Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Int J Mol Sci ; 22(21)2021 Oct 30.
Article in English | MEDLINE | ID: mdl-34769236

ABSTRACT

Galactic cosmic rays are primarily composed of protons (85%), helium (14%), and high charge/high energy ions (HZEs) such as 56Fe, 28Si, and 16O. HZE exposure is a major risk factor for astronauts during deep-space travel due to the possibility of HZE-induced cancer. A systems biology integrated omics approach encompassing transcriptomics, proteomics, lipidomics, and functional biochemical assays was used to identify microenvironmental changes induced by HZE exposure. C57BL/6 mice were placed into six treatment groups and received the following irradiation treatments: 600 MeV/n 56Fe (0.2 Gy), 1 GeV/n 16O (0.2 Gy), 350 MeV/n 28Si (0.2 Gy), 137Cs (1.0 Gy) gamma rays, 137Cs (3.0 Gy) gamma rays, and sham irradiation. Left liver lobes were collected at 30, 60, 120, 270, and 360 days post-irradiation. Analysis of transcriptomic and proteomic data utilizing ingenuity pathway analysis identified multiple pathways involved in mitochondrial function that were altered after HZE irradiation. Lipids also exhibited changes that were linked to mitochondrial function. Molecular assays for mitochondrial Complex I activity showed significant decreases in activity after HZE exposure. HZE-induced mitochondrial dysfunction suggests an increased risk for deep space travel. Microenvironmental and pathway analysis as performed in this research identified possible targets for countermeasures to mitigate risk.


Subject(s)
Cosmic Radiation/adverse effects , Electron Transport Complex I/metabolism , Gamma Rays/adverse effects , Liver/enzymology , Mitochondria, Liver/enzymology , Radiation Injuries, Experimental/enzymology , Animals , Dose-Response Relationship, Radiation , Liver/pathology , Male , Mice , Mitochondria, Liver/pathology , Proteomics , Radiation Injuries, Experimental/pathology , Space Flight
2.
Int J Mol Sci ; 22(6)2021 Mar 17.
Article in English | MEDLINE | ID: mdl-33802822

ABSTRACT

Future space missions will include a return to the Moon and long duration deep space roundtrip missions to Mars. Leaving the protection that Low Earth Orbit provides will unavoidably expose astronauts to higher cumulative doses of space radiation, in addition to other stressors, e.g., microgravity. Immune regulation is known to be impacted by both radiation and spaceflight and it remains to be seen whether prolonged effects that will be encountered in deep space can have an adverse impact on health. In this study, we investigated the effects in the overall metabolism of three different low dose radiation exposures (γ-rays, 16O, and 56Fe) in spleens from male C57BL/6 mice at 1, 2, and 4 months after exposure. Forty metabolites were identified with significant enrichment in purine metabolism, tricarboxylic acid cycle, fatty acids, acylcarnitines, and amino acids. Early perturbations were more prominent in the γ irradiated samples, while later responses shifted towards more prominent responses in groups with high energy particle irradiations. Regression analysis showed a positive correlation of the abundance of identified fatty acids with time and a negative association with γ-rays, while the degradation pathway of purines was positively associated with time. Taken together, there is a strong suggestion of mitochondrial implication and the possibility of long-term effects on DNA repair and nucleotide pools following radiation exposure.


Subject(s)
Cosmic Radiation , Metabolome/radiation effects , Radiation Exposure , Spleen/metabolism , Spleen/radiation effects , Animals , Citric Acid Cycle/radiation effects , Dose-Response Relationship, Radiation , Linear Models , Male , Mice, Inbred C57BL , Multivariate Analysis , Purines/metabolism
3.
BMC Bioinformatics ; 21(1): 118, 2020 Mar 20.
Article in English | MEDLINE | ID: mdl-32192433

ABSTRACT

BACKGROUND: mRNA interaction with other mRNAs and other signaling molecules determine different biological pathways and functions. Gene co-expression network analysis methods have been widely used to identify correlation patterns between genes in various biological contexts (e.g., cancer, mouse genetics, yeast genetics). A challenge remains to identify an optimal partition of the networks where the individual modules (clusters) are neither too small to make any general inferences, nor too large to be biologically interpretable. Clustering thresholds for identification of modules are not systematically determined and depend on user-settable parameters requiring optimization. The absence of systematic threshold determination may result in suboptimal module identification and a large number of unassigned features. RESULTS: In this study, we propose a new pipeline to perform gene co-expression network analysis. The proposed pipeline employs WGCNA, a software widely used to perform different aspects of gene co-expression network analysis, and Modularity Maximization algorithm, to analyze novel RNA-Seq data to understand the effects of low-dose 56Fe ion irradiation on the formation of hepatocellular carcinoma in mice. The network results, along with experimental validation, show that using WGCNA combined with Modularity Maximization, provides a more biologically interpretable network in our dataset, than that obtainable using WGCNA alone. The proposed pipeline showed better performance than the existing clustering algorithm in WGCNA, and identified a module that was biologically validated by a mitochondrial complex I assay. CONCLUSIONS: We present a pipeline that can reduce the problem of parameter selection that occurs with the existing algorithm in WGCNA, for applicable RNA-Seq datasets. This may assist in the future discovery of novel mRNA interactions, and elucidation of their potential downstream molecular effects.


Subject(s)
Iron/chemistry , Liver/metabolism , Software , Algorithms , Animals , Gene Expression Profiling/methods , Gene Regulatory Networks/genetics , Ions/chemistry , Iron/toxicity , Liver/drug effects , Mice , Mice, Inbred C57BL , RNA-Seq
4.
BMC Genomics ; 21(1): 453, 2020 Jul 01.
Article in English | MEDLINE | ID: mdl-32611366

ABSTRACT

BACKGROUND: One of the health risks posed to astronauts during deep space flights is exposure to high charge, high-energy (HZE) ions (Z > 13), which can lead to the induction of hepatocellular carcinoma (HCC). However, little is known on the molecular mechanisms of HZE irradiation-induced HCC. RESULTS: We performed comparative RNA-Seq transcriptomic analyses to assess the carcinogenic effects of 600 MeV/n 56Fe (0.2 Gy), 1 GeV/n 16O (0.2 Gy), and 350 MeV/n 28Si (0.2 Gy) ions in a mouse model for irradiation-induced HCC. C3H/HeNCrl mice were subjected to total body irradiation to simulate space environment HZE-irradiation, and liver tissues were extracted at five different time points post-irradiation to investigate the time-dependent carcinogenic response at the transcriptomic level. Our data demonstrated a clear difference in the biological effects of these HZE ions, particularly immunological, such as Acute Phase Response Signaling, B Cell Receptor Signaling, IL-8 Signaling, and ROS Production in Macrophages. Also seen in this study were novel unannotated transcripts that were significantly affected by HZE. To investigate the biological functions of these novel transcripts, we used a machine learning technique known as self-organizing maps (SOMs) to characterize the transcriptome expression profiles of 60 samples (45 HZE-irradiated, 15 non-irradiated control) from liver tissues. A handful of localized modules in the maps emerged as groups of co-regulated and co-expressed transcripts. The functional context of these modules was discovered using overrepresentation analysis. We found that these spots typically contained enriched populations of transcripts related to specific immunological molecular processes (e.g., Acute Phase Response Signaling, B Cell Receptor Signaling, IL-3 Signaling), and RNA Transcription/Expression. CONCLUSIONS: A large number of transcripts were found differentially expressed post-HZE irradiation. These results provide valuable information for uncovering the differences in molecular mechanisms underlying HZE specific induced HCC carcinogenesis. Additionally, a handful of novel differentially expressed unannotated transcripts were discovered for each HZE ion. Taken together, these findings may provide a better understanding of biological mechanisms underlying risks for HCC after HZE irradiation and may also have important implications for the discovery of potential countermeasures against and identification of biomarkers for HZE-induced HCC.


Subject(s)
Iron/toxicity , Liver Neoplasms, Experimental/etiology , Oxygen/toxicity , Silicon/toxicity , Animals , Hepatitis/etiology , Hepatitis/genetics , Hepatitis/metabolism , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/metabolism , Machine Learning , Male , Mice , RNA-Seq , Time Factors
5.
Am J Physiol Lung Cell Mol Physiol ; 308(5): L416-28, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25526737

ABSTRACT

Despite considerable progress in identifying health risks to crewmembers related to exposure to galactic/cosmic rays and solar particle events (SPE) during space travel, its long-term effects on the pulmonary system are unknown. We used a murine risk projection model to investigate the impact of exposure to space-relevant radiation (SR) on the lung. C3H mice were exposed to (137)Cs gamma rays, protons (acute, low-dose exposure mimicking the 1972 SPE), 600 MeV/u (56)Fe ions, or 350 MeV/u (28)Si ions at the NASA Space Radiation Laboratory at Brookhaven National Laboratory. Animals were irradiated at the age of 2.5 mo and evaluated 23.5 mo postirradiation, at 26 mo of age. Compared with age-matched nonirradiated mice, SR exposures led to significant air space enlargement and dose-dependent decreased systemic oxygenation levels. These were associated with late mild lung inflammation and prominent cellular injury, with significant oxidative stress and apoptosis (caspase-3 activation) in the lung parenchyma. SR, especially high-energy (56)Fe or (28)Si ions markedly decreased sphingosine-1-phosphate levels and Akt- and p38 MAPK phosphorylation, depleted anti-senescence sirtuin-1 and increased biochemical markers of autophagy. Exposure to SR caused dose-dependent, pronounced late lung pathological sequelae consistent with alveolar simplification and cellular signaling of increased injury and decreased repair. The associated systemic hypoxemia suggested that this previously uncharacterized space radiation-associated lung injury was functionally significant, indicating that further studies are needed to define the risk and to develop appropriate lung-protective countermeasures for manned deep space missions.


Subject(s)
Cosmic Radiation/adverse effects , Lung Injury/etiology , Animals , Apoptosis , Autophagy , Biomarkers/metabolism , Bronchoalveolar Lavage Fluid , Cell Proliferation , Disease Models, Animal , Hypoxia/blood , Hypoxia/complications , Hypoxia/pathology , Lung Injury/blood , Lung Injury/pathology , Male , Mice, Inbred C3H , Oxidative Stress , Oxygen/blood , Pneumonia/blood , Pneumonia/complications , Pneumonia/pathology , Signal Transduction
6.
Lab Invest ; 95(11): 1222-33, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26280221

ABSTRACT

In recent years, increasing threats of radiation exposure and nuclear disasters have become a significant concern for the United States and countries worldwide. Exposure to high doses of radiation triggers a number of potentially lethal effects. Among the most severe is the gastrointestinal (GI) toxicity syndrome caused by the destruction of the intestinal barrier, resulting in bacterial translocation, systemic bacteremia, sepsis, and death. The lack of effective radioprotective agents capable of mitigating radiation-induced damage has prompted a search for novel countermeasures that can mitigate the effects of radiation post exposure, accelerate tissue repair in radiation-exposed individuals, and prevent mortality. We report that a single injection of regenerative peptide TP508 (rusalatide acetate, Chrysalin) 24 h after lethal radiation exposure (9 Gy, LD100/15) appears to significantly increase survival and delay mortality by mitigating radiation-induced intestinal and colonic toxicity. TP508 treatment post exposure prevents the disintegration of GI crypts, stimulates the expression of adherens junction protein E-cadherin, activates crypt cell proliferation, and decreases apoptosis. TP508 post-exposure treatment also upregulates the expression of DCLK1 and LGR5 markers of stem cells that have been shown to be responsible for maintaining and regenerating intestinal crypts. Thus, TP508 appears to mitigate the effects of GI toxicity by activating radioresistant stem cells and increasing the stemness potential of crypts to maintain and restore intestinal integrity. These results suggest that TP508 may be an effective emergency nuclear countermeasure that could be delivered within 24 h post exposure to increase survival and delay mortality, giving victims time to reach clinical sites for advanced medical treatment.


Subject(s)
Gastrointestinal Tract/radiation effects , Peptide Fragments/pharmacology , Stem Cells/drug effects , Thrombin/pharmacology , Adherens Junctions/drug effects , Adherens Junctions/metabolism , Animals , Cell Proliferation/drug effects , Gastrointestinal Tract/pathology , Male , Mice , Mice, Inbred ICR , Stem Cells/cytology , Survival Analysis
7.
Mutagenesis ; 28(1): 71-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22987027

ABSTRACT

Exposure to sparsely ionising gamma- or X-ray irradiation is known to increase the risk of leukaemia in humans. However, heavy ion radiotherapy and extended space exploration will expose humans to densely ionising high linear energy transfer (LET) radiation for which there is currently no understanding of leukaemia risk. Murine models have implicated chromosomal deletion that includes the hematopoietic transcription factor gene, PU.1 (Sfpi1), and point mutation of the second PU.1 allele as the primary cause of low-LET radiation-induced murine acute myeloid leukaemia (rAML). Using array comparative genomic hybridisation, fluorescence in situ hybridisation and high resolution melt analysis, we have confirmed that biallelic PU.1 mutations are common in low-LET rAML, occurring in 88% of samples. Biallelic PU.1 mutations were also detected in the majority of high-LET rAML samples. Microsatellite instability was identified in 42% of all rAML samples, and 89% of samples carried increased microsatellite mutant frequencies at the single-cell level, indicative of ongoing instability. Instability was also observed cytogenetically as a 2-fold increase in chromatid-type aberrations. These data highlight the similarities in molecular characteristics of high-LET and low-LET rAML and confirm the presence of ongoing chromosomal and microsatellite instability in murine rAML.


Subject(s)
Gamma Rays/adverse effects , Leukemia, Myeloid, Acute/etiology , Leukemia, Radiation-Induced , Microsatellite Instability , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , Animals , Cesium Radioisotopes , Chromatids/radiation effects , Chromosome Aberrations , Dose-Response Relationship, Radiation , In Situ Hybridization, Fluorescence , Iron , Leukemia, Myeloid, Acute/genetics , Leukemia, Radiation-Induced/genetics , Linear Energy Transfer , Male , Mice , Mice, Inbred CBA , Mutation , Single-Cell Analysis
8.
Sci Rep ; 12(1): 10927, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35764780

ABSTRACT

Research examining the potential for circulating miRNA to serve as markers for preneoplastic lesions or early-stage hepatocellular carcinoma (HCC) is hindered by the difficulties of obtaining samples from asymptomatic individuals. As a surrogate for human samples, we identified hub miRNAs in gene co-expression networks using HCC-bearing C3H mice. We confirmed 38 hub miRNAs as associated with HCC in F2 hybrid mice derived from radiogenic HCC susceptible and resistant founders. When compared to a panel of 12 circulating miRNAs associated with human HCC, two had no mouse ortholog and 7 of the remaining 10 miRNAs overlapped with the 38 mouse HCC hub miRNAs. Using small RNA sequencing data generated from serially collected plasma samples in F2 mice, we examined the temporal levels of these 7 circulating miRNAs and found that the levels of 4 human circulating markers, miR-122-5p, miR-100-5p, miR-34a-5p and miR-365-3p increased linearly as the time approaching HCC detection neared, suggesting a correlation of miRNA levels with oncogenic progression. Estimation of change points in the kinetics of the 4 circulating miRNAs suggested the changes started 17.5 to 6.8 months prior to HCC detection. These data establish these 4 circulating miRNAs as potential sentinels for preneoplastic lesions or early-stage HCC.


Subject(s)
Carcinoma, Hepatocellular , Circulating MicroRNA , Liver Neoplasms , MicroRNAs , Animals , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/pathology , Circulating MicroRNA/genetics , Humans , Liver Neoplasms/pathology , Mice , Mice, Inbred C3H , MicroRNAs/genetics , Radiopharmaceuticals
10.
Sci Rep ; 11(1): 14052, 2021 07 07.
Article in English | MEDLINE | ID: mdl-34234215

ABSTRACT

High-charge, high-energy ion particle (HZE) radiations are extraterrestrial in origin and characterized by high linear energy transfer (high-LET), which causes more severe cell damage than low-LET radiations like γ-rays or photons. High-LET radiation poses potential cancer risks for astronauts on deep space missions, but the studies of its carcinogenic effects have relied heavily on animal models. It remains uncertain whether such data are applicable to human disease. Here, we used genomics approaches to directly compare high-LET radiation-induced, low-LET radiation-induced and spontaneous hepatocellular carcinoma (HCC) in mice with a human HCC cohort from The Cancer Genome Atlas (TCGA). We identified common molecular pathways between mouse and human HCC and discovered a subset of orthologous genes (mR-HCC) that associated high-LET radiation-induced mouse HCC with a subgroup (mrHCC2) of the TCGA cohort. The mrHCC2 TCGA cohort was more enriched with tumor-suppressing immune cells and showed a better prognostic outcome than other patient subgroups.


Subject(s)
Carcinoma, Hepatocellular/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/radiation effects , Liver Neoplasms/genetics , Radiation, Ionizing , Transcriptome , Animals , Biomarkers, Tumor , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/mortality , Computational Biology/methods , Disease Models, Animal , Humans , Liver Neoplasms/diagnosis , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Mice , Prognosis , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
11.
ACS Omega ; 5(39): 25164-25174, 2020 Oct 06.
Article in English | MEDLINE | ID: mdl-33043195

ABSTRACT

Characterization of lipids by matrix-assisted laser desorption ionization mass spectrometry imaging (MALDI-MSI) is of great interest because not only are lipids important structural molecules in both the cell and internal organelle membranes, but they are also important signaling molecules. MALDI-MSI combined with spatial image segmentation has been previously used to identify tumor heterogeneities within tissues with distinct anatomical regions such as the brain. However, there has been no systematic study utilizing MALDI-MSI combined with spatial image segmentation to assess the tumor microenvironment in the liver. Here, we present that image segmentation can be used to evaluate the tumor microenvironment in the liver. In particular, to better understand the molecular mechanisms of irradiation-induced hepatic carcinogenesis, we used MALDI-MSI in the negative ion mode to identify lipid changes 12 months post exposure to low dose 28Si and 137Cs γ ray irradiation. We report here the changes in the lipid profiles of male C3H/HeNCrl mice liver tissues after exposure to irradiation and analyzed using the spatial shrunken centroid clustering algorithm. These findings provide valuable information as astronauts will be exposed to high-charge high-energy (HZE) particles and low-energy γ-ray irradiation during deep space travel. Even at low doses, exposure to these irradiations can lead to cancer. Previous studies infer that irradiation of mice with low-dose HZE particles induces oxidative damage and microenvironmental changes that are thought to play roles in the pathophysiology of hepatocellular carcinoma.

12.
Radiat Res ; 171(4): 484-93, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19397449

ABSTRACT

Chromosome aberrations in mitotic bone marrow cells of CBA/Ca and C57BL/6 mice were measured 1 day after exposure to 1 Gy of 1 GeV/nucleon 56Fe ions or 3 Gy of gamma rays. The proportion that have lost a region of chromosome 2 containing the PU.1 gene could be explained by a model based on these measurements. The distribution of aberrations among cells was close to the expected Poisson for the gamma-irradiated cells, but for the HZE 56Fe ions the distribution was highly dispersed. The observations were consistent with the results of an analysis similar to that of Edwards and co-workers in 1980 after ex vivo irradiation of human blood with alpha particles. The analysis used to fit the current data was based on a compound Poisson process, also used previously by others, but in addition included the random nature of parameters involved such as cell nuclear diameter, particle traversal lengths through cell nuclei, production of aberrations, and cell cycle arrest per traversal. From the measured numbers of acentric fragments produced, the relative size of chromosome 2 and the region associated with PU.1 deletions, an independent prediction of PU.1 loss agreed well with measurements described in the accompanying paper.


Subject(s)
Gene Expression Regulation, Leukemic , Iron , Leukemia/etiology , Leukemia/metabolism , Neoplasms, Radiation-Induced/etiology , Neoplasms, Radiation-Induced/metabolism , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Animals , Chromosome Aberrations , Chromosomes , Gamma Rays , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Radiometry , X-Rays
13.
Radiat Res ; 172(2): 213-9, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19630525

ABSTRACT

Abstract Estimates of cancer risks posed to space-flight crews by exposure to high atomic number, high-energy (HZE) ions are subject to considerable uncertainty because epidemiological data do not exist for human populations exposed to similar radiation qualities. We assessed the leukemogenic efficacy of one such HZE species, 1 GeV (56)Fe ions, a component of space radiation, in a mouse model for radiation-induced acute myeloid leukemia. CBA/CaJ mice were irradiated with 1 GeV/nucleon (56)Fe ions or (137)Cs gamma rays and followed until they were moribund or to 800 days of age. We found that 1 GeV/nucleon (56)Fe ions do not appear to be substantially more effective than gamma rays for the induction of acute myeloid leukemia (AML). However, (56)Fe-ion-irradiated mice had a much higher incidence of hepatocellular carcinoma (HCC) than gamma-irradiated mice, with an estimated RBE of approximately 50. These data suggest a difference in the effects of HZE iron ions on the induction of leukemia compared to solid tumors, suggesting potentially different mechanisms of tumorigenesis.


Subject(s)
Carcinoma, Hepatocellular/epidemiology , Carcinoma, Hepatocellular/veterinary , Leukemia, Myeloid/epidemiology , Leukemia, Myeloid/veterinary , Liver Neoplasms/epidemiology , Liver Neoplasms/veterinary , Neoplasms, Radiation-Induced/epidemiology , Neoplasms, Radiation-Induced/veterinary , Animals , Cosmic Radiation , Dose-Response Relationship, Radiation , Heavy Ions , Incidence , Iron , Male , Mice , Radiation Dosage , Risk Assessment/methods , Risk Factors , Whole-Body Irradiation/statistics & numerical data
14.
Radiat Res ; 171(4): 474-83, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19397448

ABSTRACT

Since deletion of the PU.1 gene on chromosome 2 is a crucial acute myeloid leukemia (AML) initiating step in the mouse model, we quantified PU.1 deleted cells in the bone marrow of gamma-, X- and 56Fe-ion-irradiated mice at various times postirradiation. Although 56Fe ions were initially some two to three times more effective than X or gamma rays in inducing PU.1 deletions, by 1 month postirradiation, the proportions of cells with PU.1 deletions were similar for the HZE particles and the sparsely ionizing radiations. These results indicate that while 56Fe ions are more effective in inducing PU.1 deletions, they are also more effective in causing collateral damage that removes hit cells from the bone marrow. After X, gamma or 56Fe-ion irradiation, AML-resistant C57BL/6 mice have fewer cells with PU.1 deletions than CBA mice, and those cells do not persist in the bone marrow of the C57B6/6 mice. Our findings suggest that quantification of PU.1 deleted bone marrow cells 1 month postirradiation can be used as surrogate for the incidence of radiation-induced AML measured in large-scale mouse studies. If so, PU.1 loss could be used to systematically assess the potential leukemogenic effects of other ions and energies in the space radiation environment.


Subject(s)
Gene Expression Regulation, Leukemic , Iron , Leukemia/etiology , Leukemia/metabolism , Neoplasms, Radiation-Induced/etiology , Neoplasms, Radiation-Induced/metabolism , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Animals , Chromosomes , Chromosomes, Artificial, Bacterial/metabolism , Dose-Response Relationship, Radiation , Gamma Rays , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , X-Rays
15.
Cancer Invest ; 26(6): 553-61, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18584345

ABSTRACT

BACKGROUND: We designed and tested, in vitro, an adenoviral construct containing the feline interleukin-12 (IL-12) gene under control of the heat-inducible promoter HSP70B. This construct, AdhspfIL12, was used in a phase I trial in feline soft tissue sarcomas. During the course of our experiments, we noted that IL-12 was being produced in the transfected Crandell Feline Kidney (CrFK) cells under certain conditions even in the absence of hyperthermia. This observation was further explored to identify the cause of this unintended HSP70B induction. MATERIALS AND METHODS: We used real-time PCR as a sensitive method to quantitatively detect the presence of even small amounts of IL-12 mRNA. This served as a surrogate indicator of HSP70B induction. Various conditions were tested to induce the heat shock promoter, including nutritional deprivation, radiation and changes in pH. RESULTS: Nutritional stresses, specifically the absence of glucose and glutamine, could induce the heat shock promoter, thus, resulting in production of the downstream gene product. Other factors known to trigger the heat shock response, pH change, and reactive oxygen species production were also studied but were not found to contribute to heat shock promoter induction in our setting. CONCLUSIONS: The human heat shock promoter (HSP70B) is reported to be an efficient and tightly regulated promoter. We discovered, using sensitive real-time PCR techniques, that it can also be induced in response to cellular nutrient stresses. The pros and cons of this phenomenon and its implications for cancer gene therapy are discussed.


Subject(s)
Genetic Therapy/methods , HSP70 Heat-Shock Proteins/genetics , Interleukin-12/biosynthesis , Promoter Regions, Genetic , Stress, Physiological/metabolism , Transcriptional Activation , Adenoviridae/genetics , Animals , Antimitotic Agents/pharmacology , Aphidicolin/pharmacology , Cats , Cell Line , Cell Proliferation , Culture Media/metabolism , Genetic Therapy/adverse effects , Genetic Vectors , Glucose/deficiency , Glutamine/deficiency , Humans , Hydrogen-Ion Concentration , Hyperthermia, Induced , Interleukin-12/genetics , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , Reactive Oxygen Species/metabolism , Stress, Physiological/genetics , Time Factors , Transcriptional Activation/drug effects , Transfection
16.
Radiat Res ; 169(6): 633-8, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18494544

ABSTRACT

Resveratrol, a polyphenol compound with reported antioxidant and anticarcinogenic effects, a wide range of molecular targets, and toxicity only at extreme doses, has received considerable attention. We evaluated the radioprotective effect of orally administered resveratrol on the frequencies of chromosome aberrations in irradiated mouse bone marrow cells. CBA/CaJ mice were divided into four groups: (1) no treatment, (2) resveratrol only, (3) radiation only, and (4) resveratrol and radiation. Resveratrol treatment (100 mg/kg daily) was initiated 2 days prior to irradiation. Bone marrow was then harvested at 1 and 30 days after a single dose of 3 Gy whole-body gamma radiation. A statistically significant (P < 0.05) reduction in the mean total chromosome aberration frequency per metaphase at both times postirradiation in the resveratrol and radiation group compared to the radiation-only group was observed. This study is the first to demonstrate that resveratrol has radioprotective effects in vivo. These results support the use of resveratrol as a radioprotector with the potential for widespread application.


Subject(s)
Bone Marrow Cells/radiation effects , Chromosome Aberrations , Chromosomes/radiation effects , Radiation-Protective Agents/pharmacology , Stilbenes/pharmacology , Animals , Bone Marrow/radiation effects , Gamma Rays , Male , Mice , Mice, Inbred CBA , Models, Statistical , Resveratrol , Whole-Body Irradiation
17.
Mol Cancer Ther ; 6(1): 380-9, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17237297

ABSTRACT

Interleukin-12 (IL-12), a proinflammatory cytokine, shows anticancer properties. Systemically administered IL-12 causes dose-dependent toxicity. To achieve localized intratumoral gene expression, an adenoviral gene therapy vector with IL-12 controlled by a heat-inducible promoter (heat shock promoter 70B) was developed and tested in a phase I clinical trial in cats with spontaneously arising soft tissue sarcoma. A feasibility study was done in 16 cats with soft tissue sarcoma using murine IL-12 and/or enhanced green fluorescent protein adenoviral vectors under cytomegalovirus or heat shock promoter 70 control. Subsequently, we conducted a phase I clinical trial using an adenoviral feline IL-12 construct in 13 cats with soft tissue sarcoma. The soft tissue sarcomas were irradiated (48 Gy/16 fractions) followed by intratumoral injection of adenovirus. Twenty-four hours postinjection, tumors were heated (41 degrees C, 60 min). Tumor expression of feline IL-12 and IFN-gamma was determined. Cats were monitored for systemic toxicity. For the murine IL-12 construct, an association was noted between viral dose and murine IL-12 levels within tumor, whereas serum levels were minimal. Mild toxicity was noted at 10(11) plaque-forming units (pfu). With the feline IL-12 construct, high levels of feline IL-12 mRNA were detected in tumor biopsies with low or absent IFN-gamma mRNA following gene therapy. Hematologic and hepatic toxicities were noted at the highest viral doses and were associated with detection of IFN-gamma mRNA in tumor. It is possible to localize gene expression and limit systemic toxicity of IL-12 using the hyperthermia-induced gene therapy approach. The maximum tolerated dose of the feline IL-12 adenoviral vector was 10(10) pfu/tumor as dose-limiting toxicities were noted at the 4 x 10(10) pfu dose.


Subject(s)
Genetic Therapy , Hyperthermia, Induced , Interleukin-12/genetics , Interleukin-12/therapeutic use , Sarcoma/veterinary , Adenoviridae , Animals , Cats , Cytomegalovirus/genetics , Feasibility Studies , Genetic Therapy/adverse effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Interleukin-12/blood , Liver/pathology , Mice , Promoter Regions, Genetic/genetics , Recombinant Proteins/adverse effects , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/therapeutic use , Sarcoma/drug therapy , Sarcoma/genetics , Sarcoma/radiotherapy
18.
Cancer Lett ; 250(1): 63-73, 2007 May 18.
Article in English | MEDLINE | ID: mdl-17095151

ABSTRACT

The correct repair of DNA double-strand breaks (DSBs) is essential to maintaining the integrity of the genome. Misrepair of DSBs is detrimental to cells and organisms, leading to gene mutation, chromosomal aberration, and cancer development. Nonhomologous end-joining (NHEJ) is one of the principal rejoining processes in most higher eukaryotic cells. NHEJ is facilitated by DNA-dependent protein kinase (DNA-PK), which is composed of a catalytic subunit, DNA-PKcs, and the heterodimeric DNA binding regulatory complex Ku70/86. Null mutation of DNA-PKcs leads to immunodeficiency, chromosomal aberration, gene mutation, telomeric end-capping failure, and cancer predisposition in animals and cells. However, it is unknown whether partial deficiency of DNA-PKcs as might occur in a fraction of the population (e.g., heterozygotes), influences cellular function. Using small interfering RNA (siRNA) transfection, we established partial deficiency of DNA-PKcs in human cells, ranging from 4 to 85% of control levels. Our results reveal for the first time, that partial deficiency of DNA-PKcs leads to increased ionizing radiation (IR)-induced mutagenesis, cell killing, and telomere dysfunction. Radiation mutagenesis was increased inversely with DNA-PKcs protein level, with the most pronounced effect being observed in cells with protein levels below 50% of controls. A small but statistically significant increase in IR-induced cell killing was observed as DNA-PKcs levels decreased, over the entire range of protein levels. Frequencies of IR-induced telomere-DSB fusion was increased at levels of DNA-PKcs as low as approximately 50%, similar to what would be expected in heterozygous individuals. Taken together, our results suggest that even partial deficiency of DNA repair proteins may represent a considerable risk to genomic stability.


Subject(s)
DNA-Activated Protein Kinase/metabolism , Lymphocytes/enzymology , Lymphocytes/radiation effects , Mutagenesis , Radiation, Ionizing , Telomere/physiology , Cell Line , Genomic Instability , Humans , Transfection
19.
Cancer Res ; 64(16): 5608-16, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15313898

ABSTRACT

The absence of p53 function increases risk for spontaneous tumorigenesis in the mammary gland. Hormonal stimulation enhances tumor risk in p53-null mammary epithelial cells as well as the incidence of aneuploidy. Aneuploidy appears in normal p53-null mammary epithelial cells within 5 weeks of hormone stimulation. Experiments reported herein assessed a possible mechanism of hormone-induced aneuploidy. Hormones increased DNA synthesis equally between wild-type (WT) and p53-null mammary epithelial cells. There were two distinct responses in p53-null cells to hormone exposure. First, Western blot analysis demonstrated that the levels of two proteins involved in regulating sister chromatid separation and the spindle checkpoint, Mad2 and separase (ESPL1) were increased in null compared with WT cells. In contrast, the levels of securin and Rad21 proteins were not increased in hormone-stimulated p53-null compared with WT cells. ESPL1 RNA was also increased in p53-null mouse mammary cells in vivo by 18 h of hormone stimulation and in human breast MCF7 cells in monolayer culture by 8 h of hormone stimulation. Furthermore, both promoters contained p53 and steroid hormone response elements. Mad2 protein was increased as a consequence of the absence of p53 function. The increase in Mad2 protein was observed also at the cellular level by immunohistochemistry. Second, hormones increased gene amplication in the distal arm of chromosome 2, as shown by comparative genomic hybridization. These results support the hypothesis that hormone stimulation acts to increase aneuploidy by several mechanisms. First, by increasing mitogenesis in the absence of the p53 checkpoint in G2, hormones allow the accumulation of cells that have experienced chromosome missegregation. Second, the absolute rate of chromosome missegregation may be increased by alterations in the levels of two proteins, separase and Mad2, which are important for maintaining chromosomal segregation and the normal spindle checkpoint during mitosis.


Subject(s)
Aneuploidy , Chromosomal Instability , Estrogens/pharmacology , Mammary Glands, Animal/physiology , Progesterone/pharmacology , Tumor Suppressor Protein p53/deficiency , Animals , Base Sequence , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Calcium-Binding Proteins/biosynthesis , Calcium-Binding Proteins/genetics , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Cell Line, Tumor , Endopeptidases/biosynthesis , Endopeptidases/genetics , Epithelial Cells/drug effects , Epithelial Cells/pathology , Epithelial Cells/physiology , Female , Mad2 Proteins , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/pathology , Mice , Mice, Inbred BALB C , Repressor Proteins , Separase , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/physiology
20.
DNA Repair (Amst) ; 3(4): 349-57, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-15010310

ABSTRACT

In addition to joining broken DNA strands, several non-homologous end-joining (NHEJ) proteins have a second seemingly antithetical role in constructing functional telomeres, the nucleoprotein structures at the termini of linear eukaryotic chromosomes that prevent joining between natural chromosome ends. Although NHEJ deficiency impairs double-strand break (DSB) repair, it also promotes inappropriate chromosomal end fusions that are observed microscopically as dicentric chromosomes with telomeric DNA sequence at points of joining. Here, we test the proposition that unprotected telomeres can fuse not only to other dysfunctional telomeres, but also to ends created by DSBs. Severe combined immunodeficiency (scid) is caused by a mutation in the catalytic subunit of DNA-dependent protein kinase (DNA-PK), an enzyme required for both efficient DSB repair and telomeric end-capping. Cells derived from wild-type, Trp53-/-, scid, and Trp53-/-/scid mice were exposed to gamma radiation to induce DSBs, and chromosomal aberrations were analyzed using a novel cytogenetic technique that can detect joining of a telomere to a DSB end. Telomere-DSB fusions were observed in both cell lines having the scid mutation, but not in wild-type nor Trp53-/- cells. Over a range of 25-340 cGy, half of the visible exchange-type chromosomal aberrations in Trp53-/-/scid cells involved telomere-DSB fusions. Our results demonstrate that unprotected telomeres are not only sensed as, but also acted upon, by the DNA repair machinery as if they were DSB ends. By opening a new pathway for misrepair, telomere-DSB fusion decreases the overall fidelity of DSB repair. The high frequency of these events in scid cells indicates telomere dysfunction makes a strong, and previously unsuspected, contribution to the characteristic radiation sensitivity associated with DNA-PK deficiency.


Subject(s)
DNA Damage , Telomere , Animals , DNA-Activated Protein Kinase , DNA-Binding Proteins , In Situ Hybridization, Fluorescence , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Nuclear Proteins
SELECTION OF CITATIONS
SEARCH DETAIL