Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
Commun Biol ; 6(1): 234, 2023 03 02.
Article in English | MEDLINE | ID: mdl-36864251

ABSTRACT

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.


Subject(s)
Shelterin Complex , Telomere , ADP-Ribosylation , DNA Damage , DNA Helicases , Telomere/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism
2.
J Exp Clin Cancer Res ; 38(1): 311, 2019 Jul 16.
Article in English | MEDLINE | ID: mdl-31311580

ABSTRACT

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.


Subject(s)
Acridines/administration & dosage , Brain Neoplasms/therapy , Glioblastoma/therapy , Neoplastic Stem Cells/drug effects , Radiation-Sensitizing Agents/administration & dosage , Acridines/pharmacology , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Checkpoint Kinase 1/genetics , Checkpoint Kinase 1/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Mice , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Radiation-Sensitizing Agents/pharmacology , Xenograft Model Antitumor Assays
3.
Mutat Res Rev Mutat Res ; 773: 204-219, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28927529

ABSTRACT

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.


Subject(s)
Neoplasms/drug therapy , Neoplasms/radiotherapy , Radiation, Ionizing , Telomerase/metabolism , Telomere/metabolism , Aminobenzoates/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Disease Models, Animal , Homeostasis/drug effects , Homeostasis/radiation effects , Humans , Naphthalenes/pharmacology , Oligonucleotides/pharmacology , RNA Interference , Radiation-Sensitizing Agents/pharmacology
4.
Cell Death Dis ; 7: e2308, 2016 07 28.
Article in English | MEDLINE | ID: mdl-27468685

ABSTRACT

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.


Subject(s)
Cell Nucleus/metabolism , DNA/metabolism , Gene Expression Regulation, Leukemic , Granulocyte Precursor Cells/metabolism , Leukemia, Promyelocytic, Acute/genetics , Oncogene Proteins, Fusion/genetics , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Nucleus/drug effects , Cell Nucleus/radiation effects , Cell Nucleus/ultrastructure , Checkpoint Kinase 2/genetics , Checkpoint Kinase 2/metabolism , DNA/genetics , DNA Breaks, Double-Stranded/radiation effects , Disease Models, Animal , Gamma Rays , Granulocyte Precursor Cells/drug effects , Granulocyte Precursor Cells/pathology , Granulocyte Precursor Cells/radiation effects , Histones/genetics , Histones/metabolism , Humans , Leukemia, Promyelocytic, Acute/metabolism , Leukemia, Promyelocytic, Acute/pathology , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , Signal Transduction , Tretinoin/pharmacology , Tumor Suppressor p53-Binding Protein 1/genetics , Tumor Suppressor p53-Binding Protein 1/metabolism
5.
Colloids Surf B Biointerfaces ; 142: 408-416, 2016 Jun 01.
Article in English | MEDLINE | ID: mdl-26977977

ABSTRACT

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.


Subject(s)
Drug Carriers , Glucose/analogs & derivatives , Gold/chemistry , Metal Nanoparticles/chemistry , Sulfhydryl Compounds/chemistry , Biological Transport , Cell Survival/drug effects , Glucose/chemistry , Gold/pharmacology , HeLa Cells , Humans , Particle Size , Rhodamines , Static Electricity , Surface Properties
6.
Article in English | MEDLINE | ID: mdl-26520373

ABSTRACT

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.


Subject(s)
Chromosome Aberrations , Chromosome Banding/methods , Chromosome Breakpoints , Chromosomes, Human, Pair 1/radiation effects , Fibroblasts/radiation effects , In Situ Hybridization, Fluorescence/methods , Carbon , Cell Line , Chromosomes, Human, Pair 1/genetics , Heavy Ions , Helium , Humans , Linear Energy Transfer , Protons , X-Rays
7.
Radiat Prot Dosimetry ; 166(1-4): 302-5, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25897136

ABSTRACT

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.


Subject(s)
Chromosome Aberrations/radiation effects , Fibroblasts/radiation effects , Heavy Ions , In Situ Hybridization, Fluorescence/methods , Linear Energy Transfer/radiation effects , Protons , Humans , Radiation Dosage , X-Rays
8.
DNA Repair (Amst) ; 25: 104-15, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25467559

ABSTRACT

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.


Subject(s)
Acridines/therapeutic use , G-Quadruplexes/drug effects , Glioblastoma/drug therapy , Radiation-Sensitizing Agents/therapeutic use , Telomerase/antagonists & inhibitors , Telomere/drug effects , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Repair/drug effects , DNA Repair/genetics , Histones/metabolism , Humans , Phosphorylation , Radiation Tolerance/drug effects , Radiation Tolerance/genetics
9.
Environ Mol Mutagen ; 56(4): 412-7, 2015 May.
Article in English | MEDLINE | ID: mdl-25263003

ABSTRACT

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.


Subject(s)
Environmental Monitoring/methods , Micronucleus Tests/methods , Triturus/genetics , Animals , Fresh Water/analysis , Gills/drug effects , Italy , Larva/drug effects , Metals, Heavy/analysis , Ponds , Water Pollutants, Chemical/analysis
10.
Front Genet ; 4: 141, 2013.
Article in English | MEDLINE | ID: mdl-23908663

ABSTRACT

This work is about the setup of an in vitro system to report low-dose of X-rays as measured as cytogenetic damage. Q- and multicolor FISH (m-FISH), for telomere length and chromosome instability analysis, respectively, were compared to evaluate their sensitivity in the low-dose range in human primary fibroblasts. No telomere length modulation was observed up to 1 Gy in cycling fibroblasts, though reported for high doses, by that frustrating the purpose of using it as a low-exposure marker. To date the m-FISH is the best setup for the assessment of the chromosome structural damage: it allows stable and instable aberrations to be detected all over the karyotype. Stable ones such as balanced translocations, are not eliminated due to cell-cycle as unstable ones, so they are considered transmissible markers for retrospective dosimetry. The induction of chromosome damage showed a clear dependence on dose delivered; unstable aberrations were demonstrated after doses of 0.1 Gy, and stable aberrations after doses higher than 0.5 Gy. Summarizing, q-FISH is unfit to report low exposures while m-FISH provides better results: unstable aberrations are sensible short-term reporters, while stable ones long report exposures but with a higher induction threshold.

11.
Mutat Res ; 756(1-2): 86-94, 2013 Aug 30.
Article in English | MEDLINE | ID: mdl-23721903

ABSTRACT

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.


Subject(s)
Chromosome Aberrations , DNA, Mitochondrial/metabolism , Fibroblasts/radiation effects , Mitochondria/radiation effects , Zalcitabine/pharmacology , Antimetabolites/pharmacology , Cell Survival/radiation effects , Cells, Cultured , DNA Damage/genetics , DNA Damage/radiation effects , DNA Repair/genetics , DNA Repair/radiation effects , DNA, Mitochondrial/drug effects , DNA, Mitochondrial/radiation effects , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , In Situ Hybridization, Fluorescence , Linear Energy Transfer , Membrane Potential, Mitochondrial/radiation effects , Mitochondria/drug effects , Mitochondria/genetics , Reactive Oxygen Species/metabolism , Telomere/genetics , X-Rays
12.
Mutat Res ; 740(1-2): 13-20, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23220250

ABSTRACT

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.


Subject(s)
Lymphocytes/radiation effects , Radiation Tolerance/genetics , Telomere Shortening/radiation effects , Cell Line , Humans , Telomerase/metabolism , Telomere Homeostasis/radiation effects
13.
Apoptosis ; 16(9): 940-9, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21671007

ABSTRACT

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.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Carcinoma, Non-Small-Cell Lung/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Stilbenes/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Bcl-2-Like Protein 11 , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Caspase 3/metabolism , Cell Line, Tumor/drug effects , Cytosol/metabolism , Gene Expression Regulation, Neoplastic , Humans , Immunoprecipitation , M Phase Cell Cycle Checkpoints , Microscopy, Confocal , Microtubules/drug effects , Microtubules/metabolism , Mitochondria/metabolism , Protein Transport , RNA, Small Interfering/metabolism , Signal Transduction , Transfection
14.
Radiat Prot Dosimetry ; 143(2-4): 274-8, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21156783

ABSTRACT

Telomeres are the end of linear chromosomes, responsible for chromosome stability and cell viability. It is well known that radiations are able to induce chromosome instability but it has not yet been investigated whether telomere structure is affected by the radiation exposure and if radiations with different quality act in a different way on telomeres. The effect of radiations with different quality on telomere structure and chromosome instability was analysed in human primary fibroblasts exposed to X rays or low-energy protons (28.5 keV µm(-1)). Telomere length was evaluated at different harvesting times from 24 h up to 360 h (15 days), whereas chromosome instability was evaluated in terms of sister chromatid exchanges (SCEs) (48 h from irradiation) and chromosome painting (360 h from irradiation). Results indicated a delayed telomere lengthening 360 h after X-ray treatment, whereas protons were able to induce such a lengthening shortly from irradiation as well as at longer harvesting times. Data obtained from chromosome instability analysis indicated an increase of SCE frequency only after proton irradiation, but, on the contrary, at the longer harvesting time chromosome painting analysis displayed a higher frequency of aberrations after X-ray treatment, suggesting a role of selective process against highly damaged cells.


Subject(s)
Chromosomal Instability/genetics , Chromosomal Instability/radiation effects , Telomere/genetics , Telomere/radiation effects , Cell Line , Dose-Response Relationship, Radiation , Humans , Photons , Radiation Dosage , Telomere/ultrastructure , X-Rays
15.
Cell Mol Biol (Noisy-le-grand) ; 56 Suppl: OL1299-317, 2010 Jun 10.
Article in English | MEDLINE | ID: mdl-20937217

ABSTRACT

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.


Subject(s)
Liver Neoplasms/metabolism , MicroRNAs/metabolism , Proteome/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism , Cell Line, Tumor , GRB2 Adaptor Protein/metabolism , Gene Expression Profiling , Hepatocytes/metabolism , Humans , Signal Transduction
16.
Radiat Res ; 174(5): 539-49, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20726710

ABSTRACT

It is well established that high-LET radiations efficiently induce chromosome aberrations. However, data on the effect of protons on telomere maintenance, as involved in genomic stability, are scarce and contradictory. Here we demonstrate that high-LET protons induce telomere lengthening in human primary fibroblasts and that this elongation does not involve the telomerase enzyme, supporting the hypothesis that high-LET radiations are able to activate a telomerase-independent mechanism. In tumor cells that lack telomerase, one or more non-telomerase mechanisms for telomere maintenance are present, which are termed alternative lengthening of telomeres (ALT). Since ALT cells are characterized by recombinational events at telomeres, known as telomeric-sister chromatid exchanges (T-SCE), and colocalization of telomeres and premyelocytic leukemia protein (PML), we analyzed both T-SCE and PML. Our results show that high-LET protons induce a 2.5-fold increase of T-SCE and a colocalization of PML protein and telomeric DNA. Furthermore, our data show that the ALT pathway can be activated in human primary cells after induction of severe DNA damage. Thus, since telomeres are known to be involved in chromosome maintenance, the present work may contribute in the elucidation of the mechanism by which ionizing radiation induces genomic instability.


Subject(s)
Fibroblasts/metabolism , Fibroblasts/radiation effects , Linear Energy Transfer , Telomere/metabolism , Telomere/radiation effects , DNA Damage , Dose-Response Relationship, Drug , Genome, Human/genetics , Humans , In Situ Hybridization, Fluorescence , Nuclear Proteins/metabolism , Promyelocytic Leukemia Protein , Protein Transport/radiation effects , Sister Chromatid Exchange/radiation effects , Telomerase/metabolism , Telomere/genetics , Time Factors , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism
17.
Radiat Res ; 170(3): 345-52, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18763866

ABSTRACT

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.


Subject(s)
Cell Cycle Proteins/metabolism , Chromatids/genetics , Chromatids/radiation effects , Chromosomal Proteins, Non-Histone/metabolism , Chromosome Aberrations , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Radiation Injuries/genetics , Signal Transduction/physiology , Tumor Suppressor Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins , Cells, Cultured , Dose-Response Relationship, Radiation , Humans , Radiation Dosage , Signal Transduction/radiation effects
18.
Amino Acids ; 34(3): 403-12, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17610126

ABSTRACT

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.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Nucleus/genetics , Etoposide/pharmacology , Gene Expression Regulation, Enzymologic , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Zea mays/enzymology , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Doxycycline/pharmacology , Etoposide/therapeutic use , Gene Expression Regulation, Enzymologic/drug effects , Humans , Oxidoreductases Acting on CH-NH Group Donors/genetics , Polyamines/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Zea mays/genetics , Polyamine Oxidase
19.
Eur J Med Genet ; 50(3): 176-87, 2007.
Article in English | MEDLINE | ID: mdl-17395558

ABSTRACT

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.


Subject(s)
Craniofacial Abnormalities/genetics , Microcephaly/genetics , Nijmegen Breakage Syndrome/genetics , Radiation Tolerance/genetics , Telomere/genetics , Cell Cycle Proteins/genetics , Chromosomes, Human/radiation effects , DNA Repair/genetics , Female , Humans , Male , Nijmegen Breakage Syndrome/diagnosis , Nuclear Proteins/genetics , Phenotype , Telomere/ultrastructure
20.
Genome Dyn ; 1: 191-205, 2006.
Article in English | MEDLINE | ID: mdl-18724061

ABSTRACT

Nijmegen breakage syndrome (NBS) is a rare recessive genetic disorder, characterized by bird-like facial appearance, early growth retardation, congenital microcephaly, immunodeficiency and high frequency of malignancies. NBS belongs to the so-called chromosome instability syndromes; in fact, NBS cells display spontaneous chromosomal aberrations and are hypersensitive to DNA double-strand break-inducing agents, such as ionizing radiations. NBS1, the gene underlying the disease, is located on human chromosome 8q21. The disease appears to be prevalent in the Eastern and Central European population where more than 90% of patients are homozygous for the founder mutation 657del5 leading to a truncated variant of the protein. NBS1 forms a multimeric complex with MRE11/RAD50 nuclease at the C-terminus and retains or recruits them at the vicinity of sites of DNA damage by direct binding to histone H2AX, which is phosphorylated by PI3-kinase family, such as ATM, in response to DNA damage. Thereafter, the NBS1-complex proceeds to rejoin double-strand breaks predominantly by homologous recombination repair in vertebrates. NBS cells also show to be defective in the activation of intra-S phase checkpoint. We review here some cellular and molecular aspects of NBS, which might contribute to the clinical symptoms of the disease.


Subject(s)
Cell Cycle Proteins/genetics , Nijmegen Breakage Syndrome/genetics , Nuclear Proteins/genetics , Cell Cycle , Chromosomal Instability , DNA Damage , DNA Repair , Founder Effect , Histones/metabolism , Homozygote , Humans , Models, Biological , Models, Genetic , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , White People
SELECTION OF CITATIONS
SEARCH DETAIL
...