Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Immunol Rev ; 296(1): 169-190, 2020 07.
Article in English | MEDLINE | ID: mdl-32594569

ABSTRACT

Therapeutic vaccination offers great promise as an intervention for a diversity of infectious and non-infectious conditions. Given that most chronic health conditions are thought to have an immune component, vaccination can at least in principle be proposed as a therapeutic strategy. Understanding the nature of protective immunity is of vital importance, and the progress made in recent years in defining the nature of pathological and protective immunity for a range of diseases has provided an impetus to devise strategies to promote such responses in a targeted manner. However, in many cases, limited progress has been made in clinical adoption of such approaches. This in part results from a lack of safe and effective vaccine adjuvants that can be used to promote protective immunity and/or reduce deleterious immune responses. Although somewhat simplistic, it is possible to divide therapeutic vaccine approaches into those targeting conditions where antibody responses can mediate protection and those where the principal focus is the promotion of effector and memory cellular immunity or the reduction of damaging cellular immune responses as in the case of autoimmune diseases. Clearly, in all cases of antigen-specific immunotherapy, the identification of protective antigens is a vital first step. There are many challenges to developing therapeutic vaccines beyond those associated with prophylactic diseases including the ongoing immune responses in patients, patient heterogeneity, and diversity in the type and stage of disease. If reproducible biomarkers can be defined, these could allow earlier diagnosis and intervention and likely increase therapeutic vaccine efficacy. Current immunomodulatory approaches related to adoptive cell transfers or passive antibody therapy are showing great promise, but these are outside the scope of this review which will focus on the potential for adjuvanted therapeutic active vaccination strategies.


Subject(s)
Adjuvants, Immunologic , Immunomodulation , Vaccination , Vaccines/immunology , Vaccines/therapeutic use , Animals , Antibody Formation/immunology , Autoimmunity , Disease Management , Humans , Immunity, Cellular , Immunity, Humoral , Molecular Targeted Therapy , Treatment Outcome , Vaccination/methods , Vaccines/administration & dosage
2.
Int J Mol Sci ; 20(20)2019 Oct 22.
Article in English | MEDLINE | ID: mdl-31652604

ABSTRACT

Chronic exposure to low-dose ionizing radiation is associated with an increased risk of cardiovascular disease. Alteration in energy metabolism has been suggested to contribute to radiation-induced heart pathology, mitochondrial dysfunction being a hallmark of this disease. The goal of this study was to investigate the regulatory role of acetylation in heart mitochondria in the long-term response to chronic radiation. ApoE-deficient C57Bl/6J mice were exposed to low-dose-rate (20 mGy/day) gamma radiation for 300 days, resulting in a cumulative total body dose of 6.0 Gy. Heart mitochondria were isolated and analyzed using quantitative proteomics. Radiation-induced proteome and acetylome alterations were further validated using immunoblotting, enzyme activity assays, and ELISA. In total, 71 proteins showed peptides with a changed acetylation status following irradiation. The great majority (94%) of the hyperacetylated proteins were involved in the TCA cycle, fatty acid oxidation, oxidative stress response and sirtuin pathway. The elevated acetylation patterns coincided with reduced activity of mitochondrial sirtuins, increased the level of Acetyl-CoA, and were accompanied by inactivation of major cardiac metabolic regulators PGC-1 alpha and PPAR alpha. These observations suggest that the changes in mitochondrial acetylation after irradiation is associated with impairment of heart metabolism. We propose a novel mechanism involved in the development of late cardiac damage following chronic irradiation.


Subject(s)
Mitochondrial Proteins/metabolism , Myocytes, Cardiac/metabolism , Protein Processing, Post-Translational , Sirtuins/genetics , Whole-Body Irradiation/adverse effects , Acetylation , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Down-Regulation , Female , Mice , Mice, Inbred C57BL , Mitochondria, Heart/metabolism , Mitochondria, Heart/radiation effects , Mitochondrial Proteins/radiation effects , Myocytes, Cardiac/radiation effects , PPAR alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism
3.
Int J Radiat Biol ; 93(9): 920-928, 2017 09.
Article in English | MEDLINE | ID: mdl-28697312

ABSTRACT

PURPOSE: Epidemiological studies indicate that radiation doses as low as 0.5 Gy increase the risk of cardiovascular disease decades after the exposure. The aim of the present study was to investigate whether this radiation dose causes late molecular alterations in endothelial cells that could support the population-based data. MATERIALS AND METHODS: Human coronary artery endothelial cells were irradiated at 0.5 Gy (X-ray) and radiation-induced changes in the proteome were investigated after different time intervals (1, 7 and 14 d) using ICPL technology. Key changes identified by proteomics and bioinformatics were validated by immunoblotting and ELISA. RESULTS: The radiation-induced alteration of the endothelial proteome was characterized by sustained perturbation of Rho GDP-dissociation inhibitor (RhoGDI) and nitric oxide (NO) signalling pathways. At later time-points, this was accompanied by reduced proteasome activity, enhanced protein carbonylation indicating augmented oxidative stress, and senescence. CONCLUSIONS: These molecular changes are indicative of long-term premature endothelial dysfunction and provide a mechanistic framework to the epidemiological data showing increased risk of cardiovascular disease at 0.5 Gy.


Subject(s)
Endothelial Cells/physiology , Endothelial Cells/radiation effects , Gene Expression Regulation/radiation effects , Nitric Oxide/metabolism , Proteome/metabolism , Signal Transduction/radiation effects , rho-Specific Guanine Nucleotide Dissociation Inhibitors/metabolism , Cells, Cultured , Cellular Senescence/physiology , Cellular Senescence/radiation effects , Gene Expression Regulation/physiology , Humans , Radiation Dosage , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , X-Rays
4.
PLoS One ; 12(4): e0175386, 2017.
Article in English | MEDLINE | ID: mdl-28384359

ABSTRACT

We propose a stochastic model for use in epidemiological analysis, describing the age-dependent development of atherosclerosis with adequate simplification. The model features the uptake of monocytes into the arterial wall, their proliferation and transition into foam cells. The number of foam cells is assumed to determine the health risk for clinically relevant events such as stroke. In a simulation study, the model was checked against the age-dependent prevalence of atherosclerotic lesions. Next, the model was applied to incidence of atherosclerotic stroke in the cohort of male workers from the Mayak nuclear facility in the Southern Urals. It describes the data as well as standard epidemiological models. Based on goodness-of-fit criteria the risk factors smoking, hypertension and radiation exposure were tested for their effect on disease development. Hypertension was identified to affect disease progression mainly in the late stage of atherosclerosis. Fitting mechanistic models to incidence data allows to integrate biological evidence on disease progression into epidemiological studies. The mechanistic approach adds to an understanding of pathogenic processes, whereas standard epidemiological methods mainly explore the statistical association between risk factors and disease outcome. Due to a more comprehensive scientific foundation, risk estimates from mechanistic models can be deemed more reliable. To the best of our knowledge, such models are applied to epidemiological data on cardiovascular diseases for the first time.


Subject(s)
Atherosclerosis/epidemiology , Nuclear Power Plants , Occupational Diseases/epidemiology , Atherosclerosis/physiopathology , Cohort Studies , Disease Progression , Female , Humans , Male , Occupational Exposure , Russia/epidemiology , Stochastic Processes
5.
Int J Radiat Biol ; 93(2): 156-164, 2017 02.
Article in English | MEDLINE | ID: mdl-27653672

ABSTRACT

PURPOSE: Ionizing radiation induces cardiovascular disease, the endothelium being the main target. The exact mechanism of the damage is unclear but the involvement of multiple signaling pathways is probable. Reversible lysine acetylation is a posttranslational protein modification that regulates activity across a broad range of signaling pathways. The aim of this study was to determine if a low radiation dose results in acetylome alteration in endothelial cells. MATERIALS AND METHODS: Human coronary artery endothelial cell line was irradiated with Cs-137 gamma-rays (0.5 Gy) and proteomics analysis was performed using enriched acetylated peptides and all peptides. Data were validated using immunoblotting, deacetylase activity assay, and RhoA activity assay. RESULTS: Nearly a hundred proteins were found to have an altered acetylation status 24 h after irradiation, primarily due to an overall decrease in acetylation. The expression of specific deacetylases was significantly increased, coinciding with an enhancement in global deacetylase activity. Proteins changed in their acetylation status belonged to several pathways including protein synthesis, cytoskeleton-related processes, protein folding and calcium signaling. The predicted changes in the RhoA/actin cytoskeleton pathway were validated by immunoassay. CONCLUSIONS: This study shows that protein acetylation is an important mediator of radiation responses in human cardiac coronary endothelial cells. Increased knowledge of the endothelial response to radiation is crucial for the development of normal tissue-sparing modalities during radiation therapy.


Subject(s)
Coronary Vessels/physiology , Coronary Vessels/radiation effects , Endothelial Cells/physiology , Endothelial Cells/radiation effects , Histone Deacetylases/biosynthesis , Proteome/biosynthesis , Acetylation/radiation effects , Cell Line , Coronary Vessels/cytology , Dose-Response Relationship, Radiation , Gene Expression Regulation, Enzymologic/physiology , Gene Expression Regulation, Enzymologic/radiation effects , Humans , Radiation Dosage
6.
Oncotarget ; 7(44): 71817-71832, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27708245

ABSTRACT

Accruing data indicate that radiation-induced consequences resemble pathologies of neurodegenerative diseases such as Alzheimer´s. The aim of this study was to elucidate the effect on hippocampus of chronic low-dose-rate radiation exposure (1 mGy/day or 20 mGy/day) given over 300 days with cumulative doses of 0.3 Gy and 6.0 Gy, respectively. ApoE deficient mutant C57Bl/6 mouse was used as an Alzheimer´s model. Using mass spectrometry, a marked alteration in the phosphoproteome was found at both dose rates. The radiation-induced changes in the phosphoproteome were associated with the control of synaptic plasticity, calcium-dependent signalling and brain metabolism. An inhibition of CREB signalling was found at both dose rates whereas Rac1-Cofilin signalling was found activated only at the lower dose rate. Similarly, the reduction in the number of activated microglia in the molecular layer of hippocampus that paralleled with reduced levels of TNFα expression and lipid peroxidation was significant only at the lower dose rate. Adult neurogenesis, investigated by Ki67, GFAP and NeuN staining, and cell death (activated caspase-3) were not influenced at any dose or dose rate. This study shows that several molecular targets induced by chronic low-dose-rate radiation overlap with those of Alzheimer´s pathology. It may suggest that ionising radiation functions as a contributing risk factor to this neurodegenerative disease.


Subject(s)
Alzheimer Disease/etiology , Apolipoproteins E/physiology , Hippocampus/radiation effects , Proteome , Alzheimer Disease/metabolism , Animals , Cyclic AMP Response Element-Binding Protein/physiology , Disease Models, Animal , Female , Hippocampus/metabolism , Lipid Peroxidation/radiation effects , Mice , Mice, Inbred C57BL , Neurogenesis/radiation effects , Neuronal Plasticity/radiation effects , Phosphorylation , Radiation Dosage , Radiation, Ionizing , Signal Transduction
7.
Oncotarget ; 7(26): 40461-40480, 2016 Jun 28.
Article in English | MEDLINE | ID: mdl-27250028

ABSTRACT

A combined transcriptome and proteome analysis of mouse radiation-induced AMLs using two primary AMLs, cell lines from these primaries, another cell line and its in vivo passage is reported. Compared to haematopoietic progenitor and stem cells (HPSC), over 5000 transcriptome alterations were identified, 2600 present in all materials. 55 and 3 alterations were detected in the proteomes of the cell lines and primary/in vivo passage material respectively, with one common to all materials. In cell lines, approximately 50% of the transcriptome changes are related to adaptation to cell culture, and in the proteome this proportion was higher. An AML 'signature' of 17 genes/proteins commonly deregulated in primary AMLs and cell lines compared to HPSCs was identified and validated using human AML transcriptome data. This also distinguishes primary AMLs from cell lines and includes proteins such as Coronin 1, pontin/RUVBL1 and Myeloperoxidase commonly implicated in human AML. C-Myc was identified as having a key role in radiation leukaemogenesis. These data identify novel candidates relevant to mouse radiation AML pathogenesis, and confirm that pathways of leukaemogenesis in the mouse and human share substantial commonality.


Subject(s)
Hematopoietic Stem Cells/metabolism , Leukemia, Myeloid, Acute/metabolism , Neoplasms, Radiation-Induced/metabolism , Proteome , Transcriptome , ATPases Associated with Diverse Cellular Activities/metabolism , Algorithms , Animals , Carrier Proteins/metabolism , Cell Line, Tumor , DNA Helicases/metabolism , Gene Expression Regulation, Leukemic , Humans , Leukemia, Myeloid, Acute/pathology , Mice , Microfilament Proteins/metabolism , Neoplasms, Radiation-Induced/pathology , Peroxidase/metabolism , Signal Transduction
8.
J Radiat Res ; 56(4): 623-32, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25840449

ABSTRACT

Reversible lysine acetylation is a highly regulated post-translational protein modification that is known to regulate several signaling pathways. However, little is known about the radiation-induced changes in the acetylome. In this study, we analyzed the acute post-translational acetylation changes in primary human cardiac microvascular endothelial cells 4 h after a gamma radiation dose of 2 Gy. The acetylated peptides were enriched using anti-acetyl conjugated agarose beads. A total of 54 proteins were found to be altered in their acetylation status, 23 of which were deacetylated and 31 acetylated. Pathway analyses showed three protein categories particularly affected by radiation-induced changes in the acetylation status: the proteins involved in the translation process, the proteins of stress response, and mitochondrial proteins. The activation of the canonical and non-canonical Wnt signaling pathways affecting actin cytoskeleton signaling and cell cycle progression was predicted. The protein expression levels of two nicotinamide adenine dinucleotide (NAD+)-dependent deacetylases, sirtuin 1 and sirtuin 3, were significantly but transiently upregulated 4 but not 24 h after irradiation. The status of the p53 protein, a target of sirtuin 1, was found to be rapidly stabilized by acetylation after radiation exposure. These findings indicate that post-translational modification of proteins by acetylation and deacetylation is essentially affecting the radiation response of the endothelium.


Subject(s)
Acetylation/radiation effects , Endothelial Cells/physiology , Endothelial Cells/radiation effects , Histone Deacetylases/metabolism , Lysine/metabolism , Proteome/metabolism , Cells, Cultured , Dose-Response Relationship, Radiation , Humans , Microvessels/cytology , Microvessels/physiology , Microvessels/radiation effects , Myocardium/cytology , Radiation Dosage , Radiation, Ionizing
9.
J Proteome Res ; 14(1): 366-73, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25299163

ABSTRACT

Tens of thousands of people are being exposed daily to environmental low-dose gamma radiation. Epidemiological data indicate that such low radiation doses may negatively affect liver function and result in the development of liver disease. However, the biological mechanisms behind these adverse effects are unknown. The aim of this study was to investigate radiation-induced damage in the liver after low radiation doses. Neonatal male NMRI mice were exposed to total body irradiation on postnatal day 10 using acute single doses ranging from 0.02 to 1.0 Gy. Early (1 day) and late (7 months) changes in the liver proteome were tracked using isotope-coded protein label technology and quantitative mass spectrometry. Our data indicate that low and moderate radiation doses induce an immediate inhibition of the glycolysis pathway and pyruvate dehydrogenase availability in the liver. Furthermore, they lead to significant long-term alterations in lipid metabolism and increased liver inflammation accompanying inactivation of the transcription factor peroxisome proliferator-activated receptor alpha. This study contributes to the understanding of the potential risk of liver damage in populations environmentally exposed to ionizing radiation.


Subject(s)
Animals, Newborn/metabolism , Liver/metabolism , Proteome/metabolism , Proteome/radiation effects , Whole-Body Irradiation/adverse effects , Animals , Computational Biology , Dose-Response Relationship, Radiation , Immunoblotting , Lipid Metabolism/radiation effects , Liver/radiation effects , Male , Mice , Proteomics , Radiation, Ionizing , Tandem Mass Spectrometry
10.
PLoS One ; 8(10): e77484, 2013.
Article in English | MEDLINE | ID: mdl-24147004

ABSTRACT

BACKGROUND: microRNAs (miRNAs) are non-coding RNAs that alter the stability and translation efficiency of messenger RNAs. Ionizing radiation (IR) induces rapid and selective changes in miRNA expression. Depletion of the miRNA processing enzymes Dicer or Ago2 reduces the capacity of cells to survive radiation exposure. Elucidation of critical radiation-regulated miRNAs and their target proteins offers a promising approach to identify new targets to increase the therapeutic effectiveness of the radiation treatment of cancer. PRINCIPAL FINDINGS: Expression of miR-525-3p is rapidly up-regulated in response to radiation. Manipulation of miR-525-3p expression in irradiated cells confirmed that this miRNA mediates the radiosensitivity of a variety of non-transformed (RPE, HUVEC) and tumor-derived cell lines (HeLa, U2-Os, EA.hy926) cell lines. Thus, anti-miR-525-3p mediated inhibition of the increase in miR-525-3p elevated radiosensitivity, while overexpression of precursor miR-525-3p conferred radioresistance. Using a proteomic approach we identified 21 radiation-regulated proteins, of which 14 were found to be candidate targets for miR-525-3p-mediated repression. Luciferase reporter assays confirmed that nine of these were indeed direct targets of miR-525-3p repression. Individual analysis of these direct targets by RNAi-mediated knockdown established that ARRB1, TXN1 and HSPA9 are essential miR-525-3p-dependent regulators of radiation sensitivity. CONCLUSION: The transient up-regulation of miR-525-3p, and the resultant repression of its direct targets ARRB1, TXN1 and HSPA9, is required for cell survival following irradiation. The conserved function of miR-525-3p across several cell types makes this microRNA pathway a promising target for modifying the efficacy of radiotherapy.


Subject(s)
Arrestins/genetics , Gene Expression Regulation , MicroRNAs/genetics , Thioredoxins/genetics , Arrestins/metabolism , Base Pairing , Base Sequence , Cell Line , Cell Survival/genetics , Cell Survival/radiation effects , Dose-Response Relationship, Radiation , Gene Expression Profiling , Gene Expression Regulation/radiation effects , Gene Regulatory Networks , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Humans , MicroRNAs/chemistry , MicroRNAs/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Molecular Sequence Annotation , Proteome , Proteomics , RNA Interference , Radiation Tolerance/genetics , Signal Transduction , Thioredoxins/chemistry , Thioredoxins/metabolism , beta-Arrestin 1 , beta-Arrestins
11.
Radiat Environ Biophys ; 52(4): 451-61, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23880982

ABSTRACT

Epidemiological studies establish that children and young adults are especially susceptible to radiation-induced cardiovascular disease (CVD). The biological mechanisms behind the elevated CVD risk following exposure at young age remain unknown. The present study aims to elucidate the long-term effects of ionizing radiation by studying the murine cardiac proteome after exposure to low and moderate radiation doses. NMRI mice received single doses of total body (60)Co gamma-irradiation on postnatal day 10 and were sacrificed 7 months later. Changes in cardiac protein expression were quantified using isotope-coded protein label and tandem mass spectrometry. We identified 32, 31, 66, and 34 significantly deregulated proteins after doses of 0.02, 0.1, 0.5, and 1.0 Gy, respectively. The four doses shared 9 deregulated proteins. Bioinformatics analysis showed that most of the deregulated proteins belonged to a limited set of biological categories, including metabolic processes, inflammatory response, and cytoskeletal structure. The transcription factor peroxisome proliferator-activated receptor alpha was predicted as a common upstream regulator of several deregulated proteins. This study indicates that both adaptive and maladaptive responses to the initial radiation damage persist well into adulthood. It will contribute to the understanding of the long-term consequences of radiation-induced injury and developmental alterations in the neonatal heart.


Subject(s)
Heart/radiation effects , Myocardium/metabolism , Proteomics , Animals , Animals, Newborn , Dose-Response Relationship, Radiation , Gene Ontology , Male , Mice , Protein Interaction Maps/radiation effects , Signal Transduction/radiation effects , Time Factors , Whole-Body Irradiation
12.
J Proteome Res ; 12(6): 2700-14, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23560462

ABSTRACT

Radiation exposure of the thorax is associated with a markedly increased risk of cardiac morbidity and mortality with a latency period of decades. Although many studies have confirmed the damaging effect of ionizing radiation on the myocardium and cardiac endothelial structure and function, the molecular mechanism behind this damage is not yet elucidated. Peroxisome proliferator-activated receptor alpha (PPAR alpha), a transcriptional regulator of lipid metabolism in heart tissue, has recently received great attention in the development of cardiovascular disease. The goal of this study was to investigate radiation-induced cardiac damage in general and the role of PPAR alpha in this process in particular. C57BL/6 mice received local heart irradiation with X-ray doses of 8 and 16 gray (Gy) at the age of 8 weeks. The mice were sacrificed 16 weeks later. Radiation-induced changes in the cardiac proteome were quantified using the Isotope Coded Protein Label (ICPL) method followed by mass spectrometry and software analysis. Significant alterations were observed in proteins involved in lipid metabolism and oxidative phosphorylation. Ionizing radiation markedly changed the phosphorylation and ubiquitination status of PPAR alpha. This was reflected as decreased expression of its target genes involved in energy metabolism and mitochondrial respiratory chain confirming the proteomics data. This study suggests that persistent alteration of cardiac metabolism due to impaired PPAR alpha activity contributes to the heart pathology after radiation.


Subject(s)
Heart/radiation effects , Lipid Metabolism/radiation effects , Mitochondria, Heart/radiation effects , PPAR alpha/genetics , Animals , Gene Expression/radiation effects , Heart/physiopathology , Male , Mice , Mice, Inbred C57BL , Mitochondria, Heart/metabolism , Oxidative Phosphorylation/radiation effects , PPAR alpha/metabolism , Protein Interaction Mapping , Proteomics , Signal Transduction , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry , X-Rays
13.
Radiother Oncol ; 106(3): 404-10, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23522698

ABSTRACT

BACKGROUND AND PURPOSE: Radiotherapy of thoracic and chest-wall tumours increases the long-term risk of radiation-induced heart disease. The aim of this study was to investigate the long-term effect of local heart irradiation on cardiac mitochondria. METHODS: C57BL/6 and atherosclerosis-prone ApoE(-/-) mice received local heart irradiation with a single X-ray dose of 2 Gy. To investigate the low-dose effect, C57BL/6 mice also received a single heart dose of 0.2 Gy. Functional and proteomic alterations of cardiac mitochondria were evaluated after 40 weeks, compared to age-matched controls. RESULTS: The respiratory capacity of irradiated C57BL/6 cardiac mitochondria was significantly reduced at 40 weeks. In parallel, protein carbonylation was increased, suggesting enhanced oxidative stress. Considerable alterations were found in the levels of proteins of mitochondria-associated cytoskeleton, respiratory chain, ion transport and lipid metabolism. Radiation induced similar but less pronounced effects in the mitochondrial proteome of ApoE(-/-) mice. In ApoE(-/-), no significant change was observed in mitochondrial respiration or protein carbonylation. The dose of 0.2 Gy had no significant effects on cardiac mitochondria. CONCLUSION: This study suggests that ionising radiation causes non-transient alterations in cardiac mitochondria, resulting in oxidative stress that may ultimately lead to malfunctioning of the heart muscle.


Subject(s)
Mitochondria, Heart/radiation effects , Animals , Apolipoproteins E/physiology , Heart/radiation effects , Mice , Mice, Inbred C57BL , Mitochondria, Heart/metabolism , Oxidative Stress , Protein Carbonylation , Radiation, Ionizing , Time Factors
14.
Proteomics ; 13(7): 1096-107, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23349028

ABSTRACT

Chronic low-dose ionizing radiation induces cardiovascular disease in human populations but the mechanism is largely unknown. We suggested that chronic radiation exposure may induce endothelial cell senescence that is associated with vascular damage in vivo. We investigated whether chronic radiation exposure is causing a change in the onset of senescence in endothelial cells in vitro. Indeed, when exposed to continuous low-dose rate gamma radiation (4.1 mGy/h), primary human umbilical vein endothelial cells (HUVECs) initiated senescence much earlier than the nonirradiated control cells. We investigated the changes in the protein expression of HUVECs before and during the onset of radiation-induced senescence. Cellular proteins were quantified using isotope-coded protein label technology after 1, 3, and 6 weeks of radiation exposure. Several senescence-related biological pathways were influenced by radiation, including cytoskeletal organization, cell-cell communication and adhesion, and inflammation. Immunoblot analysis showed an activation of the p53/p21 pathway corresponding to the progressing senescence. Our data suggest that chronic radiation-induced DNA damage and oxidative stress result in induction of p53/p21 pathway that inhibits the replicative potential of HUVECs and leads to premature senescence. This study contributes to the understanding of the increased risk of cardiovascular diseases seen in populations exposed to chronic low-dose irradiation.


Subject(s)
Cellular Senescence/radiation effects , Gamma Rays , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/radiation effects , Proteomics/methods , Cell Proliferation/radiation effects , Cell Shape/radiation effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Dose-Response Relationship, Radiation , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Immunoblotting , Metabolic Networks and Pathways/radiation effects , Proteome/metabolism , Reproducibility of Results , Signal Transduction/radiation effects , Tumor Suppressor Protein p53/metabolism
15.
Radiat Environ Biophys ; 52(1): 87-98, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23138885

ABSTRACT

High doses of ionising radiation significantly increase the risk of cardiovascular disease (CVD), the vascular endothelium representing one of the main targets. Whether radiation doses lower than 500 mGy induce cardiovascular damage is controversial. The aim of this study was to investigate radiation-induced expression changes on protein and microRNA (miRNA) level in primary human coronary artery endothelial cells after a single 200 mGy radiation dose (Co-60). Using a multiplex gel-based proteomics technology (2D-DIGE), we identified 28 deregulated proteins showing more than ±1.5-fold expression change in comparison with non-exposed cells. A great majority of the proteins showed up-regulation. Bioinformatics analysis indicated "cellular assembly and organisation, cellular function and maintenance and molecular transport" as the most significant radiation-responsive network. Caspase-3, a central regulator of this network, was confirmed to be up-regulated using immunoblotting. We also analysed radiation-induced alterations in the level of six miRNAs known to play a role either in CVD or in radiation response. The expression of miR-21 and miR-146b showed significant radiation-induced deregulation. Using miRNA target prediction, three proteins found differentially expressed in this study were identified as putative candidates for miR-21 regulation. A negative correlation was observed between miR-21 levels and the predicted target proteins, desmoglein 1, phosphoglucomutase and target of Myb protein. This study shows for the first time that a low-dose exposure has a significant impact on miRNA expression that is directly related to protein expression alterations. The data presented here may facilitate the discovery of low-dose biomarkers of radiation-induced cardiovascular damage.


Subject(s)
Endothelial Cells/metabolism , Gamma Rays , MicroRNAs/metabolism , Aged , Cells, Cultured , Coronary Vessels/cytology , Female , Humans , Proteomics
16.
J Proteomics ; 75(8): 2384-95, 2012 Apr 18.
Article in English | MEDLINE | ID: mdl-22387116

ABSTRACT

Qualitative proteome profiling of formalin-fixed, paraffin-embedded (FFPE) tissue is advancing the field of clinical proteomics. However, quantitative proteome analysis of FFPE tissue is hampered by the lack of an efficient labelling method. The usage of conventional protein labelling on FFPE tissue has turned out to be inefficient. Classical labelling targets lysine residues that are blocked by the formalin treatment. The aim of this study was to establish a quantitative proteomics analysis of FFPE tissue by combining the label-free approach with optimised protein extraction and separation conditions. As a model system we used FFPE heart tissue of control and exposed C57BL/6 mice after total body irradiation using a gamma ray dose of 3 gray. We identified 32 deregulated proteins (p≤0.05) in irradiated hearts 24h after the exposure. The proteomics data were further evaluated and validated by bioinformatics and immunoblotting investigation. In good agreement with our previous results using fresh-frozen tissue, the analysis indicated radiation-induced alterations in three main biological pathways: respiratory chain, lipid metabolism and pyruvate metabolism. The label-free approach enables the quantitative measurement of radiation-induced alterations in FFPE tissue and facilitates retrospective biomarker identification using clinical archives.


Subject(s)
Heart/radiation effects , Mitochondria, Heart/radiation effects , Muscle Proteins/metabolism , Myocardium/metabolism , Radiation, Ionizing , Animals , Fixatives/pharmacology , Formaldehyde/pharmacology , Heart/drug effects , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/radiation effects , Metabolome/physiology , Mice , Mice, Inbred C57BL , Mitochondria, Heart/chemistry , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Mitochondrial Proteins/analysis , Mitochondrial Proteins/drug effects , Mitochondrial Proteins/radiation effects , Muscle Proteins/analysis , Muscle Proteins/drug effects , Muscle Proteins/radiation effects , Myocardium/chemistry , Paraffin Embedding/methods , Staining and Labeling/methods
17.
J Proteomics ; 75(8): 2319-30, 2012 Apr 18.
Article in English | MEDLINE | ID: mdl-22370162

ABSTRACT

Epidemiological data show that ionising radiation increases the risk of cardiovascular disease. The endothelium is one of the main targets of radiation-induced damage. Rapid radiation-induced alterations in the biological processes were investigated after exposure to a clinically relevant radiation dose (2.5 Gy gamma radiation). The changes in protein expression were determined using the human endothelial cell line EA.hy926 as a model. Two complementary proteomic approaches, SILAC (Stable Isotope Labelling with Amino acids in Cell culture) and 2D-DIGE (Two Dimensional Difference-in-Gel-Electrophoresis) were used. The proteomes of the endothelial cells were analysed 4h and 24h after irradiation. Differentially expressed proteins were identified and quantified by MALDI-TOF/TOF and LTQ Orbitrap tandem mass spectrometry. The deregulated proteins were mainly categorised in four key pathways: (i) glycolysis/gluconeogenesis and synthesis/degradation of ketone bodies, (ii) oxidative phosphorylation, (iii) Rho-mediated cell motility and (iv) non-homologous end joining. We suggest that these alterations facilitate the repair processes needed to overcome the stress caused by irradiation and are indicative of the vascular damage leading to radiation-induced cardio- and cerebrovascular impairment.


Subject(s)
Endothelial Cells/chemistry , Endothelial Cells/radiation effects , Isotope Labeling/methods , Proteome/analysis , Two-Dimensional Difference Gel Electrophoresis/methods , Amino Acids , Cell Culture Techniques , Cell Proliferation/radiation effects , Cells, Cultured , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/chemistry , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/radiation effects , Humans , Metabolic Networks and Pathways/physiology , Metabolic Networks and Pathways/radiation effects , Models, Biological , Proteome/metabolism , Proteomics/methods , Radiation Injuries/metabolism , Signal Transduction/radiation effects , Validation Studies as Topic
18.
PLoS One ; 6(12): e27811, 2011.
Article in English | MEDLINE | ID: mdl-22174747

ABSTRACT

BACKGROUND: Radiation therapy treatment of breast cancer, Hodgkin's disease or childhood cancers expose the heart to high local radiation doses, causing an increased risk of cardiovascular disease in the survivors decades after the treatment. The mechanisms that underlie the radiation damage remain poorly understood so far. Previous data show that impairment of mitochondrial oxidative metabolism is directly linked to the development of cardiovascular disease. METHODOLOGY/PRINCIPAL FINDINGS: In this study, the radiation-induced in vivo effects on cardiac mitochondrial proteome and function were investigated. C57BL/6N mice were exposed to local irradiation of the heart with doses of 0.2 Gy or 2 Gy (X-ray, 200 kV) at the age of eight weeks, the control mice were sham-irradiated. After four weeks the cardiac mitochondria were isolated and tested for proteomic and functional alterations. Two complementary proteomics approaches using both peptide and protein quantification strategies showed radiation-induced deregulation of 25 proteins in total. Three main biological categories were affected: the oxidative phophorylation, the pyruvate metabolism, and the cytoskeletal structure. The mitochondria exposed to high-dose irradiation showed functional impairment reflected as partial deactivation of Complex I (32%) and Complex III (11%), decreased succinate-driven respiratory capacity (13%), increased level of reactive oxygen species and enhanced oxidation of mitochondrial proteins. The changes in the pyruvate metabolism and structural proteins were seen with both low and high radiation doses. CONCLUSION/SIGNIFICANCE: This is the first study showing the biological alterations in the murine heart mitochondria several weeks after the exposure to low- and high-dose of ionizing radiation. Our results show that doses, equivalent to a single dose in radiotherapy, cause long-lasting changes in mitochondrial oxidative metabolism and mitochondria-associated cytoskeleton. This prompts us to propose that these first pathological changes lead to an increased risk of cardiovascular disease after radiation exposure.


Subject(s)
Mitochondria/metabolism , Mitochondria/radiation effects , Myocardium/metabolism , Signal Transduction/radiation effects , Animals , Computational Biology , Cytochromes c1/metabolism , Dose-Response Relationship, Radiation , Electron Transport Complex I/metabolism , Electron Transport Complex III/metabolism , Electrophoresis, Gel, Two-Dimensional , Immunoblotting , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/ultrastructure , Mitochondrial Proteins/metabolism , Oxidation-Reduction/drug effects , Oxidation-Reduction/radiation effects , Phosphorylation/drug effects , Phosphorylation/radiation effects , Proteomics , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Reproducibility of Results , Signal Transduction/drug effects , Succinic Acid/pharmacology , X-Rays
19.
Proteomics ; 11(16): 3299-311, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21751382

ABSTRACT

Accidental nuclear scenarios lead to environmental contamination of unknown level. Immediate radiation-induced biological responses that trigger processes leading to adverse health effects decades later are not well understood. A comprehensive proteomic analysis provides a promising means to identify and quantify the initial damage after radiation exposure. Early changes in the cardiac tissue of C57BL/6 mice exposed to total body irradiation were studied, using a dose relevant to both intentional and accidental exposure (3 Gy gamma ray). Heart tissue protein lysates were analyzed 5 and 24 h after the exposure using isotope-coded protein labeling (ICPL) and 2-dimensional difference-in-gel-electrophoresis (2-D DIGE) proteomics approaches. The differentially expressed proteins were identified by LC-ESI-MS-MS. Both techniques showed similar functional groups of proteins to be involved in the initial injury. Pathway analyses indicated that total body irradiation immediately induced biological responses such as inflammation, antioxidative defense, and reorganization of structural proteins. Mitochondrial proteins represented the protein class most sensitive to ionizing radiation. The proteins involved in the initial damage processes map to several functional categories involving cardiotoxicity. This prompts us to propose that these early changes are indicative of the processes that lead to an increased risk of cardiovascular disease after radiation exposure.


Subject(s)
Gamma Rays/adverse effects , Heart/radiation effects , Proteins/analysis , Proteome/radiation effects , Animals , Blotting, Western , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Chromatography, Liquid , Electrophoresis, Gel, Two-Dimensional , Lipid Peroxidation/radiation effects , Mass Spectrometry , Mice , Mice, Inbred C57BL , Oxidative Stress , Protein Carbonylation/radiation effects , Protein Interaction Mapping , Proteins/metabolism , Proteome/analysis , Proteomics , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/pathology , Reproducibility of Results , Ventricular Remodeling/radiation effects , Whole-Body Irradiation
20.
Radiat Environ Biophys ; 50(1): 155-66, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21104263

ABSTRACT

High doses of ionising radiation damage the heart by an as yet unknown mechanism. A concern for radiological protection is the recent epidemiological data indicating that doses as low as 100-500 mGy may induce cardiac damage. The aim of this study was to identify potential molecular targets and/or mechanisms involved in the pathogenesis of low-dose radiation-induced cardiovascular disease. The vascular endothelium plays a pivotal role in the regulation of cardiac function and is therefore a potential target tissue. We report here that low-dose radiation induced rapid and time-dependent changes in the cytoplasmic proteome of the human endothelial cell line EA.hy926. The proteomes were investigated at 4 and 24 h after irradiation at two different dose rates (Co-60 gamma ray total dose 200 mGy; 20 mGy/min and 190 mGy/min) using 2D-DIGE technology. Differentially expressed proteins were identified, after in-gel trypsin digestion, by MALDI-TOF/TOF tandem mass spectrometry, and peptide mass fingerprint analyses. We identified 15 significantly differentially expressed proteins, of which 10 were up-regulated and 5 down-regulated, with more than ±1.5-fold difference compared with unexposed cells. Pathways influenced by the low-dose exposures included the Ran and RhoA pathways, fatty acid metabolism and stress response.


Subject(s)
Endothelial Cells/diagnostic imaging , Endothelial Cells/metabolism , Proteome/metabolism , Apoptosis/radiation effects , Cell Proliferation/radiation effects , Cytosol/metabolism , Cytosol/radiation effects , Dose-Response Relationship, Drug , Endothelial Cells/cytology , Gene Expression Profiling , Humans , Proteomics , Radiography , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL