Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mutat Res ; 756(1-2): 86-94, 2013 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-23721903

RESUMEN

Mitochondria are the main cellular source of Reactive Oxygen Species (ROS). Alterations of mitochondrial metabolism and consequent loss of mitochondrial membrane potential may lead to redox imbalance and in turn to DNA damage, chromosomal instability and apoptosis. On the other hand, impaired mitochondrial functions may either exacerbate the detrimental effects of geno- and cytotoxic agents or may bring beneficial cellular responses. To study the role of mitochondria within this framework, AG01522 human primary fibroblasts were incubated with the mitochondrial polymerase γ inhibitor 2',3'-dideoxycytidine (ddC), leading to mitochondrial DNA (mtDNA) depletion and to mitochondrial dysfunctions. The successful treatment toward mtDNA depletion was confirmed by Complex-IV subunit I (COX-I) immunofluorescence and western blot assays. mtDNA-depleted cells and their counterparts were ultrastructurally characterized by transmission electron microscopy. mtDNA-depleted cells showed dramatic mitochondrial alterations such as fragmentation and cristae disruption along with a reduction of the mitochondrial membrane potential and elevated levels of ROS. Despite increased ROS levels, we did not find any difference in telomere length between ddC-treated and untreated cells. The spontaneous rate of DNA double-strand breaks (DSBs) and chromosome aberrations was significantly enhanced in mtDNA-depleted cells whereas the induction of DSBs by low-Linear Energy Transfer (LET) (X-rays; 7.7keV/µm protons) and high-LET radiations (28.5keV/µm protons) did not differ when compared with normal cells. However, in irradiated cells impaired mitochondrial functions seemed to bring beneficial cellular responses to the detrimental effect of radiations. In fact, after X-irradiation mtDNA-depleted cells show less remaining unrejoined DSBs than normal cells and furthermore a lower induction of cytogenetic damage. Overall, these data show that active mitochondrial functions are required for the proper maintenance of cellular genome stability in primary fibroblasts.


Asunto(s)
Aberraciones Cromosómicas , ADN Mitocondrial/metabolismo , Fibroblastos/efectos de la radiación , Mitocondrias/efectos de la radiación , Zalcitabina/farmacología , Antimetabolitos/farmacología , Supervivencia Celular/efectos de la radiación , Células Cultivadas , Daño del ADN/genética , Daño del ADN/efectos de la radiación , Reparación del ADN/genética , Reparación del ADN/efectos de la radiación , ADN Mitocondrial/efectos de los fármacos , ADN Mitocondrial/efectos de la radiación , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Hibridación Fluorescente in Situ , Transferencia Lineal de Energía , Potencial de la Membrana Mitocondrial/efectos de la radiación , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Especies Reactivas de Oxígeno/metabolismo , Telómero/genética , Rayos X
2.
Commun Biol ; 6(1): 234, 2023 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-36864251

RESUMEN

Telomeres are nucleoprotein structures at eukaryotic chromosome termini. Their stability is preserved by a six-protein complex named shelterin. Among these, TRF1 binds telomere duplex and assists DNA replication with mechanisms only partly clarified. Here we found that poly (ADP-ribose) polymerase 1 (PARP1) interacts and covalently PARylates TRF1 in S-phase modifying its DNA affinity. Therefore, genetic and pharmacological inhibition of PARP1 impairs the dynamic association of TRF1 and the bromodeoxyuridine incorporation at replicating telomeres. Inhibition of PARP1 also affects the recruitment of WRN and BLM helicases in TRF1 containing complexes during S-phase, triggering replication-dependent DNA-damage and telomere fragility. This work unveils an unprecedented role for PARP1 as a "surveillant" of telomere replication, which orchestrates protein dynamics at proceeding replication fork.


Asunto(s)
Complejo Shelterina , Telómero , ADP-Ribosilación , Daño del ADN , ADN Helicasas , Telómero/genética , Poli(ADP-Ribosa) Polimerasa-1/metabolismo
3.
Mutat Res ; 740(1-2): 13-20, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23220250

RESUMEN

Many and varied are the proposed mechanisms that lead to resistance to ionizing radiation treatment. Among them, an inverse relationship between telomere length and radioresistance has been recently advanced. Investigating such a relationship in TK6 lymphoblasts, we found that clones originating from cells survived to 4Gy of X-rays showed a significantly higher telomere length when compared with clones grown from untreated cells. The lengthening observed was not attributable to a radiation-induced increase in telomerase activity, as demonstrated by TRAP assay performed in the dose range of 1-10Gy. Given the evidence that TK6 whole population was characterized by heterogeneity in cellular mean telomere length and telomere loss, we tested the hypothesis that a process of selection may favour cells with longer telomeres (more radioresistant cells) following exposure to irradiation. In order to do this 15 independent TK6 clones were selected and characterized for telomere length and loss on the basis of q-FISH and flow-FISH analysis. Among the screened clones four characterized by long telomeres and four characterized by short telomeres were tested for their radiosensitivity by means of clonogenic assay. The results obtained showed that, in our experimental conditions (cellular model, radiation doses) no significant correlation was observed between radiosensitivity and mean telomere lengths, whereas a positive correlation was observed with respect to telomere loss. Overall, these results indicate that telomere loss and not mean telomere length plays a critical role in the phenomenon of radiosensitivity/radioresistance.


Asunto(s)
Linfocitos/efectos de la radiación , Tolerancia a Radiación/genética , Acortamiento del Telómero/efectos de la radiación , Línea Celular , Humanos , Telomerasa/metabolismo , Homeostasis del Telómero/efectos de la radiación
4.
Apoptosis ; 16(9): 940-9, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21671007

RESUMEN

The BH3-only Bcl-2 subfamily member Bim is a well known apoptosis promoting protein. However, the mechanisms upstream of mitochondrion membrane permeability by which Bim is involved in apoptosis have been poorly investigated, particularly in response to agents capable of interfering with the cytoskeleton architecture and arresting cells in mitosis. Based on the observation that Bim is sequestered on the microtubule-array by interaction with the light chain of dynein, we have investigated upon depolymerisation, whether Bim could be involved in the commitment of apoptosis. With this purpose H460 Non Small Lung Cancer Cells (NSLC) were treated with the microtubule damaging agent combretastatin-A4 (CA-4) (7.5 nM; 8-48 h), and various parameters were investigated. Upon treatment, cells arrested in mitosis and died through a caspase-3-dependent mitotic catastrophe. Transient knock down of Bim drastically reduced apoptosis, indicating that this protein was involved in cell death as induced by microtubules disorganisation. In response to increasing conditions of microtubules depolymerisation, we found that the protein level of Bim was strongly upregulated in a time-dependent manner at transcriptional level. Furthermore, Bim was released from microtubule-associated components. Bim was translocated to mitochondria, even in a condition of protein synthesis inhibition, where it showed a markedly increased interaction with Bcl-2. In turn, the fraction of Bax bound to Bcl-2 decreases in response to treatment, thereby indicating that Bim possibly promotes Bax release from the pro-survival protein Bcl-2. Overall, we demonstrated that Bim is required for the CA-4-induced cell death in the H460 lung cancer cell line via activation of the mitochondrial signalling pathway. Defining the contribution of Bim to the mechanism of apoptosis may offer some different clues in view of developing new strategies for chemotherapy with CA-4, underlining the relevance of the cytoskeleton integrity in the apoptotic response.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Estilbenos/farmacología , Antineoplásicos Fitogénicos/farmacología , Proteína 11 Similar a Bcl2 , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Caspasa 3/metabolismo , Línea Celular Tumoral/efectos de los fármacos , Citosol/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunoprecipitación , Puntos de Control de la Fase M del Ciclo Celular , Microscopía Confocal , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Mitocondrias/metabolismo , Transporte de Proteínas , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Transfección
5.
Cell Mol Biol (Noisy-le-grand) ; 56 Suppl: OL1299-317, 2010 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-20937217

RESUMEN

Somatic mutations in the genes members of WNT/ß-catenin pathway, especially in CTNNB1 codifying for ß-catenin, have been found to play an important role in hepatocarcinogenesis. The purpose of this work is to characterize alterations of the WNT/ß-catenin signalling pathway, and to study the expression pattern of a panel of microRNAs and proteins potentially involved in the pathogenesis of liver cancer. In this respect, the molecular characterization of the most used liver cancer cell lines HuH6, Hep3B, HepG2, and HLE, could represent a useful tool to identify novel molecular markers for hepatic tumour. A significant modulation of FZD7, NLK, RHOU, SOX17, TCF7L2, TLE1, SLC9A3R1 and WNT10A transcripts was observed in all the four liver cancer cell lines. The analysis of selected microRNAs showed that miR-122a, miR-125a and miR-150 could be suitable candidates to discriminate tumoural versus normal human primary hepatocytes. Finally, Grb-2 protein expression resulted to be increased more than two-fold in liver cancer cell lines in comparison to normal human primary hepatocytes. These advances in the knowledge of molecular mechanisms involved in the pathogenesis of liver cancer may provide new potential biomarkers and molecular targets for the diagnosis and therapy.


Asunto(s)
Neoplasias Hepáticas/metabolismo , MicroARNs/metabolismo , Proteoma/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Línea Celular Tumoral , Proteína Adaptadora GRB2/metabolismo , Perfilación de la Expresión Génica , Hepatocitos/metabolismo , Humanos , Transducción de Señal
6.
J Exp Clin Cancer Res ; 38(1): 311, 2019 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-31311580

RESUMEN

BACKGROUND: Glioblastoma is the most aggressive and most lethal primary brain tumor in the adulthood. Current standard therapies are not curative and novel therapeutic options are urgently required. Present knowledge suggests that the continued glioblastoma growth and recurrence is determined by glioblastoma stem-like cells (GSCs), which display self-renewal, tumorigenic potential, and increased radio- and chemo-resistance. The G-quadruplex ligand RHPS4 displays in vitro radiosensitizing effect in GBM radioresistant cells through the targeting and dysfunctionalization of telomeres but RHPS4 and Ionizing Radiation (IR) combined treatment efficacy in vivo has not been explored so far. METHODS: RHPS4 and IR combined effects were tested in vivo in a heterotopic mice xenograft model and in vitro in stem-like cells derived from U251MG and from four GBM patients. Cell growth assays, cytogenetic analysis, immunoblotting, gene expression and cytofluorimetric analysis were performed in order to characterize the response of differentiated and stem-like cells to RHPS4 and IR in single and combined treatments. RESULTS: RHPS4 administration and IR exposure is very effective in blocking tumor growth in vivo up to 65 days. The tumor volume reduction and the long-term tumor control suggested the targeting of the stem cell compartment. Interestingly, RHPS4 treatment was able to strongly reduce cell proliferation in GSCs but, unexpectedly, did not synergize with IR. Lack of radiosensitization was supported by the GSCs telomeric-resistance observed as the total absence of telomere-involving chromosomal aberrations. Remarkably, RHPS4 treatment determined a strong reduction of CHK1 and RAD51 proteins and transcript levels suggesting that the inhibition of GSCs growth is determined by the impairment of the replication stress (RS) response and DNA repair. CONCLUSIONS: We propose that the potent antiproliferative effect of RHPS4 in GSCs is not determined by telomeric dysfunction but is achieved by the induction of RS and by the concomitant depletion of CHK1 and RAD51, leading to DNA damage and cell death. These data open to novel therapeutic options for the targeting of GSCs, indicating that the combined inhibition of cell-cycle checkpoints and DNA repair proteins provides the most effective means to overcome resistance of GSC to genotoxic insults.


Asunto(s)
Acridinas/administración & dosificación , Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Células Madre Neoplásicas/efectos de los fármacos , Fármacos Sensibilizantes a Radiaciones/administración & dosificación , Acridinas/farmacología , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/genética , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Ratones , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Fármacos Sensibilizantes a Radiaciones/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Radiat Res ; 170(3): 345-52, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18763866

RESUMEN

Nijmegen breakage syndrome shares several common cellular features with ataxia telangiectasia, including chromosomal instability and aberrant S- and G2-phase checkpoint regulation. We show here that after irradiation, NBS1 interacts physically with both BRCA1 and SMC1, a component of the cohesin complex, and that their interactions are completely abolished in AT cells. It is noted that BRCA1 is required for the interaction of NBS1 with SMC1, whereas the reverse is not the case, since BRCA1 is able to bind to NBS1 in the absence of an NBS1/SMC1 interaction as observed in MRE11- or RAD50-deficient cells. This indicates that ATM and BRCA1 are upstream of the NBS1/SMC1 interaction. Furthermore, the interaction of NBS1 with SMC1 requires both conserved domains of NBS in the N-terminus and the C-terminus, since they are indispensable for binding of NBS1 to BRCA1 and to MRE11/ATM, respectively. The interaction of NBS1 with SMC1 and the resulting phosphorylation are compromised in the clones lacking either the N- or C-terminus of NBS1, and as a consequence, chromatid-type aberrations are enhanced after irradiation. Our results reveal that ATM plays a fundamental role in promoting the radiation-induced interaction of NBS1 with SMC1 in the presence of BRCA1, leading to the maintenance of chromosomal integrity.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Cromátides/genética , Cromátides/efectos de la radiación , Proteínas Cromosómicas no Histona/metabolismo , Aberraciones Cromosómicas , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Traumatismos por Radiación/genética , Transducción de Señal/fisiología , Proteínas Supresoras de Tumor/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada , Células Cultivadas , Relación Dosis-Respuesta en la Radiación , Humanos , Dosis de Radiación , Transducción de Señal/efectos de la radiación
8.
Amino Acids ; 34(3): 403-12, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17610126

RESUMEN

In this study, polyamine oxidase from maize (MPAO), which is involved in the terminal catabolism of spermidine and spermine to produce an aminoaldehyde, 1,3-diaminopropane and H(2)O(2), has been conditionally expressed at high levels in the nucleus of MCF-7 human breast cancer cells, with the aim to interfere with polyamine homeostasis and cell proliferation. Recombinant MPAO expression induced accumulation of a high amount of 1,3-diaminopropane, an increase of putrescine levels and no alteration in the cellular content of spermine and spermidine. Furthermore, recombinant MPAO expression did not interfere with cell growth of MCF-7 cells under normal conditions but it did confer higher growth sensitivity to etoposide, a DNA topoisomerase II inhibitor widely used as antineoplastic drug. These data suggest polyamine oxidases as a potential tool to improve the efficiency of antiproliferative agents despite the difficulty to interfere with cellular homeostasis of spermine and spermidine.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Núcleo Celular/genética , Etopósido/farmacología , Regulación Enzimológica de la Expresión Génica , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Zea mays/enzimología , Antineoplásicos Fitogénicos/farmacología , Antineoplásicos Fitogénicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Doxiciclina/farmacología , Etopósido/uso terapéutico , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Poliaminas/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Zea mays/genética , Poliamino Oxidasa
9.
Eur J Med Genet ; 50(3): 176-87, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17395558

RESUMEN

Genetic heterogeneity in Nijmegen breakage syndrome (NBS) is highlighted by patients showing clinical and cellular features of NBS but with no mutations in NBS1 and normal levels of nibrin. NBS is an autosomal recessive disorder, whose clinical cellular signs include growth and developmental defects, dysmorphic facies, immunodeficiency, cancer predisposition, chromosomal instability and radiosensitivity. NBS is caused by mutations in the NBS1 gene, whose product is part of the MRE11/RAD50/NBS1 complex involved in the DNA double-strand break (DSB) response pathway. Since the identification of the NBS1 gene, patients with NBS clinical signs, particularly severe congenital microcephaly, are screened for mutations in the NBS1 gene. Further analyses include X-ray-induced chromosome aberrations, telomere analysis, kinetics of DSBs repair, levels of a panel of proteins involved in the maintenance of genetic stability, radiation-induced phosphorylation of various substrates and cell cycle analysis. We describe a patient with a NBS clinical phenotype, chromosomal sensitivity to X-rays but without mutations in the whole NBS1 or in the Cernunnos gene. Enhanced response to irradiation was mediated neither by DSBs rejoining defects nor by the NBS/AT-dependent DNA-damage response pathway. Notably, we found that primary fibroblasts from this patient displayed telomere length alterations. Cross-talk between pathways controlling response to DSBs and those involved in maintaining telomeres has been shown in the present patient. Dissecting the cellular phenotype of radiosensitive NBS-like patients represents a useful tool for the research of new genes involved in the cellular response to DSBs.


Asunto(s)
Anomalías Craneofaciales/genética , Microcefalia/genética , Síndrome de Nijmegen/genética , Tolerancia a Radiación/genética , Telómero/genética , Proteínas de Ciclo Celular/genética , Cromosomas Humanos/efectos de la radiación , Reparación del ADN/genética , Femenino , Humanos , Masculino , Síndrome de Nijmegen/diagnóstico , Proteínas Nucleares/genética , Fenotipo , Telómero/ultraestructura
10.
Mutat Res Rev Mutat Res ; 773: 204-219, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28927529

RESUMEN

One of the hallmarks of cancer consists in the ability of tumor cells to divide indefinitely, and to maintain stable telomere lengths throughout the activation of specific telomere maintenance mechanisms (TMM). Therefore in the last fifteen years, researchers proposed to target telomerase or telomeric structure in order to block limitless replicative potential of cancer cells providing a fascinating strategy for a broad-spectrum cancer therapy. In the present review, we report in vitro and in vivo evidence regarding the use of chemical agents targeting both telomerase or telomere structure and showing promising antitumor effects when used in combination with ionizing radiation (IR). RNA interference, antisense oligonucleotides (e.g., GRN163L), non-nucleoside inhibitors (e.g., BIBR1532) and nucleoside analogs (e.g., AZT) represent some of the most potent strategies to inhibit telomerase activity used in combination with IR. Furthermore, radiosensitizing effects were demonstrated also for agents acting directly on the telomeric structure such as G4-ligands (e.g., RHPS4 and Telomestatin) or telomeric-oligos (T-oligos). To date, some of these compounds are under clinical evaluation (e.g., GRN163L and KML001). Advantages of Telomere/Telomerase Targeting Compounds (T/TTCs) coupled with radiotherapy may be relevant in the treatment of radioresistant tumors and in the development of new optimized treatment plans with reduced dose adsorbed by patients and consequent attenuation of short- end long-term side effects. Pros and cons of possible future applications in cancer therapy based on the combination of T/TCCs and radiation treatment are discussed.


Asunto(s)
Neoplasias/tratamiento farmacológico , Neoplasias/radioterapia , Radiación Ionizante , Telomerasa/metabolismo , Telómero/metabolismo , Aminobenzoatos/farmacología , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Modelos Animales de Enfermedad , Homeostasis/efectos de los fármacos , Homeostasis/efectos de la radiación , Humanos , Naftalenos/farmacología , Oligonucleótidos/farmacología , Interferencia de ARN , Fármacos Sensibilizantes a Radiaciones/farmacología
11.
Radiat Prot Dosimetry ; 122(1-4): 176-9, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17223635

RESUMEN

Telomeres are specialised nucleoprotein complexes that serve as protective caps of linear eukaryotic chromosomes. The loss of the ends of the chromosomes due to these un-rejoined double strand breaks (DSBs) may not be lethal to the cell, but may instead result in the loss of functional telomeres, chromosome fusions and initiation of breakage/fusion/bridge cycle-induced chromosome instability. The telomeres also participate in the process of DNA repair, as evidenced by 'de novo' synthesis of telomere repeats at DSBs and by the capacity of telomeres to binding the essential components of the DNA repair machinery. Based on the observation that high-LET radiations efficiently induce chromosome aberrations, it was tested whether protons were able to affect telomere structure. Human primary fibroblasts (HFFF2) and mouse embryonic fibroblasts (MEFs) were irradiated with 4 Gy of 3 MeV protons at the radiobiology facility of the INFN-LNL. Experiments with X rays were also carried out. Cells were fixed after either 24 h or 15 d from treatment. A difference in average telomere length, measured by quantitative fluorescence in situ hybridisation (Q-FISH), between X rays and protons treatment was observed. X rays are able to modify telomere length in HFFF2 harvested at a later time. On the other hand, 3 MeV low-energy protons induced, both in HFFF2 and in MEFs, a significant increase in telomere length at short as well as at long harvesting time periods from treatment. These results seem to indicate that lesions characterised by different complexity, as those expected after low-energy protons and those induced by damage similar to that induced by sparsely ionising radiation, are able to modulate telomere elongation at different time periods.


Asunto(s)
Daño del ADN , ADN/genética , ADN/efectos de la radiación , Fibroblastos/fisiología , Fibroblastos/efectos de la radiación , Telómero/genética , Telómero/efectos de la radiación , Animales , Células Cultivadas , Relación Dosis-Respuesta en la Radiación , Fibroblastos/citología , Humanos , Transferencia Lineal de Energía/fisiología , Transferencia Lineal de Energía/efectos de la radiación , Ratones , Dosis de Radiación , Radiación Ionizante , Telómero/ultraestructura
12.
Colloids Surf B Biointerfaces ; 142: 408-416, 2016 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-26977977

RESUMEN

The synthesis, characterization and assessment of biological behavior of innovative negatively charged functionalized gold nanoparticles is herein reported, for potential applications in the field of radiotherapy and drug delivery. Gold nanoparticles (AuNPs) functionalized with two capping agents, i.e., the 3-mercapto-1-propansulfonate (3-MPS) and 1-ß-thio-D-glucose (TG), have been on purpose synthesized and fully characterized. Advanced characterization techniques including X-Ray Photoelectron Spectroscopy (XPS) were applied to probe the chemical structure of the synthesized nanomaterials. Z-potential and Dynamic Light Scattering measurements allowed assessing the nanodimension, dispersity, surface charge and stability of AuNPs. Transmission Electron Microscopy (TEM) and Flame Atomic Absorption Spectroscopy (FAAS) were applied to the "in vitro" HSG cell model, to investigate the nanoparticles-cells interaction and to evaluate the internalization efficiency, whereas short term cytotoxicity and long term cell killing were evaluated by means of MTT and SRB assays, respectively. In conclusion, in order to increase the amount of gold atoms inside the cell we have optimized the synthesis for a new kind of biocompatible and very stable negatively charged TG-functionalized nanoparticles, with diameters in a range that maximize the uptake in cells (i.e., ∼15nm). Such particles are very promising for radiotherapy and drug delivery application.


Asunto(s)
Portadores de Fármacos , Glucosa/análogos & derivados , Oro/química , Nanopartículas del Metal/química , Compuestos de Sulfhidrilo/química , Transporte Biológico , Supervivencia Celular/efectos de los fármacos , Glucosa/química , Oro/farmacología , Células HeLa , Humanos , Tamaño de la Partícula , Rodaminas , Electricidad Estática , Propiedades de Superficie
13.
Cell Death Dis ; 7: e2308, 2016 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-27468685

RESUMEN

Proteins involved in DNA double-strand break (DSB) repair localize within the promyelocytic leukemia nuclear bodies (PML-NBs), whose disruption is at the root of the acute promyelocytic leukemia (APL) pathogenesis. All-trans-retinoic acid (RA) treatment induces PML-RARα degradation, restores PML-NB functions, and causes terminal cell differentiation of APL blasts. However, the precise role of the APL-associated PML-RARα oncoprotein and PML-NB integrity in the DSB response in APL leukemogenesis and tumor suppression is still lacking. Primary leukemia blasts isolated from APL patients showed high phosphorylation levels of H2AX (γ-H2AX), an initial DSBs sensor. By addressing the consequences of ionizing radiation (IR)-induced DSB response in primary APL blasts and RA-responsive and -resistant myeloid cell lines carrying endogenous or ectopically expressed PML-RARα, before and after treatment with RA, we found that the disruption of PML-NBs is associated with delayed DSB response, as revealed by the impaired kinetic of disappearance of γ-H2AX and 53BP1 foci and activation of ATM and of its substrates H2AX, NBN, and CHK2. The disruption of PML-NB integrity by PML-RARα also affects the IR-induced DSB response in a preleukemic mouse model of APL in vivo. We propose the oncoprotein-dependent PML-NB disruption and DDR impairment as relevant early events in APL tumorigenesis.


Asunto(s)
Núcleo Celular/metabolismo , ADN/metabolismo , Regulación Leucémica de la Expresión Génica , Células Precursoras de Granulocitos/metabolismo , Leucemia Promielocítica Aguda/genética , Proteínas de Fusión Oncogénica/genética , Animales , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/efectos de los fármacos , Núcleo Celular/efectos de la radiación , Núcleo Celular/ultraestructura , Quinasa de Punto de Control 2/genética , Quinasa de Punto de Control 2/metabolismo , ADN/genética , Roturas del ADN de Doble Cadena/efectos de la radiación , Modelos Animales de Enfermedad , Rayos gamma , Células Precursoras de Granulocitos/efectos de los fármacos , Células Precursoras de Granulocitos/patología , Células Precursoras de Granulocitos/efectos de la radiación , Histonas/genética , Histonas/metabolismo , Humanos , Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/patología , Ratones , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Transducción de Señal , Tretinoina/farmacología , Proteína 1 de Unión al Supresor Tumoral P53/genética , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo
14.
DNA Repair (Amst) ; 25: 104-15, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25467559

RESUMEN

G-quadruplex (G4) interacting agents are a class of ligands that can bind to and stabilise secondary structures located in genomic G-rich regions such as telomeres. Stabilisation of G4 leads to telomere architecture disruption with a consequent detrimental effect on cell proliferation, which makes these agents good candidates for chemotherapeutic purposes. RHPS4 is one of the most effective and well-studied G4 ligands with a very high specificity for telomeric G4. In this work, we tested the in vitro efficacy of RHPS4 in astrocytoma cell lines, and we evaluated whether RHPS4 can act as a radiosensitising agent by destabilising telomeres. In the first part of the study, the response to RHPS4 was investigated in four human astrocytoma cell lines (U251MG, U87MG, T67 and T70) and in two normal primary fibroblast strains (AG01522 and MRC5). Cell growth reduction, histone H2AX phosphorylation and telomere-induced dysfunctional foci (TIF) formation were markedly higher in astrocytoma cells than in normal fibroblasts, despite the absence of telomere shortening. In the second part of the study, the combined effect of submicromolar concentrations of RHPS4 and X-rays was assessed in the U251MG glioblastoma radioresistant cell line. Long-term growth curves, cell cycle analysis and cell survival experiments, clearly showed the synergistic effect of the combined treatment. Interestingly the effect was greater in cells bearing a higher number of dysfunctional telomeres. DNA double-strand breaks rejoining after irradiation revealed delayed repair kinetics in cells pre-treated with the drug and a synergistic increase in chromosome-type exchanges and telomeric fusions. These findings provide the first evidence that exposure to RHPS4 radiosensitizes astrocytoma cells, suggesting the potential for future therapeutic applications.


Asunto(s)
Acridinas/uso terapéutico , G-Cuádruplex/efectos de los fármacos , Glioblastoma/tratamiento farmacológico , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Telomerasa/antagonistas & inhibidores , Telómero/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Reparación del ADN/genética , Histonas/metabolismo , Humanos , Fosforilación , Tolerancia a Radiación/efectos de los fármacos , Tolerancia a Radiación/genética
15.
Artículo en Inglés | MEDLINE | ID: mdl-26520373

RESUMEN

High-resolution multicolour banding FISH (mBAND) and multiplex FISH (mFISH) were used to analyse the aberrations of chromosome 1 in irradiated-AG01522 human primary fibroblasts. The cells were exposed to 1Gy of a panel of radiation of different qualities, such as X-rays, low-energy protons (28keV/µm), helium-ions (62keV/µm) and carbon-ions (96 and 252keV/µm). mBAND and mFISH analysis in calyculin-A G2-condensed chromosome spreads allowed us to detect intra- and interchromosome aberrations involving chromosome 1, including simple and complex-type exchanges, inversions (both para- and pericentric ones), deletions and rings. The data indicate that the induction of chromosomal exchanges was influenced by both Linear energy transfer (LET) and particle types. Moreover, the complex-to-simple exchanges ratio (C-ratio) and interchromosome to intrachromosome exchanges ratio (F-ratio) were evaluated by mFISH and mBAND techniques, respectively. Our results indicate that the C-ratio is a more reliable marker of radiation quality, with values that increased linearly in an LET-dependent manner. In addition, by means of mBAND analysis, the distribution of radiation-induced breakpoints along chromosome 1 was analyzed and compared with the expected distributions of the breaks. The expected values were calculated assuming a random distribution of the breakpoints. The data indicate that, irrespective of the radiation that was used, the breakpoints were non-randomly distributed along chromosome 1. In particular, breaks in the pericentromeric region were encountered at a higher frequency than expected. A deeper analysis revealed that breaks were not located in the constitutive heterochromatin (G-bands 1p11/1q11 and 1q12), but rather in a region comprised between 1p11.2 and 1p22.1, which includes G-light and G-dark bands.


Asunto(s)
Aberraciones Cromosómicas , Bandeo Cromosómico/métodos , Puntos de Rotura del Cromosoma , Cromosomas Humanos Par 1/efectos de la radiación , Fibroblastos/efectos de la radiación , Hibridación Fluorescente in Situ/métodos , Carbono , Línea Celular , Cromosomas Humanos Par 1/genética , Iones Pesados , Helio , Humanos , Transferencia Lineal de Energía , Protones , Rayos X
16.
Environ Mol Mutagen ; 56(4): 412-7, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25263003

RESUMEN

The amphibian micronucleus test has been widely used during the last 30 years to test the genotoxic properties of several chemicals and as a tool for ecogenotoxic monitoring. The vast majority of these studies were performed on peripheral blood of urodelan larvae and anuran tadpoles and to a lesser extent adults were also used. In this study, we developed protocols for measuring micronuclei in adult shed skin cells and larval gill cells of the Italian crested newt (Triturus carnifex). Amphibians were collected from ponds in two protected areas in Italy that differed in their radon content. Twenty-three adult newts and 31 larvae were captured from the radon-rich pond, while 20 adults and 27 larvae were taken from the radon-free site. The animals were brought to the laboratory and the micronucleus test was performed on peripheral blood and shed skins taken from the adults and on larval gills. Samples from the radon-rich site showed micronucleus frequencies higher than those from the radon-free site and the difference was statistically significant in gill cells (P < 0.00001). Moreover, the larval gills seem to be more sensitive than the adult tissues. This method represents an easy (and noninvasive in the case of the shed skin) application of the micronucleus assay that can be useful for environmental studies in situ.


Asunto(s)
Monitoreo del Ambiente/métodos , Pruebas de Micronúcleos/métodos , Triturus/genética , Animales , Agua Dulce/análisis , Branquias/efectos de los fármacos , Italia , Larva/efectos de los fármacos , Metales Pesados/análisis , Estanques , Contaminantes Químicos del Agua/análisis
17.
Radiat Prot Dosimetry ; 166(1-4): 302-5, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25897136

RESUMEN

The present investigation aimed to characterise the shape of dose-response curve and determining the frequency distribution of various aberration types as a function of dose and radiation quality in AG01522 primary human fibroblasts in the 0.1- to 1-Gy dose range. For this purpose, the cells were irradiated with 7.7 and 28.5 keV µm(-1) low-energy protons, 62 keV µm(-1 4)He(2+) ions (LNL Radiobiology facility) or X rays and samples collected for 24-colour mFISH analysis. X rays and 7.7 keV µm(-1) protons displayed a quadratic dose-response curve solely for total and simple exchanges, whereas for high-linear energy transfer radiations, a linear dose-response curve was observed for all the aberration categories, with the exception of complex exchanges.


Asunto(s)
Aberraciones Cromosómicas/efectos de la radiación , Fibroblastos/efectos de la radiación , Iones Pesados , Hibridación Fluorescente in Situ/métodos , Transferencia Lineal de Energía/efectos de la radiación , Protones , Humanos , Dosis de Radiación , Rayos X
18.
Cell Prolif ; 35(2): 93-104, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11952644

RESUMEN

The relationship between G2-phase checkpoint activation, cytoplasmic cyclin-B1 accumulation and nuclear phosphorylation of p34CDC2 was studied in Nijmegen breakage syndrome cells treated with DNA damaging agents. Experiments were performed on lymphoblastoid cell lines from four Nijmegen breakage syndrome patients with different mutations, as well as on cells from an ataxia telangiectasia patient. Lymphoblastoid cell lines were irradiated with 0.50-2 Gy X-rays and the percentage of G2-phase accumulated cells was evaluated by means of flow cytometry in samples that were harvested 24 h later. The G2-checkpoint activation was analysed by scoring the mitotic index at 2 and 4 h after treatment with 0.5 and 1 Gy X-rays and treatment with the DNA double-strand break inducer calicheamicin-gamma1. Cytoplasmic accumulation of cyclin-B1 was evaluated by means of fluorescence immunostaining or Western blotting, in cells harvested shortly after irradiation with 1 and 2 Gy. The extent of tyrosine 15-phosphorylated p34CDC2 was assessed in the nuclear fractions. Nijmegen breakage syndrome cells showed suboptimal G2-phase checkpoint activation respect to normal cells and were greatly different from ataxia telangiectasia cells. Increased cytoplasmic cyclin-B1 accumulation was detected by both immunofluorescence and immunoblot in normal as well as in Nijmegen breakage syndrome cells. Furthermore, nuclear p34CDC2. phosphorylation was detected at a higher level in Nijmegen breakage syndrome than in ataxia telangiectasia cells. In conclusion, our data do not suggest that failure to activate checkpoints plays a major role in the radiosensitivity of Nijmegen breakage syndrome cells.


Asunto(s)
Rotura Cromosómica , Fase G2/efectos de la radiación , Linfocitos/citología , Linfocitos/efectos de la radiación , Western Blotting , Proteína Quinasa CDC2/metabolismo , Línea Celular , Ciclina B/análisis , Ciclina B1 , Citoplasma/química , Citoplasma/enzimología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Fase G2/fisiología , Humanos , Linfocitos/enzimología
19.
Radiat Res ; 156(3): 225-31, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11500131

RESUMEN

To study the origin of micronuclei induced in human primary fibroblasts by low-energy protons (7.7 and 28.5 keV/microm) and X rays, we have developed a combined antikinetochore-antibody (CREST) and FISH staining with pancentromeric probes. This technique allowed us to analyze the integrity of the kinetochore and centromeric DNA structures and to assess their role in induced aneuploidy. The effect of LET on radiation-induced chromosome nondisjunction was studied in binucleated cells with centromeric-specific DNA probes for chromosomes 7 and 11. Our results indicate that, though more than 90% of radiation-induced micronuclei were CREST(-)/FISH(-), 28.5 keV/microm protons and X rays were also able to induce statistically significant increases in the number of micronuclei that were CREST(-)/FISH(+) and CREST(+)/FISH(+), respectively. One interpretation of these results could be that the protons induced chromosome loss by kinetochore detachment or by breakage in the centromeric DNA region, whereas X rays induced aneuploidy through a non-DNA damage mechanism. Nondisjunction appears to be a far more important mechanism leading to radiation-induced aneuploidy. Irrespective of the higher frequency of micronuclei induced by 28.5 keV/microm protons, the frequency of chromosome loss was markedly higher for X rays than for 28.5 keV/microm protons, strengthening the hypothesis that non-DNA targets, such as components of the mitotic spindle apparatus, may be involved in aberrations in chromosome segregation after X irradiation.


Asunto(s)
Aneuploidia , Centrómero , Deleción Cromosómica , No Disyunción Genética , Protones , Rayos X , Línea Celular , Fibroblastos/efectos de la radiación , Fibroblastos/ultraestructura , Humanos , Hibridación Fluorescente in Situ
20.
Environ Mol Mutagen ; 32(2): 130-8, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9776175

RESUMEN

Molecular cytogenetic methods were applied to investigate the effect of the occupational exposure to low concentrations of benzene and petroleum fuels on genomic stability. Twelve male gasoline station attendants (average benzene exposure of 0.32 mg/m3 as 8h TWA) and 12 age- and smoking-matched unexposed controls were selected for the study. The incidence of hyperploidy and polyploidy in peripheral lymphocytes was evaluated through in situ hybridization of interphase cells, harvested 24 hr after stimulation, with centromeric probes of chromosomes 7, 11, 18, and X. For half of the subjects, metaphases harvested 24 hr later were analyzed. The incidence of chromosome loss in vitro was determined in cytokinesis-blocked cells, harvested at 66 hr, through the hybridization of micronuclei with a pancentromeric probe. Ten thousand chromosomes (more than 200 metaphases equivalent) and 2,000 binucleated cells/person were scored for hyperploidy and micronucleus analysis, respectively. The results obtained did not show any exposure-related excess of hyperploidy or micronucleus formation. Conversely, the age of the subjects was significantly correlated with several markers of genomic instability, such as the incidence of chromosome X and chromosome 18 hyperploidy, total hyperploidy and polyploidy, and close to statistical significance with chromosome loss. Smoking habits did not appear to contribute significantly to the effects measured. The parallel analysis of hyperploidy and polyploidy in interphase nuclei in 24-hr cultures and in metaphase cells harvested 24 hr later showed basically similar incidences of aneuploid cells, indicating that no significant selection against hyperploid and polyploid types occurred during the first cell cycle in vitro.


Asunto(s)
Contaminantes Ocupacionales del Aire/efectos adversos , Aneuploidia , Benceno/efectos adversos , Cromosomas Humanos/efectos de los fármacos , Gasolina/efectos adversos , Exposición Profesional , Contaminantes Ocupacionales del Aire/análisis , Contaminantes Ocupacionales del Aire/farmacología , Contaminantes Ocupacionales del Aire/orina , Benceno/análisis , Benceno/farmacología , Ciclo Celular , Células Cultivadas , Deleción Cromosómica , Gasolina/toxicidad , Humanos , Hibridación Fluorescente in Situ , Interfase , Masculino , Metafase , Pruebas de Micronúcleos , Ciudad de Roma , Muestreo , Fumar/epidemiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA