Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 47(17): e100, 2019 09 26.
Article in English | MEDLINE | ID: mdl-31318974

ABSTRACT

The majority of the proteins involved in processing of DNA double-strand breaks (DSBs) accumulate at the damage sites. Real-time imaging and analysis of these processes, triggered by the so-called microirradiation using UV lasers or heavy particle beams, yielded valuable insights into the underlying DSB repair mechanisms. To study the temporal organization of DSB repair responses triggered by a more clinically-relevant DNA damaging agent, we developed a system coined X-ray multi-microbeam microscope (XM3), capable of simultaneous high dose-rate (micro)irradiation of large numbers of cells with ultra-soft X-rays and imaging of the ensuing cellular responses. Using this setup, we analyzed the changes in real-time kinetics of MRE11, MDC1, RNF8, RNF168 and 53BP1-proteins involved in the signaling axis of mammalian DSB repair-in response to X-ray and UV laser-induced DNA damage, in non-cancerous and cancer cells and in the presence or absence of a photosensitizer. Our results reveal, for the first time, the kinetics of DSB signaling triggered by X-ray microirradiation and establish XM3 as a powerful platform for real-time analysis of cellular DSB repair responses.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , DNA-Binding Proteins/metabolism , Time-Lapse Imaging/methods , X-Rays , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins/metabolism , Cell Line , Humans , MRE11 Homologue Protein , Microscopy, Electron, Scanning , Osteosarcoma/metabolism , Pigment Epithelium of Eye/metabolism , Signal Transduction , Tumor Suppressor p53-Binding Protein 1/metabolism , Ubiquitin-Protein Ligases/metabolism , Ultraviolet Rays
2.
Biol Chem ; 399(12): 1409-1419, 2018 11 27.
Article in English | MEDLINE | ID: mdl-30138105

ABSTRACT

Histatins are histidine-rich peptides present in the saliva of humans and higher primates and have been implicated in the protection of the oral cavity. Histatin 1 is one of the most abundant histatins and recent reports show that it has a stimulating effect on cellular adherence, thereby suggesting a role in maintaining the quality of the epithelial barrier and stimulating mesenchymal-to-epithelial transition. Here we summarize these findings and discuss them in the context of previous reports. The recent findings also provide new insights in the physiological functions of histatin 1, which are discussed here. Furthermore, we put forward a possible role of histatin 1 in various pathologies and its potential function in clinical applications.


Subject(s)
Epithelial-Mesenchymal Transition , Histatins/metabolism , Amino Acid Sequence , Cell Adhesion , Histatins/chemistry , Histatins/genetics , Humans
3.
FASEB J ; 31(9): 3922-3933, 2017 09.
Article in English | MEDLINE | ID: mdl-28522595

ABSTRACT

Histatins are multifunctional histidine-rich peptides secreted by the salivary glands and exclusively present in the saliva of higher primates, where they play a fundamental role in the protection of the oral cavity. Our previously published results demonstrated that histatin-1 (Hst1) promotes cell-substrate adhesion in various cell types and hinted that it could also be involved in cell-cell adhesion, a process of fundamental importance to epithelial and endothelial barriers. Here we explore the effects of Hst1 on cellular barrier function. We show that Hst1 improved endothelial barrier integrity, decreased its permeability for large molecules, and prevented translocation of bacteria across epithelial cell layers. These effects are mediated by the adherens junction protein E-cadherin (E-cad) and by the tight junction protein zonula occludens 1, as Hst1 increases the levels of zonula occludens 1 and of active E-cad. Hst1 may also promote epithelial differentiation as Hst1 induced transcription of the epithelial cell differentiation marker apolipoprotein A-IV (a downstream E-cad target). In addition, Hst1 counteracted the effects of epithelial-mesenchymal transition inducers on the outgrowth of oral cancer cell spheroids, suggesting that Hst1 affects processes that are implicated in cancer progression.-Van Dijk, I. A., Ferrando, M. L., van der Wijk, A.-E., Hoebe, R. A., Nazmi, K., de Jonge, W. J., Krawczyk, P. M., Bolscher, J. G. M., Veerman, E. C. I., Stap, J. Human salivary peptide histatin-1 stimulates epithelial and endothelial cell adhesion and barrier function.


Subject(s)
Endothelial Cells/physiology , Epithelial Cells/physiology , Gene Expression Regulation/physiology , Histatins/metabolism , Cell Line , Histatins/genetics , Humans
4.
Int J Mol Sci ; 17(11)2016 Oct 26.
Article in English | MEDLINE | ID: mdl-27792189

ABSTRACT

The structural maintenance of chromosomes (SMC) protein complexes shape and regulate the structure and dynamics of chromatin, thereby controlling many chromosome-based processes such as cell cycle progression, differentiation, gene transcription and DNA repair. The SMC5/6 complex is previously described to promote DNA double-strand breaks (DSBs) repair by sister chromatid recombination, and found to be essential for resolving recombination intermediates during meiotic recombination. Moreover, in budding yeast, SMC5/6 provides structural organization and topological stress relief during replication in mitotically dividing cells. Despite the essential nature of the SMC5/6 complex, the versatile mechanisms by which SMC5/6 functions and its molecular regulation in mammalian cells remain poorly understood. By using a human osteosarcoma cell line (U2OS), we show that after the CRISPR-Cas9-mediated removal of the SMC5/6 subunit NSMCE2, treatment with the topoisomerase II inhibitor etoposide triggered an increased sensitivity in cells lacking NSMCE2. In contrast, NSMCE2 appeared not essential for a proper DNA damage response or cell survival after DSB induction by ionizing irradiation (IR). Interestingly, by way of immunoprecipitations (IPs) and mass spectrometry, we found that the SMC5/6 complex physically interacts with the DNA topoisomerase II α (TOP2A). We therefore propose that the SMC5/6 complex functions in resolving TOP2A-mediated DSB-repair intermediates generated during replication.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Breaks, Double-Stranded , DNA Repair , Ligases/metabolism , Antigens, Neoplasm/metabolism , CRISPR-Cas Systems , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Survival/radiation effects , Chromosomal Proteins, Non-Histone , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , Etoposide/adverse effects , Humans , Ligases/genetics , Poly-ADP-Ribose Binding Proteins , Protein Interaction Maps , Topoisomerase II Inhibitors/adverse effects
5.
Am J Physiol Heart Circ Physiol ; 311(5): H1097-H1107, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27521422

ABSTRACT

Excess catecholamine levels are suggested to be cardiotoxic and to underlie stress-induced heart failure. The cardiotoxic effects of norepinephrine and epinephrine are well recognized. However, although cardiac and circulating dopamine levels are also increased in stress cardiomyopathy patients, knowledge regarding putative toxic effects of excess dopamine levels on cardiomyocytes is scarce. We now studied the effects of elevated dopamine levels in H9c2 cardiomyoblasts. H9c2 cells were cultured and treated with dopamine (200 µM) for 6, 24, and 48 h. Subsequently, the effects on lipid accumulation, cell viability, flippase activity, reactive oxygen species (ROS) production, subcellular NADPH oxidase (NOX) protein expression, and ATP/ADP and GTP/GDP levels were analyzed. Dopamine did not result in cytotoxic effects after 6 h. However, after 24 and 48 h dopamine treatment induced a significant increase in lipid accumulation, nitrotyrosine levels, indicative of ROS production, and cell death. In addition, dopamine significantly reduced flippase activity and ATP/GTP levels, coinciding with phosphatidylserine exposure on the outer plasma membrane. Furthermore, dopamine induced a transient increase in cytoplasmic and (peri)nucleus NOX1 and NOX4 expression after 24 h that subsided after 48 h. Moreover, while dopamine induced a similar transient increase in cytoplasmic NOX2 and p47phox expression, in the (peri)nucleus this increased expression persisted for 48 h where it colocalized with ROS. Exposure of H9c2 cells to elevated dopamine levels induced lipid accumulation, oxidative stress, and a proinflammatory status of the plasma membrane. This can, in part, explain the inflammatory response in patients with stress-induced heart failure.


Subject(s)
Dopamine Agents/pharmacology , Dopamine/pharmacology , Inflammation/metabolism , Lipid Metabolism/drug effects , Myoblasts, Cardiac/drug effects , NADPH Oxidases/drug effects , Oxidative Stress/drug effects , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Caspase 3/drug effects , Caspase 3/metabolism , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Survival , Flow Cytometry , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Hydrogen-Ion Concentration , Microscopy, Electron , Microscopy, Fluorescence , Myoblasts, Cardiac/metabolism , Myoblasts, Cardiac/ultrastructure , NADH, NADPH Oxidoreductases/drug effects , NADH, NADPH Oxidoreductases/metabolism , NADPH Oxidase 1 , NADPH Oxidase 4 , NADPH Oxidases/metabolism , Nuclear Proteins/drug effects , Nuclear Proteins/metabolism , Peroxidase/drug effects , Peroxidase/metabolism , Rats , Reactive Oxygen Species/metabolism , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins , Tyrosine/analogs & derivatives , Tyrosine/drug effects , Tyrosine/metabolism
6.
J Leukoc Biol ; 100(3): 453-62, 2016 09.
Article in English | MEDLINE | ID: mdl-27034403

ABSTRACT

IL-17A, a major proinflammatory cytokine, can be produced by a variety of leukocytes, but its exact cellular source in human inflammatory diseases remains incompletely understood. IL-17A protein is abundantly found in mast cells in human tissues, such as inflamed synovium, but surprisingly, mechanistic murine studies failed to demonstrate IL-17A production by mast cells. Here, we demonstrate that primary human tissue mast cells do not produce IL-17A themselves but actively capture exogenous IL-17A through receptor-mediated endocytosis. The exogenous IL-17A is stored in intracellular granules and can subsequently be released in a bioactive form. This novel mechanism confers to mast cells the capacity to steer IL-17A-mediated tissue inflammation by the rapid release of preformed cytokine.


Subject(s)
Endocytosis/physiology , Interleukin-17/metabolism , Mast Cells/metabolism , Palatine Tonsil/metabolism , Synoviocytes/metabolism , Cells, Cultured , Humans , Interleukin-17/genetics , Mast Cells/cytology , Palatine Tonsil/cytology , Synoviocytes/cytology
7.
J Leukoc Biol ; 100(3): 513-23, 2016 09.
Article in English | MEDLINE | ID: mdl-26957213

ABSTRACT

Osteoclasts are bone-resorbing cells and targets for treating bone diseases. Previously, we reported that distinct murine osteoclast precursor subsets, such as early blasts (CD31(hi) Ly-6C(-)), myeloid blasts (CD31(+) Ly-6C(+)), and monocytes (CD31(-) Ly-6C(hi)), respond differently to the osteoclastogenesis-inducing cytokines, macrophage colony-stimulating factor, and receptor activator for nuclear factor κB ligand. It is unknown, however, how these cell types respond to the osteoclast-stimulating inflammatory cytokine interleukin 1ß. This study aims to investigate the effect of interleukin 1ß on osteoclastogenesis derived from different mouse bone marrow precursors. Early blasts, myeloid blasts, and monocytes were sorted from mouse bone marrow cells using flow cytometry. Cells were cultured on plastic or on bone slices in the presence of macrophage colony-stimulating factor and receptor activator for nuclear factor κB ligand, without or with interleukin 1ß (0.1-10 ng/ml). We found that interleukin 1ß stimulated multinucleation and bone resorption of osteoclasts derived from the 3 precursors at different rates. The most large osteoclasts (>20 nuclei) and highest level of bone resorption (16.3%) was by myeloid blast-derived osteoclasts. Interleukin 1ß particularly accelerated proliferation of early blasts and the most small osteoclasts (3-5 nuclei) formed on plastic. Life span varied among osteoclasts derived from different precursors: large osteoclasts (>2400 µm(2)) formed most rapidly (75 h) from myeloid blasts but had a short life span (30 h). Monocytes needed the longest time (95 h) for the generation of such large osteoclasts, but these cells had a longer life span (50 h). Our results indicate that the different bone marrow osteoclast precursors are differently stimulated by interleukin 1ß with respect to proliferation, multinucleation, life span, and bone resorption.


Subject(s)
Bone Marrow/pathology , Bone Resorption/pathology , Cell Proliferation/drug effects , Interleukin-1beta/pharmacology , Monocytes/pathology , Myeloid Cells/pathology , Osteoclasts/pathology , Animals , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone Resorption/chemically induced , Bone Resorption/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Gene Expression Regulation/drug effects , Male , Mice , Mice, Inbred C57BL , Monocytes/drug effects , Monocytes/metabolism , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteogenesis/drug effects
8.
Cancer Res ; 75(22): 4790-802, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26363012

ABSTRACT

Isocitrate dehydrogenase 1 (IDH1) is mutated in various types of human cancer to IDH1(R132H), a structural alteration that leads to catalysis of α-ketoglutarate to the oncometabolite D-2-hydroxyglutarate. In this study, we present evidence that small-molecule inhibitors of IDH1(R132H) that are being developed for cancer therapy may pose risks with coadministration of radiotherapy. Cancer cells heterozygous for the IDH1(R132H) mutation exhibited less IDH-mediated production of NADPH, such that after exposure to ionizing radiation (IR), there were higher levels of reactive oxygen species, DNA double-strand breaks, and cell death compared with IDH1 wild-type cells. These effects were reversed by the IDH1(R132H) inhibitor AGI-5198. Exposure of IDH1 wild-type cells to D-2-hydroxyglutarate was sufficient to reduce IDH-mediated NADPH production and increase IR sensitivity. Mechanistic investigations revealed that the radiosensitivity of heterozygous cells was independent of the well-described DNA hypermethylation phenotype in IDH1-mutated cancers. Thus, our results argue that altered oxidative stress responses are a plausible mechanism to understand the radiosensitivity of IDH1-mutated cancer cells. Further, they offer an explanation for the relatively longer survival of patients with IDH1-mutated tumors, and they imply that administration of IDH1(R132H) inhibitors in these patients may limit irradiation efficacy in this setting.


Subject(s)
Antineoplastic Agents/pharmacology , Benzeneacetamides/pharmacology , Glioblastoma/genetics , Imidazoles/pharmacology , Isocitrate Dehydrogenase/genetics , Radiation Tolerance/drug effects , Blotting, Western , Cell Line, Tumor , Chemoradiotherapy/adverse effects , DNA Methylation/drug effects , DNA Methylation/radiation effects , Enzyme Inhibitors/pharmacology , Fluorescent Antibody Technique , Gene Knock-In Techniques , Glioblastoma/pathology , Humans , In Vitro Techniques , Mutation , NADP/biosynthesis , Oxidative Stress/drug effects , Oxidative Stress/radiation effects
9.
Mol Cell Proteomics ; 14(8): 2177-93, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26041847

ABSTRACT

Tripeptidyl peptidase II (TPP2) is a serine peptidase involved in various biological processes, including antigen processing, cell growth, DNA repair, and neuropeptide mediated signaling. The underlying mechanisms of how a peptidase can influence this multitude of processes still remain unknown. We identified rapid proteomic changes in neuroblastoma cells following selective TPP2 inhibition using the known reversible inhibitor butabindide, as well as a new, more potent, and irreversible peptide phosphonate inhibitor. Our data show that TPP2 inhibition indirectly but rapidly decreases the levels of active, di-phosphorylated extracellular signal-regulated kinase 1 (ERK1) and ERK2 in the nucleus, thereby down-regulating signal transduction downstream of growth factors and mitogenic stimuli. We conclude that TPP2 mediates many important cellular functions by controlling ERK1 and ERK2 phosphorylation. For instance, we show that TPP2 inhibition of neurons in the hippocampus leads to an excessive strengthening of synapses, indicating that TPP2 activity is crucial for normal brain function.


Subject(s)
Aminopeptidases/metabolism , Cell Nucleus/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Serine Endopeptidases/metabolism , Aminopeptidases/antagonists & inhibitors , Animals , Cell Line , Cell Nucleus/drug effects , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Gene Knockdown Techniques , Gene Ontology , Humans , Inhibitory Concentration 50 , Isotope Labeling , Mice , Models, Biological , Neurites/drug effects , Neurites/metabolism , Neuronal Plasticity/drug effects , Phosphorylation/drug effects , Protein Phosphatase 2/metabolism , Proteomics , RNA, Messenger/genetics , RNA, Messenger/metabolism , SOXC Transcription Factors/genetics , SOXC Transcription Factors/metabolism
10.
FASEB J ; 29(8): 3124-32, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25903106

ABSTRACT

Histatins (Hsts) are histidine-rich peptides exclusively present in the saliva of higher primates. In this study, we explored the effects of Hsts on cell-substrate and cell-cell adhesion. Histatin (Hst)-1 caused a significant (>2-fold) increase (EC50 = 1 µM) in the ability of human adherent cells to attach and spread, even in conditions that impaired cell spreading. Other tested Hsts did not stimulate cell spreading, indicating a specific effect of Hst1. The effect of Hst1 on cell-cell adhesion was investigated by using transepithelial resistance (TER) measurements in the human cell line Caco-2, a widely used model for the epithelial layer. We found that 10 µM Hst1 caused a 20% increase in TER compared to the negative control, indicating a function for Hst1 in intercellular cell adhesion and epithelial integrity. A role for Hst1 in both cell-substrate and cell-cell adhesion is highly conceivable, because these 2 modes of adhesion are closely related via shared components and connected signaling pathways.


Subject(s)
Cell Adhesion/physiology , Histatins/metabolism , Histidine/metabolism , Peptides/metabolism , Saliva/metabolism , Salivary Proteins and Peptides/metabolism , Caco-2 Cells , Cell Line , Cell Line, Tumor , Epithelial Cells/metabolism , Humans
11.
PLoS One ; 9(4): e94931, 2014.
Article in English | MEDLINE | ID: mdl-24736599

ABSTRACT

The LIM-only protein FHL2, also known as DRAL or SLIM3, has a function in fine-tuning multiple physiological processes. FHL2 is expressed in the vessel wall in smooth muscle cells (SMCs) and endothelial cells and conflicting data have been reported on the regulatory function of FHL2 in SMC phenotype transition. At present the function of FHL2 in SMCs in vascular injury is unknown. Therefore, we studied the role of FHL2 in SMC-rich lesion formation. In response to carotid artery ligation FHL2-deficient (FHL2-KO) mice showed accelerated lesion formation with enhanced Ki67 expression compared with wild-type (WT)-mice. Consistent with these findings, cultured SMCs from FHL2-KO mice showed increased proliferation through enhanced phosphorylation of extracellular-regulated kinase-1/2 (ERK1/2) and induction of CyclinD1 expression. Overexpression of FHL2 in SMCs inhibited CyclinD1 expression and CyclinD1-knockdown blocked the enhanced proliferation of FHL2-KO SMCs. We also observed increased CyclinD1 promoter activity in FHL2-KO SMCs, which was reduced upon ERK1/2 inhibition. Furthermore, FHL2-KO SMCs showed enhanced migration compared with WT SMCs. In conclusion, FHL2 deficiency in mice results in exacerbated SMC-rich lesion formation involving increased proliferation and migration of SMCs via enhanced activation of the ERK1/2-CyclinD1 signaling pathway.


Subject(s)
LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Carotid Arteries/metabolism , Carotid Arteries/pathology , Cell Movement/genetics , Cell Proliferation , Cyclin D1/metabolism , Male , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Signal Transduction
12.
Cell Mol Biol Lett ; 19(1): 37-51, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24363165

ABSTRACT

The influence of p53 status on potentially lethal damage repair (PLDR) and DNA double-strand break (DSB) repair was studied in two isogenic human colorectal carcinoma cell lines: RKO (p53 wild-type) and RC10.1 (p53 null). They were treated with different doses of ionizing radiation, and survival and the induction of DNA-DSB were studied. PLDR was determined by using clonogenic assays and then comparing the survival of cells plated immediately with the survival of cells plated 24 h after irradiation. Doses varied from 0 to 8 Gy. Survival curves were analyzed using the linear-quadratic formula: S(D)/S(0) = exp-(αD+ßD(2)). The γ-H2AX foci assay was used to study DNA DSB kinetics. Cells were irradiated with single doses of 0, 0.5, 1 and 2 Gy. Foci levels were studied in non-irradiated control cells and 30 min and 24 h after irradiation. Irradiation was performed with gamma rays from a (137)Cs source, with a dose rate of 0.5 Gy/min. The RKO cells show higher survival rates after delayed plating than after immediate plating, while no such difference was found for the RC10.1 cells. Functional p53 seems to be a relevant characteristic regarding PLDR for cell survival. Decay of γ-H2AX foci after exposure to ionizing radiation is associated with DSB repair. More residual foci are observed in RC10.1 than in RKO, indicating that decay of γ-H2AX foci correlates with p53 functionality and PLDR in RKO cells.


Subject(s)
Cell Survival/genetics , Colorectal Neoplasms/genetics , Histones/genetics , Tumor Suppressor Protein p53/metabolism , Cell Survival/radiation effects , Colorectal Neoplasms/pathology , DNA Breaks, Double-Stranded/radiation effects , DNA Damage/radiation effects , DNA Repair/genetics , DNA Repair/radiation effects , Histones/metabolism , Humans , Radiation, Ionizing , Tumor Suppressor Protein p53/genetics
13.
Arterioscler Thromb Vasc Biol ; 33(10): 2380-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23950142

ABSTRACT

OBJECTIVE: In aortic aneurysms the arterial vessel wall is dilated because of destruction of its integrity, which may lead to lethal vessel rupture. Chronic infiltration of inflammatory cells into the vessel wall is fundamental to aneurysm pathology. We aim to limit aneurysm growth by inhibition of inflammation and reducing endothelial cell (EC) activation with immunosuppressive drug azathioprine (Aza). APPROACH AND RESULTS: Aza and its metabolite 6-mercaptopurine have anti-inflammatory effects on leukocytes. We here demonstrate that treatment of ECs with 6-mercaptopurine inhibits cell activation as illustrated by reduced expression of interleukin-12, CCL5, CCL2, and vascular cell adhesion molecule-1 and inhibition of monocyte-EC adhesion. The underlying mechanism of 6-mercaptopurine involves suppression of GTPase Rac1 activation, resulting in reduced phosphorylation of c-Jun-terminal-N-kinase and c-Jun. Subsequently, the effect of Aza was investigated in aneurysm formation in the angiotensin II aneurysm mouse model in apolipoprotein E-deficient mice. We demonstrated that Aza decreases de novo aortic aneurysm formation from an average aneurysm severity score of 2.1 (control group) to 0.6 (Aza group), and that Aza effectively delays aorta pathology in a progression experiment, resulting in a reduced severity score from 2.8 to 1.7 in Aza-treated mice. In line with the in vitro observations, Aza-treated mice showed less c-Jun-terminal-N-kinase activation in ECs and reduced leukocyte influx in the aortic wall. CONCLUSIONS: The immunosuppressive drug Aza has an anti-inflammatory effect and in ECs inhibits Rac1 and c-Jun-terminal-N-kinase activation, which may explain the protective effect of Aza in aneurysm development and, most importantly for clinical implications, aneurysm severity.


Subject(s)
Aortic Aneurysm/prevention & control , Azathioprine/pharmacology , Endothelial Cells/drug effects , Immunosuppressive Agents/pharmacology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neuropeptides/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , rac1 GTP-Binding Protein/antagonists & inhibitors , Angiotensin II , Animals , Anti-Inflammatory Agents/pharmacology , Aortic Aneurysm/chemically induced , Aortic Aneurysm/enzymology , Aortic Aneurysm/genetics , Aortic Aneurysm/immunology , Aortic Aneurysm/pathology , Aortic Rupture/enzymology , Aortic Rupture/immunology , Aortic Rupture/prevention & control , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Cell Line, Tumor , Coculture Techniques , Disease Models, Animal , Disease Progression , Endothelial Cells/enzymology , Endothelial Cells/immunology , Enzyme Activation , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Inflammation Mediators/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Mercaptopurine/metabolism , Mice , Mice, Knockout , Monocytes/drug effects , Monocytes/enzymology , Monocytes/immunology , Neuropeptides/metabolism , Phosphorylation , Signal Transduction/drug effects , rac1 GTP-Binding Protein/metabolism
14.
DNA Repair (Amst) ; 12(1): 38-45, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23237939

ABSTRACT

In S and G2 phase mammalian cells DNA double strand breaks (DSBs) can potentially be repaired by homologous recombination (HR) or non-homologous end-joining (NHEJ). Results of several studies suggest that these two mechanistically distinct repair pathways can compete for DNA ends. Because HR and NHEJ differ with respect to error susceptibility, generation of chromosome rearrangements, which are potentially carcinogenic products of DSB repair, may depend on the pathway choice. To investigate this hypothesis, the influence of HR and NHEJ inhibition on the frequencies of chromosome aberrations in G2 phase cells was investigated. SW-1573 and RKO cells were treated with mild (41 °C) hyperthermia in order to disable HR and/or NU7441/cisplatin to inactivate NHEJ and frequencies of chromosomal fragments (resulting from unrepaired DSBs) and translocations (products of erroneous DSB rejoining) were studied using premature chromosome condensation (PCC) combined with fluorescence in situ hybridization (FISH). It is shown here that temporary inhibition of HR by hyperthermia results in increased frequency of ionizing-radiation (IR)-induced chromosomal translocations and that this effect is abrogated by NU7441- or cisplatin-mediated inhibition of NHEJ. The results suggest that in the absence of HR, DSB repair is shifted to the error-prone NHEJ pathway resulting in increased frequencies of chromosomal rearrangements. These results might be of consequence for clinical cancer treatment approaches that aim at inhibition of one or more DSB repair pathways.


Subject(s)
DNA End-Joining Repair , Homologous Recombination , Hot Temperature , Recombinational DNA Repair , Animals , Cell Line, Tumor , Chromones , Cisplatin/toxicity , DNA Breaks, Double-Stranded , G2 Phase , Gamma Rays , Humans , Mice , Morpholines , Radiation Tolerance , Translocation, Genetic/drug effects , Translocation, Genetic/radiation effects
15.
Int J Hyperthermia ; 28(6): 509-17, 2012.
Article in English | MEDLINE | ID: mdl-22834701

ABSTRACT

Local hyperthermia is an effective treatment modality to augment radio- and chemotherapy-based anti-cancer treatments. Although the effect of hyperthermia is pleotropic, recent experiments revealed that homologous recombination, a pathway of DNA repair, is directly inhibited by hyperthermia. The hyperthermia-induced DNA repair deficiency is enhanced by inhibitors of the cellular heat-shock response. Taken together, these results provide the rationale for the development of novel anti-cancer therapies that combine hyperthermia-induced homologous recombination deficiency with the systemic administration of drugs that specifically affect the viability of homologous recombination deficient cells and/or inhibit the heat-shock response, to locally sensitise cancer cells to DNA damaging agents.


Subject(s)
DNA Repair-Deficiency Disorders/etiology , DNA Repair , Hyperthermia, Induced , DNA Breaks, Double-Stranded/drug effects , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/physiology , Heat-Shock Response , Homologous Recombination/physiology , Humans , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors
16.
Calcif Tissue Int ; 90(6): 515-22, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22527205

ABSTRACT

Osteoclasts are specialized multinucleated cells with the unique capacity to resorb bone. Despite insight into the various steps of the interaction of osteoclast precursors leading to osteoclast formation, surprisingly little is known about what happens with the multinucleated cell itself after it has been formed. Is fusion limited to the short period of its formation, or do osteoclasts have the capacity to change their size and number of nuclei at a later stage? To visualize these processes we analyzed osteoclasts generated in vitro with M-CSF and RANKL from mouse bone marrow and native osteoclasts isolated from rabbit bones by live cell microscopy. We show that osteoclasts fuse not only with mononuclear cells but also with other multinucleated cells. The most intriguing finding was fission of the osteoclasts. Osteoclasts were shown to have the capacity to generate functional multinucleated compartments as well as compartments that contained apoptotic nuclei. These compartments were separated from each other, each giving rise to a novel functional osteoclast or to a compartment that contained apoptotic nuclei. Our findings suggest that osteoclasts have the capacity to regulate their own population in number and function, probably to adapt quickly to changing situations.


Subject(s)
Osteoclasts/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Marrow Cells/ultrastructure , Bone Resorption/pathology , Cell Differentiation , Cells, Cultured , Mice , Mice, Inbred C57BL , Osteoclasts/cytology , RANK Ligand , Rabbits
17.
Oncol Rep ; 27(3): 769-74, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22200791

ABSTRACT

Ionizing radiation-induced foci (IRIF) of DNA repair-related proteins accumulated at DNA double-strand break (DSB) sites have been suggested to be a powerful biodosimetric tool. However, the relationship between IRIF induction and biologically relevant endpoints, such as cell death and formation of chromosome rearrangements is less clear, especially for high linear energy transfer (LET) radiation. It is thus not sufficiently established whether IRIF are valid indicators of biological effectiveness of the various radiation types. This question is more significant in light of the recent advancements in light ion-beam and radionuclide therapy. Dose-effect relationships were determined for the induction of DNA-DSBs, chromosome aberrations and reproductive cell death in cultured SW-1573 cells irradiated with γ-rays from a Cs-137 source or with α-particles from an Am-241 source. Values of relative biological effectiveness (RBE) of the high LET α-particles were derived for these effects. DNA-DSB were detected by scoring of γ-H2AX foci, chromosome aberrations by fragments and translocations using premature chromosome condensation and cell survival by colony formation. Analysis of dose-effect relations was based on the linear-quadratic model. Except for the survival curves, for other effects no significant contribution was derived of the quadratic term in the range of doses up to 2 Gy of γ-rays. Calculated RBE values derived for the linear component of dose-effect relations for γ-H2AX foci, cell reproductive death, chromosome fragments and colour junctions are 1.0±0.3, 14.7±5.1, 15.3±5.9 and 13.3±6.0, respectively. RBE values calculated at a certain biological effect level are 1, 4, 13 and 13, respectively. The RBE values derived from the LQ model are preferred as they are based on clinically relevant doses. The results show that with low LET radiation only a small fraction of the numerous DNA-DSBs yield chromosome damage and reproductive cell death. It is concluded that many of the chromosomal aberrations detected by premature chromosome condensation do not cause reproductive cell death. Furthermore, RBE values for DNA-DSB detectable by γ-H2AX foci shortly after irradiation, provide no information relevant to applications of high LET radiation in radiotherapy. The RBE values of chromosome aberrations assessed by premature chromosome condensation are close to the value for reproductive cell death. This suggests possible relevance to assess RBE values for radiotherapy with high LET ions.


Subject(s)
Alpha Particles/therapeutic use , Cell Death/radiation effects , Cell Division/radiation effects , Chromosome Aberrations/radiation effects , DNA Breaks, Double-Stranded , DNA, Neoplasm/radiation effects , Linear Energy Transfer/radiation effects , Lung Neoplasms/radiotherapy , Carcinoma, Squamous Cell/radiotherapy , Cell Line, Tumor , Cell Survival/radiation effects , DNA Repair/radiation effects , Dose-Response Relationship, Radiation , Gamma Rays/therapeutic use , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Relative Biological Effectiveness
18.
Radiat Oncol ; 6: 64, 2011 Jun 08.
Article in English | MEDLINE | ID: mdl-21651780

ABSTRACT

BACKGROUND: Various types of radiation effects in mammalian cells have been studied with the aim to predict the radiosensitivity of tumours and normal tissues, e.g. DNA double strand breaks (DSB), chromosome aberrations and cell reproductive inactivation. However, variation in correlations with clinical results has reduced general application. An additional type of information is required for the increasing application of high-LET radiation in cancer therapy: the Relative Biological Effectiveness (RBE) for effects in tumours and normal tissues. Relevant information on RBE values might be derived from studies on cells in culture. METHODS: To evaluate relationships between DNA-DSB, chromosome aberrations and the clinically most relevant effect of cell reproductive death, for ionizing radiations of different LET, dose-effect relationships were determined for the induction of these effects in cultured SW-1573 cells irradiated with gamma-rays from a Cs-137 source or with α-particles from an Am-241 source. RBE values were derived for these effects. Ionizing radiation induced foci (IRIF) of DNA repair related proteins, indicative of DSB, were assessed by counting gamma-H2AX foci. Chromosome aberration frequencies were determined by scoring fragments and translocations using premature chromosome condensation. Cell survival was measured by colony formation assay. Analysis of dose-effect relations was based on the linear-quadratic model. RESULTS: Our results show that, although both investigated radiation types induce similar numbers of IRIF per absorbed dose, only a small fraction of the DSB induced by the low-LET gamma-rays result in chromosome rearrangements and cell reproductive death, while this fraction is considerably enhanced for the high-LET alpha-radiation. Calculated RBE values derived for the linear components of dose-effect relations for gamma-H2AX foci, cell reproductive death, chromosome fragments and colour junctions are 1.0 ± 0.3, 14.7 ± 5.1, 15.3 ± 5.9 and 13.3 ± 6.0 respectively. CONCLUSIONS: These results indicate that RBE values for IRIF (DNA-DSB) induction provide little valid information on other biologically-relevant end points in cells exposed to high-LET radiations. Furthermore, the RBE values for the induction of the two types of chromosome aberrations are similar to those established for cell reproductive death. This suggests that assays of these aberrations might yield relevant information on the biological effectiveness in high-LET radiotherapy.


Subject(s)
Chromosome Aberrations , DNA Breaks, Double-Stranded/radiation effects , DNA/radiation effects , Alpha Particles , Americium/pharmacology , Carcinoma, Squamous Cell/radiotherapy , Cell Line, Tumor , Cell Proliferation/radiation effects , Cell Survival/radiation effects , Cesium Radioisotopes , Chromosomes/radiation effects , DNA Repair/radiation effects , Dose-Response Relationship, Radiation , Humans , Lung Neoplasms/radiotherapy , Radiotherapy/methods
19.
Proc Natl Acad Sci U S A ; 108(24): 9851-6, 2011 Jun 14.
Article in English | MEDLINE | ID: mdl-21555554

ABSTRACT

Defective homologous recombination (HR) DNA repair imposed by BRCA1 or BRCA2 deficiency sensitizes cells to poly (ADP-ribose) polymerase (PARP)-1 inhibition and is currently exploited in clinical treatment of HR-deficient tumors. Here we show that mild hyperthermia (41-42.5 °C) induces degradation of BRCA2 and inhibits HR. We demonstrate that hyperthermia can be used to sensitize innately HR-proficient tumor cells to PARP-1 inhibitors and that this effect can be enhanced by heat shock protein inhibition. Our results, obtained from cell lines and in vivo tumor models, enable the design of unique therapeutic strategies involving localized on-demand induction of HR deficiency, an approach that we term induced synthetic lethality.


Subject(s)
BRCA2 Protein/metabolism , Hot Temperature , Poly(ADP-ribose) Polymerases/metabolism , Recombination, Genetic/genetics , Animals , BRCA2 Protein/genetics , Benzoquinones/pharmacology , Cell Line , Cell Line, Tumor , Cells, Cultured , DNA Repair , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/radiation effects , Female , HeLa Cells , Humans , Immunoblotting , Lactams, Macrocyclic/pharmacology , Mice , Mice, Nude , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/genetics , Quinazolines/pharmacology , RNA Interference , Rats , Recombination, Genetic/drug effects , Recombination, Genetic/radiation effects , Transplantation, Heterologous , Tumor Burden/drug effects
20.
Oncol Lett ; 1(4): 765-769, 2010 Jul.
Article in English | MEDLINE | ID: mdl-22966377

ABSTRACT

Agents that enhance the effectiveness of ionizing radiation have been investigated over many decades. A relatively new group of potential radiosensitizers consists of agents that inhibit histone acetyltransferases (HATs). This study evaluated the radiosensitizing properties of the HAT inhibitor anacardic acid (AA), used at a low-toxic concentration of 100 µM in V79, SW1573 and U2OS cells. Radiation survival curves were analyzed according to the linear quadratic model. Significant radiosensitization by AA was only obtained in U2OS cells. AA significantly increased the value of the linear parameter α, but not of the quadratic parameter ß, indicating fixation of potentially lethal damage and an intact repair function of sublethal damage. The increase of the α value was also observed in SW1573 cells, but was not accompanied by a significant radiosensitization. A likely explanation for the enhancement of the α value may be an increase in the amount of lethal lesions due to the compacted chromatin structure. Despite the conflicting results of the radiosensitizing effect of AA in the three cell lines tested, the ability of AA to increase the α value suggests potential advantages for clinical application.

SELECTION OF CITATIONS
SEARCH DETAIL
...