Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Placenta ; 35(2): 85-91, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24418702

ABSTRACT

INTRODUCTION: Exposure to low-dose radiation is widespread and attributable to natural sources. However, occupational, medical, accidental, and terrorist-related exposures remain a significant threat. Information on radiation injury to the feto-placental unit is scant and largely observational. We hypothesized that radiation causes trophoblast injury, and alters the expression of injury-related transcripts in vitro or in vivo, thus affecting fetal growth. METHODS: Primary human trophoblasts (PHTs), BeWo or NCCIT cells were irradiated in vitro, and cell number and viability were determined. Pregnant C57Bl/6HNsd mice were externally irradiated on E13.5, and placentas examined on E17.5. RNA expression was analyzed using microarrays and RT-qPCR. The experiments were repeated in the presence of the gramicidin S (GS)-derived nitroxide JP4-039, used to mitigate radiation-induced cell injury. RESULTS: We found that survival of in vitro-irradiated PHT cell was better than that of irradiated BeWo trophoblast cell line or the radiosensitive NCCIT mixed germ cell tumor line. Radiation altered the expression of several trophoblast genes, with a most dramatic effect on CDKN1A (p21, CIP1). Mice exposed to radiation at E13.5 exhibited a 25% reduction in mean weight by E17.5, and a 9% reduction in placental weight, which was associated with relatively small changes in placental gene expression. JP4-039 had a minimal effect on feto-placental growth or on gene expression in irradiated PHT cells or mouse placenta. DISCUSSION AND CONCLUSION: While radiation affects placental trophoblasts, the established placenta is fairly resistant to radiation, and changes in this tissue may not fully account for fetal growth restriction induced by ionizing radiation.


Subject(s)
Fetal Development/radiation effects , Gene Expression Regulation, Developmental/radiation effects , Radiation, Ionizing , Trophoblasts/radiation effects , Animals , Cell Line , Female , Fetal Growth Retardation/etiology , Humans , Mice , Nitrogen Oxides/therapeutic use , Placenta/radiation effects , Pregnancy , Radiation Injuries/drug therapy , Whole-Body Irradiation/adverse effects
2.
Gene Ther ; 18(6): 579-83, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21248791

ABSTRACT

To determine the effects of manganese superoxide dismutase (MnSOD) plasmid liposome (PL) maternal radioprotection on fetal mice, timed pregnant female mice (E14 gestation) were irradiated to 3.0 Gy total body irradiation (TBI) dose, and the number, weight and growth and development over 6 months after birth of newborn mice was quantitated compared with irradiated controls. Maternal MnSOD-PL treatment at E13 improved pup survival at birth (5.4±0.9 per litter) compared with non-irradiated 3.0 Gy controls 4.9±1.1. There was no statistically significant difference in newborn abnormalities, male to female ratio in newborn litters, or other evidence of teratogenesis in surviving newborn mice from MnSOD-PL treated compared with irradiated controls. However, E14 3 Gy irradiated pups from gene therapy-treated mothers showed a significant increase in both growth and overall survival over 6 months after birth (P=0.0022). To determine if transgene product crossed the placenta pregnant E13 mice were injected intravenously with hemagglutinin-epitope-tagged MnSOD (100 µg plasmid in 100 µl liposomes), then after 24 h, fetal mice, placentas and maternal tissues were removed and tested by both immunohistochemistry and reverse transcriptase-PCR for transgene and product. There was no evidence of transgene or product in placenta or any fetal tissue while maternal liver was positive by both assays. The data provide evidence for fetal radioprotection by maternal MnSOD-PL gene therapy before irradiation, which is mediated by an indirect bystander effect and is associated with a significant improvement in both survival at birth and growth and development of newborn mice.


Subject(s)
Animals, Newborn/growth & development , Genetic Therapy/methods , Liposomes , Pregnancy, Animal , Prenatal Exposure Delayed Effects , Radiation-Protective Agents/administration & dosage , Superoxide Dismutase/genetics , Whole-Body Irradiation/adverse effects , Animals , Female , Fetal Death/prevention & control , Fetal Growth Retardation/prevention & control , Fetus/radiation effects , Maternal-Fetal Exchange , Mice , Mice, Inbred C57BL , Pregnancy
3.
Gene Ther ; 17(9): 1181-90, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20827278

ABSTRACT

Duchenne muscular dystrophy is a fatal, genetic disorder in which dystrophin-deficient muscle progressively degenerates, for which dystrophin gene transfer could provide effective treatment. The host immune response to dystrophin, however, is an obstacle to therapeutic gene expression. Understanding the dystrophin-induced host immune response will facilitate the discovery of strategies to prolong expression of recombinant dystrophin in dystrophic muscle. Using whole-body irradiation of the dystrophic mdx mouse before gene transfer, we temporally removed the immune system; a 600 rad dose removed peripheral immune cells, which were restored by self-reconstitution, and a 900 rad dose removed central and peripheral immune cells, which were restored by adoptive transfer of bone marrow from a syngeneic, dystrophin-normal donor. The anti-dystrophin humoral response was delayed and dystrophin expression was partially preserved in irradiated, vector-treated mice. Nonirradiated, vector-treated control mice lost muscle dystrophin expression completely, had an earlier anti-dystrophin humoral response and demonstrated muscle fibers focally surrounded with T cells. We conclude that dystrophin gene transfer induced anti-dystrophin humoral immunity and cell-mediated responses that were significantly diminished and delayed by temporal removal of the host central or peripheral immune cells. Furthermore, manipulation of central immunity altered the pattern of regulatory T cells in muscle.


Subject(s)
Dystrophin/genetics , Immunity, Humoral/radiation effects , Muscular Dystrophy, Duchenne/immunology , Whole-Body Irradiation , Animals , DNA, Complementary/administration & dosage , Dystrophin/immunology , Gene Transfer Techniques , Genetic Vectors , Mice , Mice, Inbred mdx , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/radiation effects
4.
Gene Ther ; 15(5): 347-56, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18097469

ABSTRACT

We evaluated whether the improved esophageal radiation tolerance following Manganese Superoxide Dismutase (MnSOD)-Plasmid Liposomes was explained by improved engraftment of bone marrow-derived progenitors. C57BL/6NHsd female mice pretreated with intraesophageal MnSOD-PL were irradiated to 29 Gy to the esophagus and intravenously transplanted with marrow from male B6. 129S7-Gt (ROSA) 26S OR/J ROSA (Lac-Z+, G418-resistant) mice. After 14 days, esophagi were removed and side population and non-side population cells evaluated for donor multilineage (endothelin/vimentin/F480) positive esophageal cells. Serial intravenous transplantability was tested in second generation 29 Gy esophagus-irradiated mice. Esophagi from recipients receiving swallowed MnSOD-PL 24 h prior to irradiation demonstrated significantly increased esophageal repopulation with donor bone marrow-derived Lac-Z+, G418+, Y-probe+ multilineage cells (37.8+/-1.8>50 cell Lac-Z+ foci per esophagus) compared to irradiated controls (19.8+/-1.8) P<0.0001. Serial transfer to second-generation irradiated C57BL/6NHsd mice of intravenously injected SP or NSP first generation recipient esophagus cells was also significantly enhanced by MnSOD-PL intraesophageal pretreatment (74.4+/-3.6 SP-derived Lac-Z+ foci per esophagus, 48.6+/-5.4 NSP-derived) compared to irradiation controls (23.4+/-1.8 SP, 6.0+/-3.0 NSP), P<0.0001. Thus, intraesophageal MnSOD-PL administration enhances engraftment of marrow-derived progenitors.


Subject(s)
Esophagus/injuries , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Hematopoietic Stem Cell Transplantation/methods , Radiation Injuries, Experimental/therapy , Superoxide Dismutase/genetics , Administration, Oral , Animals , Combined Modality Therapy , Esophagus/metabolism , Esophagus/pathology , Female , Gene Expression , Genetic Vectors/genetics , In Situ Hybridization , Lac Operon , Liposomes/administration & dosage , Male , Mice , Mice, Inbred C57BL , Plasmids , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/pathology , Superoxide Dismutase/metabolism , Wound Healing , Y Chromosome
5.
Gene Ther ; 12(8): 685-93, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15750616

ABSTRACT

Intratracheal injection of manganese superoxide dismutase-plasmid/liposome (MnSOD-PL) complexes has been demonstrated to delay the onset and reduce the extent of ionizing irradiation-induced murine pulmonary organizing alveolitis/fibrosis. To facilitate translation of this modality to clinical fractionated radiotherapy, inhalation delivery of MnSOD-PL was developed using an ultrasonic nebulizer. Transgene product was quantitated by immunohistochemical quantitation and pulmonary tissue levels of MnSOD biochemical activity. C57BL/6NHsd female mice demonstrated a plasmid dose-dependent increased expression of MnSOD transgene product over the range of 250 microg-2.5 mg of MnSOD-PL administered over a constant 5 min interval. Delivery of a constant concentration of 500 microg of MnSOD-PL with varying times of administration ranging from 0.5 to 10 min demonstrated optimal MnSOD expression at 5 min. Mice pretreated by inhalation delivery of MnSOD-PL demonstrated significantly improved survival after 20 Gy single fraction irradiation to both lungs compared to LacZ-PL inhalation-treated or irradiated control mice. Mice receiving 10 fractions of 3.5 cGy demonstrated increased pulmonary MnSOD transgene product activity by a protocol of every Monday-Wednesday or daily inhalation of MnSOD-PL. Thus, inhalation radioprotective gene therapy using MnSOD-PL provides a practical and effective method for delivery of lung-specific radioprotection during fractionated radiotherapy protocols in a mouse model.


Subject(s)
DNA/administration & dosage , Genetic Therapy/methods , Lung/radiation effects , Pulmonary Fibrosis/prevention & control , Radiotherapy/adverse effects , Superoxide Dismutase/genetics , Administration, Inhalation , Animals , Female , Gene Expression , Immunohistochemistry/methods , Liposomes , Lung/enzymology , Lung/metabolism , Mice , Mice, Inbred C57BL , Models, Animal , Nebulizers and Vaporizers , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/metabolism , Radiotherapy Dosage , Superoxide Dismutase/analysis , Superoxide Dismutase/metabolism , Transgenes , Ultrasonics
6.
Curr Gene Ther ; 3(3): 183-95, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12762478

ABSTRACT

Control of cancer by irradiation therapy alone or in conjunction with combination chemotherapy is often limited by organ specific toxicity. Ionizing irradiation toxicity is initiated by damage to normal tissue near the tumor target and within the transit volume of radiotherapy beams. Irradiation-induced cellular, tissue, and organ damage is mediated by acute effects, which can be dose limiting. A latent period follows recovery from the acute reaction, then chronic irradiation fibrosis (late effects) pose a second cause of organ failure. We have developed the technology for radioprotective gene therapy using the transgene for the antioxidant manganese superoxide dismutase, delivered to specific target organs (lung, esophagus, oral cavity, oropharynx, and bladder) using gene transfer vectors including plasmid/liposomes (PL) and adenovirus. Irradiation protection by MnSOD transgene overexpression at the cellular level has been demonstrated to be localized to the mitochondrial membrane. Using MnSOD transgene constructs lacking the mitochondrial localization leader sequence, and in other experiments attaching this localization signal to otherwise non-radioprotective cytoplasmic Cu/ZnSOD, mitochondrial localization has been demonstrated to be critical to protection. Organ specific injection of MnSOD-PL prior to irradiation demonstrates transgene expression for 48-72 hours, and an associated decrease in ionizing irradiation-induced expression of inflammatory cytokine mRNA and protein. Significant reduction of organ specific tissue injury has been demonstrated in several organ systems in rodent models. Application of MnSOD-PL gene therapy in the setting of fractionated chemo-radiotherapy is being tested in clinical trials for prevention of esophagitis during treatment of non-small cell carcinoma of the lung, and in prevention of mucositis during combination therapy of carcinomas of the head and neck. Encouraging results in pre-clinical models suggest that radioprotective gene therapy may facilitate dose escalation protocols to allow increases in the therapeutic ratio of cancer radiotherapy.


Subject(s)
Genetic Therapy , Neoplasms, Radiation-Induced/therapy , Radiation-Protective Agents/therapeutic use , Superoxide Dismutase/genetics , Animals , Mice , Mice, Inbred Strains , Transgenes
7.
Gene Ther ; 10(2): 163-71, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12571645

ABSTRACT

Intratracheal (IT) injection of manganese superoxide dismutase-plasmid/liposome (MnSOD-PL) complexes prior to whole lung irradiation of C57BL/6J mice provides significant protection from acute and chronic irradiation damage. We determined the duration of increased MnSOD biochemical activity and differential expression of a hemagglutinin (HA) epitope-tagged MnSOD transgene. HA-MnSOD-PL was IT injected at doses of 0-1000 microg, and mice were killed 1,2,3 or 4 days later. Other groups of mice were irradiated to 20 Gy to the pulmonary cavity 24 h after injection and killed at the same time points as non-irradiated mice. Both non-irradiated and irradiated groups of mice showed increased MnSOD biochemical activity with plasmid dose that plateaued at 100 microg of MnSOD plasmid DNA. In control mice, MnSOD biochemical activity decreased at 2, 3 or 4 days after injection. In irradiated mice, MnSOD biochemical activity decreased at day 2 but increased on days 3 and 4. HA-MnSOD expression decreased in broncheoalveolar macrophages and alveolar type-II cells 3 days after injection in non-irradiated and irradiated mice, but remained elevated in endothelial and epithelial cells past 4 days. The data provide a rationale for every second-day administration of intrapulmonary MnSOD-PL in clinical trials of radioprotective gene therapy. This should be sufficient to provide radioprotection during radiation treatments.


Subject(s)
Genetic Therapy/methods , Lung/enzymology , Lung/radiation effects , Radiation Injuries, Experimental/prevention & control , Radiation-Protective Agents , Superoxide Dismutase/genetics , Animals , Endothelium, Vascular/enzymology , Epithelial Cells/enzymology , Gene Expression , Hemagglutinins/genetics , Injections , Liposomes , Macrophages, Alveolar/enzymology , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Pulmonary Alveoli/enzymology , Radiation Injuries, Experimental/enzymology , Time Factors
8.
Antioxid Redox Signal ; 3(3): 347-59, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11491649

ABSTRACT

Reactive oxygen species (ROS)-mediated damage to DNA is associated with induction of stress-activated protein kinases leading to secondary and tertiary effects on the nuclear matrix, cytoplasmic transport mechanisms, and altered mitochondrial and cell membranes. The cellular defenses against ROS damage are associated with up-regulation of gene products that can significantly alter cell biology, including antiapoptotic Bax family proteins and inflammatory proteins. Altered cell integrity can occur either directly or by indirect paracrine and juxtacrine interactions within tissues. Previous approaches toward therapeutic intervention against ROS damage have included administration of radical scavenger compounds, use of novel drugs that increase cellular production of constitutive antioxidants, or pharmacologic agents that modify the intracellular transport of antioxidants. Strategies to modify the cellular effects of ROS in hyperbaric oxygen injury to the lung, reperfusion injury to transplanted organs, and cancer have led to novel approaches of gene therapy in which the transgenes for antioxidant proteins can be expressed in specific tissues. Reducing tissue-damaging effects of ROS may have relevance to cancer patients by ameliorating normal tissue damage from ionizing irradiation therapy, photodynamic therapy, and cancer chemotherapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Antioxidants/therapeutic use , Genetic Therapy , Neoplasms/therapy , Reactive Oxygen Species/metabolism , Superoxide Dismutase/genetics , Animals , Carcinoma, Lewis Lung/pathology , Carcinoma, Lewis Lung/radiotherapy , Carcinoma, Lewis Lung/therapy , Cell Line , Cytokines/pharmacology , Dose-Response Relationship, Radiation , Electron Transport/radiation effects , Free Radical Scavengers/therapeutic use , Genetic Therapy/methods , Glutathione/metabolism , Mice , Mice, Inbred C57BL , Neoplasms/metabolism , Neoplasms/prevention & control , Oxidation-Reduction , Radiation-Protective Agents/therapeutic use , Signal Transduction , Superoxide Dismutase/physiology , Superoxide Dismutase/therapeutic use , Tumor Cells, Cultured
9.
Int J Cancer ; 96(4): 221-31, 2001 Aug 20.
Article in English | MEDLINE | ID: mdl-11474496

ABSTRACT

Intraesophageal administration of manganese superoxide dismutase-plasmid/liposome (MnSOD-PL) prior to single fraction radiation has been shown to protect mice from lethal esophagitis. In our study, C3H/HeNsd mice received fractionated radiation in two protocols: (i) 18 Gy daily for four days with MnSOD-PL administration 24 hr prior to the first and third fraction, or (ii) 12 Gy daily for six days with MnSOD-PL 24 hr prior to the first, third, and fifth fraction. Control radiated mice received either no liposomes only or LacZ (bacterial beta-galactosidase gene)-plasmid/liposome (LacZ-PL) by the same schedules. We measured thiol depletion and lipid peroxidation (LP) in whole esophagus and tested the effectiveness of a new plasmid, hemagglutinin (HA) epitope-tagged MnSOD (HA-MnSOD). In fractionation protocols, mice receiving MnSOD-PL, but not LacZ-PL (200 microl of plasmid/liposomes containing 200 microg of plasmid DNA), showed a significant reduction in morbidity, decreased weight loss, and improved survival. Four and seven days after 37 Gy single fraction radiation, the esophagus demonstrated a significant increase in peroxidized lipids and reduction in overall antioxidant levels, reduced thiols, and decreased glutathione (GSH). These reductions were modulated by MnSOD-PL administration. The HA-MnSOD plasmid product was detected in the basal layers of the esophageal epithelium 24 hr after administration and provided significant radiation protection compared to glutathione peroxidase-plasmid/liposome (GPX-PL), or liposomes containing MnSOD protein, vitamin E, co-enzyme Q10, or 21-aminosteroid. Thus, MnSOD-PL administration significantly improved tolerance to fractionated radiation and modulated radiation effects on levels of GSH and lipid peroxidation (LP). These studies provide further support for translation of MnSOD-PL treatment into human esophageal radiation protection.


Subject(s)
Esophagitis/etiology , Esophagitis/prevention & control , Liposomes/therapeutic use , Plasmids/therapeutic use , Superoxide Dismutase/therapeutic use , Animals , Biomarkers , Cells, Cultured , Chromatography, High Pressure Liquid , Dinoprost/metabolism , Dose-Response Relationship, Drug , Epitopes/metabolism , Fatty Acids, Unsaturated/metabolism , Female , Hemagglutinins/metabolism , Lac Operon , Lipid Metabolism , Lipid Peroxidation/radiation effects , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Radiotherapy/adverse effects , Superoxide Dismutase/chemistry , Time Factors
10.
Nitric Oxide ; 5(2): 128-36, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11292362

ABSTRACT

In 32D cl 3 hematopoietic progenitor cells, the overexpression of manganese superoxide dismutase (MnSOD, SOD2), the enzyme normally found in mitochondria, protects against the damaging effects of ionizing radiation. In the presence of a nitric oxide donor, which exacerbates the damage, inhibition of mitochondrial function can be demonstrated to be associated with respiratory complexes I (NADH dehydrogenase) and III (cytochrome c reductase), but not II (succinate dehydrogenase), IV (cytochrome c oxidase), or V (ATP synthase). The same pattern of inhibition is observed in the case of isolated bovine heart mitochondria exposed to ionizing radiation and the nitric oxide donor. The addition of authentic peroxynitrite (ONO2(-)) to isolated mitochondria also results in damage to complexes I and III (but not II, IV, and V), as shown by assays of electron-transfer activities and electron paramagnetic resonance (EPR) spectroscopic measurements, suggesting ONO2(-) to be responsible for most of the observed radiation damage in both the cultured cell lines and isolated mitochondria. It is argued that, in general, production of ONO2(-) is an important contributor to radiation damage in biological systems and the implications of these findings in relation to possible mechanisms of oxidant-linked apoptosis are briefly considered.


Subject(s)
Mitochondria/drug effects , Mitochondria/radiation effects , NADH Dehydrogenase/metabolism , Nitric Oxide/pharmacology , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cattle , Cell Line , Electron Spin Resonance Spectroscopy , Electron Transport/drug effects , Half-Life , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/enzymology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/radiation effects , Intracellular Membranes/drug effects , Intracellular Membranes/pathology , Mitochondria/enzymology , Mitochondria/pathology , Nitrates/metabolism , Nitrates/pharmacology , Nitric Oxide/metabolism , Oxidants/metabolism , Oxidants/pharmacology , Oxidative Stress/drug effects , Rabbits , Radiation, Ionizing , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxides/metabolism
11.
Int J Cancer ; 96(1): 11-21, 2001 Feb 20.
Article in English | MEDLINE | ID: mdl-11241326

ABSTRACT

Intratracheal (IT) injection of the transgene for human manganese superoxide dismutase in plasmid/liposome (SOD2-PL) complex prior to irradiation protects C57BL/6J mice from whole lung irradiation-induced organizing alveolitis/fibrosis. Transgene mRNA was detected in alveolar type II (AT-II) and tracheobronchial tree cells explanted to culture 48 hours after gene therapy. To determine whether constitutive overexpression of murine MnSOD (Sod2) in whole lung or surfactant promoter-restricted AT-II cells (SP1)-SOD2 mice would provide intrinsic radioresistance, transgenic mice of two strains were compared with age-matched controls. Other groups of surfactant promoter-restricted (SP1)-SOD2 transgenic mice or control FeVB/NHsd mice received IT SOD2-PL gene therapy prior to irradiation. There was no significant intrinsic lung protection in either strain of MnSOD transgenic mice. The SP1-SOD2 transgenic mice were protected from lung damage by IT injection of the human SOD2-PL complex 24 hours prior to irradiation. Thus, overexpression of either human SOD2 or murine Sod2 in the lungs of transgenic mice does not provide intrinsic lung irradiation protection. The overexpression of SOD2 in the SP1-SOD2 mice may have made the mice more sensitive to irradiation.


Subject(s)
Lung/enzymology , Superoxide Dismutase/biosynthesis , Animals , Bronchi/cytology , Bronchi/radiation effects , Cells, Cultured , Dose-Response Relationship, Radiation , Fibrosis/etiology , Genetic Therapy , Humans , Liposomes/metabolism , Lung/radiation effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plasmids/metabolism , Promoter Regions, Genetic , Pulmonary Alveoli/radiation effects , RNA, Messenger/metabolism , Radiation Tolerance/genetics , Reverse Transcriptase Polymerase Chain Reaction , Superoxide Dismutase/genetics , Surface-Active Agents/metabolism , Time Factors , Trachea/cytology , Trachea/radiation effects
12.
Radiat Res ; 155(1 Pt 1): 2-14, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11121210

ABSTRACT

Radiation of the esophagus of C3H/HeNsd mice with 35 or 37 Gy of 6 MV X rays induces significantly increased RNA transcription for interleukin 1 (Il1), tumor necrosis factor alpha (Tnf), interferon gamma inducing factor (Ifngr), and interferon gamma (Ifng). These elevations are associated with DNA damage that is detectable by a comet assay of explanted esophageal cells, apoptosis of the esophageal basal lining layer cells in situ, and micro-ulceration leading to dehydration and death. The histopathology and time sequence of events are comparable to the esophagitis in humans that is associated with chemoradiotherapy of non-small cell lung carcinoma (NSCLC). Intraesophageal injection of clinical-grade manganese superoxide dismutase-plasmid/liposome (SOD2-PL) 24 h prior to irradiation produced an increase in SOD2 biochemical activity in explanted esophagus. An equivalent therapeutic plasmid weight of 10 microgram ALP plasmid in the same 500 microliter of liposomes, correlated to around 52-60% of alkaline phosphatase-positive cells in the squamous layer of the esophagus at 24 h. Administration of SOD2-PL prior to irradiation mediated a significant decrease in induction of cytokine mRNA by radiation and decreased apoptosis of squamous lining cells, micro-ulceration, and esophagitis. Groups of mice receiving 35 or 37 Gy esophageal irradiation by a technique protecting the lungs and treating only the central mediastinal area were followed to assess the long-term effects of radiation. SOD2-PL-treated irradiated mice demonstrated a significant decrease in esophageal wall thickness at day 100 compared to irradiated controls. Mice with orthotopic thoracic tumors composed of 32D-v-abl cells that received intraesophageal SOD2-PL treatment showed transgenic mRNA in the esophagus at 24 h, but no detectable human SOD2 transgene mRNA in explanted tumors by nested RT-PCR. These data provide support for translation of this strategy of SOD2-PL gene therapy to studies leading to a clinical trial in fractionated irradiation to decrease the acute and chronic side effects of radiation-induced damage to the esophagus.


Subject(s)
Cytokines/biosynthesis , Esophageal Stenosis/prevention & control , Esophagitis/prevention & control , Genetic Therapy/methods , Radiation Injuries/prevention & control , Radiation Protection/methods , Superoxide Dismutase/genetics , Animals , Apoptosis/radiation effects , Cytokines/genetics , Esophageal Stenosis/ethnology , Esophageal Stenosis/metabolism , Esophagitis/etiology , Esophagitis/metabolism , Female , Gene Expression , Humans , Liposomes , Male , Mediastinal Neoplasms/genetics , Mediastinal Neoplasms/metabolism , Mice , Mice, Inbred C3H , Plasmids , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Radiation Injuries/ethnology , Radiation Injuries/metabolism , Radiation Tolerance/genetics , Reverse Transcriptase Polymerase Chain Reaction , Superoxide Dismutase/metabolism , Transgenes
13.
Radiat Res ; 154(4): 365-74, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11023599

ABSTRACT

The pulmonary ionizing radiation sensitivity of C57BL/6 Sod2(+/-) mice heterozygous for MnSOD deficiency was compared to that Sod2(+/+) control littermates. Embryo fibroblast cell lines from Sod2(-/-) (neonatal lethal) or Sod2(+/-) mice produced less biochemically active MnSOD and demonstrated a significantly greater in vitro radiosensitivity. No G(2)/M-phase cell cycle arrest after 5 Gy was observed in Sod2(-/-) cells compared to the Sod2(+/-) or Sod2(+/+) lines. Subclonal Sod2(-/-) or Sod2(+/-) embryo fibroblast lines expressing the human SOD2 transgene showed increased biochemical activity of MnSOD and radioresistance. Sod2(+/-) mice receiving 18 Gy whole-lung irradiation died sooner and had an increased percentage of lung with organizing alveolitis between 100 and 160 days compared to Sod2(+/+) wild-type littermates. Both Sod2(+/-) and Sod2(+/+) littermates injected intratracheally with human manganese superoxide dismutase-plasmid/liposome (SOD2-PL) complex 24 h prior to whole-lung irradiation showed decreased DNA strand breaks and improved survival with decreased organizing alveolitis. Thus underexpression of MnSOD in the lungs of heterozygous Sod2(+/-) knockout mice is associated with increased pulmonary radiation sensitivity and parallels increased radiation sensitivity of embryo fibroblast cell lines in vitro. The restoration of cellular radioresistance in vitro and in lungs in vivo by SOD2-PL transgene expression supports a potential role for SOD2-PL gene therapy in organ-specific radioprotection.


Subject(s)
Genetic Therapy , Lung/radiation effects , Radiation Pneumonitis/therapy , Superoxide Dismutase/deficiency , Animals , Cell Cycle/radiation effects , Cell Line , Comet Assay , DNA/radiation effects , DNA Damage , Dose-Response Relationship, Radiation , Embryo, Mammalian , Fibroblasts/enzymology , Fibroblasts/radiation effects , Humans , Injections , Liposomes/administration & dosage , Lung/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress , Radiation Pneumonitis/enzymology , Radiation Pneumonitis/etiology , Radiation Pneumonitis/genetics , Radiation Tolerance/genetics , Reactive Oxygen Species , Reverse Transcriptase Polymerase Chain Reaction , Species Specificity , Superoxide Dismutase/genetics , Trachea , Transgenes
14.
Int J Cancer ; 90(3): 128-37, 2000 Jun 20.
Article in English | MEDLINE | ID: mdl-10900424

ABSTRACT

Esophagitis is a major limiting factor in the treatment of lung cancer by radiation alone or in combination with chemotherapy. We have previously demonstrated that intraesophageal injection of manganese superoxide dismutase-plasmid/liposome (MnSOD-PL) complex into C3H/HeNsd mice blocks irradiation-induced esophagitis. To determine whether the human esophagus can be similarly transfected, normal human esophageal sections obtained from the margins of esophagectomy specimens from esophageal cancer patients were transfected in vitro with alkaline phosphatase (AlkP)-PL complex and stained for AlkP activity, and the percent of cells expressing AlkP was calculated. At 24 hr after transfection with 20 or 200 microgram of AlkP-PL complex, 55.0% and 85.8% of esophageal epithelial cells expressed detectable AlkP, respectively. Other sections transfected with MnSOD-PL complex showed transgene mRNA by nested reverse transcriptase-polymerase chain reaction (RT-PCR) assay and increased MnSOD biochemical activity for at least 96 hr after transfection. Irradiated MnSOD-PL complex-transfected sections demonstrated a significantly decreased percentage of apoptotic cells when compared to irradiated control sections. Following 1,000 cGy, MnSOD-PL-treated samples showed 7.5 +/- 2.8% and 33.3 +/- 7.3% apoptotic cells at 24 and 48 hr compared to 53.6 +/- 6.9% and 59.0 +/- 13.8% for nontransfected controls (P < 0.0001 and P < 0.1175). After 2,000 cGy, results at 24 and 48 hr were 25.0 +/- 7.6% and 66.9 +/- 4.9% for MnSOD-transfected sections compared to 65.6 +/- 4.3% and 90.0 +/- 4.1% for control sections (P < 0.0001 and P = 0.0353), respectively. Thus, human esophageal sections can be transfected with MnSOD-PL complex in vitro and thereby protected against ionizing irradiation-induced apoptosis. Int. J. Cancer (Radiat. Oncol. Invest.) 90, 128-137 (2000).


Subject(s)
Apoptosis/radiation effects , Esophagus/radiation effects , Genetic Therapy , Radiation Protection , Superoxide Dismutase/genetics , Alkaline Phosphatase/genetics , Animals , Esophagus/enzymology , Humans , Liposomes , Mice , Mice, Inbred C3H , Plasmids , Transgenes
15.
Radiat Res ; 154(1): 73-86, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10856968

ABSTRACT

Reverse transcription-polymerase chain reaction and immunofluorescence analysis of D2XRII murine bone marrow stromal cells showed that gamma irradiation with doses of 2-50 Gy from (137)Cs stimulated expression of nitric oxide synthase 2 (Nos2, also known as iNos). The activation of Nos2 was accompanied by an increase in the fluorescence of 4,5-diaminofluorescein diacetate, a nitric oxide trap, and accumulation of 3-nitrotyrosine within cellular proteins in a dose-dependent manner. These effects were inhibited by actinomycin D and by N-[3-(aminomethyl)benzyl]acetamidine dihydrochloride, a specific inhibitor of Nos2. The induction of Nos2 expression and Nos2-dependent release of nitric oxide in D2XRII cells was observed within 24 h after irradiation and was similar in magnitude to that observed in cultures incubated with Il1b and Tnf. We conducted (1) confocal fluorescence imaging of 3-nitrotyrosine in bone marrow cells of irradiated C57BL/6J mice and (2) 3-nitrotyrosine fluorescence imaging of FDC-P1JL26 hematopoietic cells that were cocultured with previously irradiated D2XRII bone marrow stromal cells. Exposure to ionizing radiation increased the production of 3-nitrotyrosine in irradiated bone marrow cells in vivo and in nonirradiated FDC-P1JL26 cells cocultured with irradiated D2XRII cells for 1 or 4 h. We suggest that nitrative/oxidative stress to the transplanted multilineage hematopoietic cells due to exposure to nitric oxide released by host bone marrow stromal cells may contribute to the genotoxic events associated with malignant alterations in bone marrow tissue of transplant recipients who are prepared for engraftment by total-body irradiation.


Subject(s)
Bone Marrow Cells/radiation effects , Nitric Oxide Synthase/metabolism , Tyrosine/analogs & derivatives , Animals , Bone Marrow Cells/enzymology , Cell Communication , Enzyme Activation , Fluorescent Antibody Technique , Hematopoietic Stem Cells/physiology , Humans , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Nitric Oxide Synthase/analysis , Nitric Oxide Synthase Type II , Radiation Dosage , Radiation, Ionizing , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/enzymology , Stromal Cells/radiation effects , Tyrosine/metabolism
16.
Gene Ther ; 7(12): 1011-8, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10871749

ABSTRACT

To determine whether intratracheal (IT) lung protective manganese superoxide-plasmid/liposomes (MnSOD-PL) complex provided 'bystander' protection of thoracic tumors, mice with orthotopic Lewis lung carcinoma-bacterial beta-galactosidase gene (3LL-LacZ) were studied. There was no significant difference in irradiation survival of 3LL-LacZ cells irradiated, then cocultured with MnSOD-PL-treated compared with control lung cells (D0 2.022 and 2.153, respectively), or when irradiation was delivered 24 h after coculture (D0 0.934 and 0.907, respectively). Tumor-bearing control mice showed 50% survival at 18 days and 10% survival at 21 days. Mice receiving liposomes with no insert or LacZ-PL complex plus 18 Gy had 50% survival at 22 days, and a 20% and 30% survival at day 50, respectively. Mice receiving MnSOD-PL complex followed by 18 Gy showed prolonged survival of 45% at 50 days after irradiation (P < 0.001). Nested RT-PCR assay for the human MnSOD transgene demonstrated expression at 24 h in normal lung, but not in orthotopic tumors. Decreased irradiation induction of TGF-beta1, TGF-beta2, TGF-beta3, MIF, TNF-alpha, and IL-1 at 24 h was detected in lungs, but not orthotopic tumors from MnSOD-PL-injected mice (P < 0.001). Thus, pulmonary radioprotective MnSOD-PL therapy does not provide detectable 'bystander' protection to thoracic tumors.


Subject(s)
Carcinoma, Lewis Lung/pathology , Genetic Therapy/methods , Lung Neoplasms/pathology , Lung/radiation effects , Radiation Injuries, Experimental/prevention & control , Superoxide Dismutase/genetics , Animals , Cell Survival/radiation effects , Cytokines/genetics , Cytokines/metabolism , Female , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Liposomes , Mice , Mice, Inbred C57BL , Plasmids , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Radiation Tolerance , Tumor Cells, Cultured
17.
Radiat Oncol Investig ; 7(4): 204-17, 1999.
Article in English | MEDLINE | ID: mdl-10492161

ABSTRACT

Esophagitis is a major toxicity of radiation therapy for nonsmall-cell lung cancer. Intraesophageal injection of manganese superoxide dismutase (MnSOD) plasmid/liposome complexes (1 mg of the pRK5-MnSOD plasmid containing the human MnSOD transgene in a 0.15 ml volume of lipofectin) before irradiation was carried out to attempt to prevent irradiation esophagitis. In control noninjected male C3H/HeNsd mice, esophagitis was induced by single fraction 3,500 cGy irradiation. Histopathology at 4 days revealed vacuole formation in squamous lining cells, separation of the squamous layer from the underlying muscle layer, ulceration at 7 days, and dehydration and death by 30 days. MnSOD plasmid/liposome complex-injected mice showed transcription of the human MnSOD transgene message in esophageal squamous lining cells by nested reverse transcriptase-polymerase chain reaction (RT-PCR) increased MnSOD biochemical activity 24 h after injection, decreased vacuole formation at day 4 (P < 0.001) after 3,500 cGy thoracic irradiation, and improved survival (P = 0.0009). In contrast, groups of mice receiving LacZ (bacterial beta-galactosidase gene) plasmid/liposome complexes or liposomes containing no DNA before 3,500 cGy irradiation showed an unaltered irradiation histopathology and decreased survival. Mice receiving intraesophageal MnSOD plasmid/liposomes followed 8 h later by human equivalent doses of Taxol (1.4 mg/kg) and carboplatin (2.5 mg/kg), then 15 h later 3,300 cGy irradiation, showed increased survival, compared with irradiated control or LacZ plasmid/liposome groups. Thus, overexpression of the human MnSOD transgene in the esophagus can prevent irradiation-induced esophagitis in the mouse model.


Subject(s)
Esophagitis/prevention & control , Esophagus/radiation effects , Gene Expression Regulation, Enzymologic , Genetic Therapy , Radiation Injuries, Experimental/prevention & control , Superoxide Dismutase/genetics , Animals , DNA/genetics , DNA Primers/chemistry , Drug Delivery Systems , Esophagitis/enzymology , Esophagitis/etiology , Esophagitis/pathology , Esophagus/enzymology , Liposomes , Male , Mice , Mice, Inbred C3H , Plasmids , Polymerase Chain Reaction , Radiation Injuries, Experimental/enzymology , Radiation Injuries, Experimental/etiology , Radiation Injuries, Experimental/pathology , Superoxide Dismutase/administration & dosage , Superoxide Dismutase/metabolism , Transgenes
18.
Biol Blood Marrow Transplant ; 5(4): 204-14, 1999.
Article in English | MEDLINE | ID: mdl-10465100

ABSTRACT

Radiation pneumonitis remains a critical dose-limiting toxicity of total body irradiation (TBI) for use in bone marrow transplantation. The acute and chronic phases of radiation damage in the mouse lung have been shown to correlate with mouse strain genotype and are dependent on fraction size, total dose, and total lung volume. Our prior studies demonstrated effective prevention of irradiation-induced lung damage and improved survival in C57BL/6J mice by MnSOD plasmid/liposome gene therapy. In the present studies, we investigated the kinetics of irradiation-induced upregulation of mRNA for acute phase cytokines interleukin (IL)-1 and tumor necrosis factor (TNF)-alpha, and fibrosis-associated transforming growth factor (TGF)-beta and isoforms (TGF-beta1, TGF-beta2 and TGF-beta3) in 2000 cGy whole-lung irradiated C57BL/6J mice, a strain known to develop dose and volume-dependent organizing alveolitis/fibrosis. The results demonstrate increase in mRNA for IL-1 between days 1 and 14 after irradiation with return to baseline levels out to 120 days. TNF-alpha mRNA levels were not initially elevated but increased between 80 and 100 days and then decreased by 120 days. The mRNA levels for TGF-beta1 demonstrated an initial increase within the first 14 days after total lung irradiation with a decrease to baseline levels out to 100 days. Then, in striking contrast to the other two cytokines, an increase in TGF-beta2 mRNA occurred at around 120 days and correlated with the detection of organizing alveolitis/radiation fibrosis and mortality. These results are consistent with a two-phase mechanism in the molecular pathology of irradiation lung injury, in which IL-1 cytokine mRNA levels correlated with the acute pneumonitis phase and delayed elevation of TNF-alpha (80-100 days), TGF-beta1 (100 days), and TGF-beta2 (120 days) were associated with the fibrosis phase. Insight into the cell-specific and tissue-specific molecular mechanisms of ionizing irradiation induction of mRNA for pulmonary cytokines may provide new strategies for treatment of radiation pneumonitis in TBI patients.


Subject(s)
Radiation-Protective Agents/pharmacology , Superoxide Dismutase/genetics , Superoxide Dismutase/pharmacology , Animals , Cytokines/drug effects , Cytokines/genetics , Cytokines/radiation effects , Female , Genetic Therapy , Interleukin-1/genetics , Interleukin-1/metabolism , Interleukin-1/radiation effects , Liposomes , Lung/pathology , Lung/radiation effects , Lung Diseases/etiology , Lung Diseases/genetics , Lung Injury , Mice , Mice, Inbred C57BL , Plasmids , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/pathology , Pulmonary Alveoli/radiation effects , Pulmonary Emphysema/etiology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/etiology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , RNA, Messenger/radiation effects , Superoxide Dismutase/administration & dosage , Time Factors , Transforming Growth Factor beta/drug effects , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta/radiation effects , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/radiation effects
19.
Radiat Res ; 152(1): 29-40, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10381838

ABSTRACT

To investigate the effect of Trp53 (formerly known as p53) on stromal cells of the hematopoietic microenvironment, long-term bone marrow cultures were established from mice in which the Trp53 gene had been inactivated by homologous recombination (Trp53(-/-)) or their wild-type littermates (Trp53(+/+)). Long-term bone marrow cultures from Trp53(-/-) mice continued to produce nonadherent cells for 22 weeks, while Trp53(+/+) cultures ceased production after 15 weeks. There was a significant increase in the number of nonadherent cells produced in Trp53(-/-) long-term bone marrow cultures beginning at week 9 and continuing to week 22 (P < 0.02). The Trp53(-/-) cultures also showed significantly increased cobblestone island formation indicative of early hematopoietic stem cell-containing colonies beginning at week 10 (P < 0.01). Cobblestone islands persisted until weeks 15 and 22 in Trp53(+/+) and Trp53(-/-) cultures, respectively. Co-cultivation experiments in which Trp53(+/+) Sca1(+)lin- enriched hematopoietic stem cells were plated on Trp53(-/-) stromal cells showed increased cobblestone island formation compared to Trp53(-/-) Scal+lin- cells plated on Trp53(+/+) or Trp53(-/-) stromal cells. Radiation survival curves for clonal bone marrow stromal cells revealed a similar D0 for the Trp53(+/+) and Trp53(-/-) cell lines (1.62 +/- 0.16 and 1.49 +/- 0. 08 Gy, respectively; P = 0.408), and similar n (8.60 +/- 3.23 and 10.71 +/- 0.78, respectively) (P = 0.491). Cell cycle analysis demonstrated a G2/M-phase arrest that occurred 6 h after irradiation for both Trp53(+/+) and Trp53(-/-) stromal cell lines. After 10 Gy irradiation, there was no significant increase in the frequency of apoptosis detected in Trp53(+/+) compared to Trp53(-/-) marrow stromal cell lines. In the stromal cell lines, ICAM-1 was constitutively expressed on Trp53(+/+) but not Trp53(-/-) cells; however, a 24-h exposure to TNF-alpha induced detectable ICAM-1 on Trp53(-/-) cells and increased expression on Trp53(+/+) cells. To test the effect of Trp53 on the radiation biology of hematopoietic progenitor cells, the 32D cl 3 cell line was compared with a subclone in which expression of an E6 inserted transgene accelerates ubiquitin-dependent degradation of Trp53, thus preventing accumulation of Trp53 after genotoxic stress. The radiation survival curves were similar with no significant difference in the D0 or n, or in the percentage of cells undergoing apoptosis after 10 Gy irradiation between the two cell lines. Cells of the 32D-E6 cell line displayed a G2/M-phase arrest 6 h after 10 Gy, while cells of the parent line exhibited both a G2/M-phase arrest and a G1-phase arrest at 24 and 48 h. The results suggest a complex mechanism of action of Trp53 on the interactions between stromal and hematopoietic cells in long-term bone marrow cultures.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Cells/radiation effects , Cell Cycle/radiation effects , Genes, p53 , Tumor Suppressor Protein p53/physiology , Animals , Bone Marrow Cells/physiology , Cell Adhesion , Cell Line , Cell Survival/radiation effects , Crosses, Genetic , Female , Hematopoietic Stem Cells/cytology , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/genetics , Kinetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Recombination, Genetic , Stromal Cells , Time Factors , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
20.
Int J Radiat Oncol Biol Phys ; 43(1): 169-81, 1999 Jan 01.
Article in English | MEDLINE | ID: mdl-9989524

ABSTRACT

PURPOSE: A dose and volume limiting factor in radiation treatment of thoracic cancer is the development of fibrosis in normal lung. The goal of the present study was to determine whether expression prior to irradiation of a transgene for human manganese superoxide dismutase (MnSOD) or human copper/zinc superoxide dismutase (Cu/ZnSOD) protects against irradiation-induced lung damage in mice. METHODS AND MATERIALS: Athymic Nude (Nu/J) mice were intratracheally injected with 10(9) plaque-forming units (PFU) of a replication-incompetent mutant adenovirus construct containing the gene for either human MnSOD, human copper/zinc superoxide dismutase (Cu/ZnSOD) or LacZ. Four days later the mice were irradiated to the pulmonary cavity to doses of 850, 900, or 950 cGy. To demonstrate adenoviral infection, nested reverse transcriptase-polymerase chain reaction (RT-PCR) was carried out with primers specific for either human MnSOD or Cu/ZnSOD transgene on freshly explanted lung, trachea, or alveolar type II cells, and immunohistochemistry was used to measure LacZ expression. RNA was extracted on day 0, 1, 4, or 7 after 850 cGy of irradiation from lungs of mice that had previously received adenovirus or had no treatment. Slot blot analysis was performed to quantitate RNA expression for IL-1, tumor necrosis factor (TNF)-alpha, TGF-beta, MnSOD, or Cu/ZnSOD. Lung tissue was explanted and tested for biochemical activity of MnSOD or Cu/ZnSOD after adenovirus injection. Other mice were sacrificed 132 days after irradiation, lungs excised, frozen in OCT, (polyvinyl alcohol, polyethylene glycol mixture) sectioned, H&E stained, and evaluated for percent of the lung demonstrating organizing alveolitis. RESULTS: Mice injected intratracheally with adenovirus containing the gene for human MnSOD had significantly reduced chronic lung irradiation damage following 950 cGy, compared to control mice or mice injected with adenovirus containing the gene for human Cu/ZnSOD or LacZ. Immunohistochemistry for LacZ protein in adenovirus LacZ (Ad-LacZ)-injected mice demonstrated expression of LacZ in both the upper and lower airway. Nested RT-PCR showed lung expression of MnSOD and Cu/ZnSOD for at least 11 days following infection with each respective adenovirus construct. Nested RT-PCR using primers specific for human MnSOD demonstrated increased expression of the human MnSOD transgene in the trachea and alveolar type II cells 4 days after virus injection on the day of irradiation. At this time point, increased biochemical activity of MnSOD and Cu/ZnSOD respectively, was detected in lungs from these two adenovirus groups, compared to each other or to control or adenovirus LacZ mice. Slot blot analysis of RNA from lungs of mice in each group following 850 cGy irradiation demonstrated decreased expression of mRNA for interleukin-I (IL-1), TNF-alpha, and transforming growth factor-beta (TGF-beta) in the MnSOD adenovirus-injected mice, compared to irradiated control, LacZ, or Cu/ZnSOD adenovirus-injected, irradiated mice. Mice receiving adenovirus MnSOD showed decreased organizing alveolitis at 132 days in all three dose groups, compared to irradiated control or Ad-LacZ, or Ad-Cu/ZnSOD mice. CONCLUSIONS: Overexpression of MnSOD in the lungs of mice prior to irradiation prevents irradiation-induced acute and chronic damage quantitated as decreased levels of mRNA for IL-1, TNF-alpha, and TGF-beta in the days immediately following irradiation, and decrease in the percent of lung demonstrating fibrosis or organizing alveolitis at 132 days. These data provide a rational basis for development of gene therapy as a method of protection of the normal lung from acute and chronic sequelae of ionizing irradiation.


Subject(s)
Adenoviridae/genetics , Cryptogenic Organizing Pneumonia/prevention & control , Genetic Therapy/methods , Radiation Pneumonitis/prevention & control , Superoxide Dismutase/genetics , Transgenes , Animals , Gene Expression , Genetic Vectors/administration & dosage , Humans , Lung/metabolism , Lung/radiation effects , Mice , Mice, Nude , RNA, Messenger/metabolism , Superoxide Dismutase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...