demonstrate that multiple and distinct human cancer precursor lesions, but not corresponding malignant cancers, are comprised of cells that display hallmarks of TDIS. Furthermore, we demonstrate that oncogenic signalling, frequently associated with initiating cancer growth in humans, dramatically affected telomere structure and function by causing telomeric replication stress, rapid and stochastic telomere attrition, and consequently telomere dysfunction in cells that lack hTERT activity. DNA replication stress induced by drugs also resulted in telomere dysfunction and cellular senescence in normal human cells, demonstrating that telomeric repeats indeed are hypersensitive to DNA replication stress. Our data reveal that TDIS, accelerated by oncogene-induced DNA replication stress, is a biological response of cells in human cancer precursor lesions and provide strong evidence that TDIS is a critical tumour suppressing mechanism in humans.


Asunto(s)
Senescencia Celular/fisiología , Oncogenes/fisiología , Telómero/fisiología , Línea Celular , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Senescencia Celular/efectos de los fármacos , Senescencia Celular/genética , Replicación del ADN/efectos de los fármacos , Replicación del ADN/genética , Replicación del ADN/fisiología , Humanos , Oncogenes/efectos de los fármacos , Oncogenes/genética , Inhibidores de la Síntesis de la Proteína/farmacología , Puromicina/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/fisiología , Telómero/efectos de los fármacos , Telómero/genética
18.
Nucleic Acids Res ; 40(12): 5332-42, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22379135

RESUMEN

Bromodeoxyuridine (5-bromo-2'-deoxyuridine, BrdU) is a halogenated nucleotide of low toxicity commonly used to monitor DNA replication. It is considered a valuable tool for in vitro and in vivo studies, including the detection of the small population of neural stem cells (NSC) in the mammalian brain. Here, we show that NSC grown in self-renewing conditions in vitro, when exposed to BrdU, lose the expression of stem cell markers like Nestin, Sox2 and Pax6 and undergo glial differentiation, strongly up-regulating the astrocytic marker GFAP. The onset of GFAP expression in BrdU exposed NSC was paralleled by a reduced expression of key DNA methyltransferases (DNMT) and a rapid loss of global DNA CpG methylation, as we determined by our specially developed analytic assay. Remarkably, a known DNA demethylating compound, 5-aza-2'-deoxycytidine (Decitabine), had similar effect on demethylation and differentiation of NSC. Since our key findings apply also to NSC derived from murine forebrain, our observations strongly suggest more caution in BrdU uses in stem cells research. We also propose that BrdU and its related substances may also open new opportunities for differentiation therapy in oncology.


Asunto(s)
Astrocitos/citología , Bromodesoxiuridina/farmacología , Metilación de ADN/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Puntos de Control del Ciclo Celular , Diferenciación Celular/efectos de los fármacos , Línea Celular , Células Cultivadas , Células-Madre Neurales/citología , Prosencéfalo/citología
19.
Semin Cancer Biol ; 21(6): 360-6, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21946622

RESUMEN

Cellular senescence is eminently characterized by a permanent cell cycle arrest and the acquisition of morphological, physiological and epigenetic changes. The establishment of cellular senescence can occur in response to telomere attrition associated with cell turnover and ageing or following oncogene activation. Although seemingly two distinct phenomena, cellular senescence and cancer share similarly altered global epigenetic profiles comprising changes in DNA methylation, involving global hypomethylation of repetitive DNA sequences and regional hypermethylation of some gene promoters, and in histone post-translational modifications. As epigenetic and genetic alterations are likely to act synergistically in cancer, anomalous epigenetic marks acquired during ageing or in response to oncogene activation might play important roles in tumorigenesis and cancer progression. These potentially tumor-promoting epigenetic alterations include transcriptional repression of genes encoding tumor suppressors or developmentally regulated proteins, expression of non-coding repetitive RNAs and acquisition of distinct heterochromatin marks that may contribute to suppress cell death by reducing DNA damage response. Cellular senescence may thus be viewed as a double-edged sword that, although acting as a potent anti-proliferative barrier, may pave the way to tumorigenesis in senescence-escaping cells by altering their epigenetic make up.


Asunto(s)
Transformación Celular Neoplásica/genética , Senescencia Celular/genética , Epigénesis Genética , Neoplasias/patología , Islas de CpG , Metilación de ADN , Humanos , Neoplasias/genética , Oncogenes , Regiones Promotoras Genéticas
20.
Cell Death Dis ; 14(2): 96, 2023 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-36759506

RESUMEN

Telomere maintenance is necessary to maintain cancer cell unlimited viability. However, the mechanisms maintaining telomere length in colorectal cancer (CRC) have not been extensively investigated. Telomere maintenance mechanisms (TMM) include the re-expression of telomerase or alternative lengthening of telomeres (ALT). ALT is genetically associated with somatic alterations in alpha-thalassemia/mental retardation X-linked (ATRX) and death domain-associated protein (DAXX) genes. Cells displaying ALT present distinctive features including C-circles made of telomeric DNA, long and heterogenous telomeric tracts, and telomeric DNA co-localized with promyelocytic leukemia (PML) bodies forming so-called ALT-associated PML bodies (APBs). Here, we identified mutations in ATRX and/or DAXX genes in an extensive collection of CRC samples including 119 patient-derived organoids (PDOs) and 232 established CRC cell lines. C-circles measured in CRC PDOs and cell lines showed low levels overall. We also observed that CRC PDOs and cell lines did not display a significant accumulation of APBs or long telomeres with no appreciable differences between wild-type and mutated ATRX/DAXX samples. Overall, our extensive analyses indicate that CRC is not prone to engage ALT, even when carrying genetic lesions in ATRX and/or DAXX, and support the notion that ATRX/DAXX genomic footprints are not reliable predictors of ALT.


Asunto(s)
Neoplasias Colorrectales , Discapacidad Intelectual , Telomerasa , Talasemia alfa , Humanos , Proteína Nuclear Ligada al Cromosoma X/genética , Proteína Nuclear Ligada al Cromosoma X/metabolismo , Homeostasis del Telómero/genética , Proteínas Co-Represoras/genética , Proteínas Co-Represoras/metabolismo , Telomerasa/genética , Telomerasa/metabolismo , Mutación/genética , Línea Celular , Telómero/genética , Telómero/metabolismo , Organoides/metabolismo , Neoplasias Colorrectales/genética , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA