Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
Add more filters











Publication year range
1.
Gene Ther ; 23(6): 500-9, 2016 06.
Article in English | MEDLINE | ID: mdl-26934099

ABSTRACT

Our previous work showed that a Sca-1(+) cell-based FGF2 therapy was capable of promoting robust increases in trabecular bone formation and connectivity on the endosteum of long bones. Past work reported that administration of FGF2 protein promoted bone formation in red marrow but not in yellow marrow. The issue as to whether the Sca-1(+) cell-based FGF2 therapy is effective in yellow marrow is highly relevant to its clinical potential for osteoporosis, as most red marrows in a person of an advanced age are converted to yellow marrows. Accordingly, this study sought to compare the osteogenic effects of this stem cell-based FGF2 therapy on red marrow-filled lumbar vertebrae with those on yellow marrow-filled caudal vertebrae of young adult W(41)/W(41) mice. The Sca-1(+) cell-based FGF2 therapy drastically increased trabecular bone formation in lumbar vertebrae, but the therapy not only did not promote bone formation but instead caused substantial loss of trabecular bone in caudal vertebrae. The lack of an osteogenic response was not due to insufficient engraftment of FGF2-expressing Sca-1(+) cells or inadequate FGF2 expression in caudal vertebrae. Previous studies have demonstrated that recipient mice of this stem cell-based FGF2 therapy developed secondary hyperparathyroidism and increased bone resorption. Thus, the loss of bone mass in caudal vertebrae might in part be due to an increase in resorption without a corresponding increase in bone formation. In conclusion, the Sca-1(+) cell-based FGF2 therapy is osteogenic in red marrow but not in yellow marrow.


Subject(s)
Antigens, Ly/genetics , Antigens, Ly/metabolism , Fibroblast Growth Factor 2/genetics , Genetic Therapy/methods , Membrane Proteins/genetics , Membrane Proteins/metabolism , Animals , Bone Marrow Cells/metabolism , Bone Marrow Transplantation/methods , Cancellous Bone/cytology , Cancellous Bone/transplantation , Caspase 3/genetics , Female , Fibroblast Growth Factor 2/administration & dosage , Fibroblast Growth Factor 2/biosynthesis , Fibroblast Growth Factor 2/blood , Humans , Lumbar Vertebrae , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Osteogenesis/genetics , Osteomalacia/etiology , Osteomalacia/genetics , Stem Cell Transplantation/methods
2.
Free Radic Res ; 48(8): 890-7, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24796731

ABSTRACT

There is evidence that space flight condition-induced biological damage is associated with increased oxidative stress and extracellular matrix (ECM) remodeling. To explore possible mechanisms, changes in gene expression profiles implicated in oxidative stress and in ECM remodeling in mouse skin were examined after space flight. The metabolic effects of space flight in skin tissues were also characterized. Space Shuttle Atlantis (STS-135) was launched at the Kennedy Space Center on a 13-day mission. Female C57BL/6 mice were flown in the STS-135 using animal enclosure modules (AEMs). Within 3-5 h after landing, the mice were euthanized and skin samples were harvested for gene array analysis and metabolic biochemical assays. Many genes responsible for regulating production and metabolism of reactive oxygen species (ROS) were significantly (p < 0.05) altered in the flight group, with fold changes >1.5 compared to AEM control. For ECM profile, several genes encoding matrix and metalloproteinases involved in ECM remodeling were significantly up-/down-regulated following space flight. To characterize the metabolic effects of space flight, global biochemical profiles were evaluated. Of 332 named biochemicals, 19 differed significantly (p < 0.05) between space flight skin samples and AEM ground controls, with 12 up-regulated and 7 down-regulated including altered amino acid, carbohydrate metabolism, cell signaling, and transmethylation pathways. Collectively, the data demonstrated that space flight condition leads to a shift in biological and metabolic homeostasis as the consequence of increased regulation in cellular antioxidants, ROS production, and tissue remodeling. This indicates that astronauts may be at increased risk for pathophysiologic damage or carcinogenesis in cutaneous tissue.


Subject(s)
Skin/metabolism , Skin/pathology , Space Flight , Animals , Extracellular Matrix/metabolism , Female , Metabolomics , Mice , Mice, Inbred C57BL , Oxidative Stress/physiology
3.
Radiat Res ; 180(2): 166-76, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23883319

ABSTRACT

The effectiveness of simulated solar particle event (SPE) proton radiation to induce retching and vomiting was evaluated in the ferret experimental animal model. The endpoints measured in the study included: (1) the fraction of animals that retched or vomited, (2) the number of retches or vomits observed, (3) the latency period before the first retch or vomit and (4) the duration between the first and last retching or vomiting events. The results demonstrated that γ ray and proton irradiation delivered at a high dose rate of 0.5 Gy/min induced dose-dependent changes in the endpoints related to retching and vomiting. The minimum radiation doses required to induce statistically significant changes in retching- and vomiting-related endpoints were 0.75 and 1.0 Gy, respectively, and the relative biological effectiveness (RBE) of proton radiation at the high dose rate did not significantly differ from 1. Similar but less consistent and smaller changes in the retching- and vomiting-related endpoints were observed for groups irradiated with γ rays and protons delivered at a low dose rate of 0.5 Gy/h. Since this low dose rate is similar to a radiation dose rate expected during a SPE, these results suggest that the risk of SPE radiation-induced vomiting is low and may reach statistical significance only when the radiation dose reaches 1 Gy or higher.


Subject(s)
Gamma Rays/adverse effects , Protons/adverse effects , Radiation Injuries, Experimental/etiology , Solar Activity , Vomiting/etiology , Animals , Disease Models, Animal , Dose-Response Relationship, Radiation , Female , Ferrets , Radiation Injuries, Experimental/physiopathology , Random Allocation , Relative Biological Effectiveness
4.
Free Radic Res ; 47(10): 811-20, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23869974

ABSTRACT

This study evaluated liver from C57BL/6 mice irradiated with low-dose/low-dose-rate (LDR) γ-rays (0.01 Gy, 0.03 cGy/h), with and without subsequent exposure to acute 2 Gy gamma or proton radiation. Analyses were performed on day 56 post-exposure. Expression patterns of apoptosis-related genes were strikingly different among irradiated groups compared with 0 Gy (p < 0.05). Two genes were affected in the Gamma group, whereas 10 were modified in the LDR + Gamma group. In Proton and LDR + Proton groups, there were six and 12 affected genes, respectively. Expression of genes in the Gamma (Traf3) and Proton (Bak1, Birc2, Birc3, Mcl1) groups was no longer different from 0 Gy control group when mice were pre-exposed to LDR γ-rays. When each combined regimen was compared with the corresponding group that received acute radiation alone, two genes in the LDR + Gamma group and 17 genes in the LDR + Proton group were modified; greatest effect was on Birc2 and Nol3 (> 5-fold up-regulated by LDR + Protons). Oxygen radical production in livers from the LDR + Proton group was higher in LDR, Gamma, and LDR + Gamma groups (p < 0.05 vs. 0 Gy), but there were no differences in phagocytosis of E. coli. Sections stained with hematoxylin and eosin (H&E) suggested more inflammation, with and without necrosis, in some irradiated groups. The data demonstrate that response to acute radiation is dependent on radiation quality and regimen and that some LDR γ-ray-induced modifications in liver response were still evident nearly 2 months after exposure.


Subject(s)
Gamma Rays , Liver/radiation effects , Protons , Animals , Apoptosis/radiation effects , Dose-Response Relationship, Radiation , Female , Gene Expression/radiation effects , Liver/metabolism , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Respiratory Burst/radiation effects
5.
Radiat Res ; 180(1): 7-16, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23672458

ABSTRACT

Acute radiation sickness (ARS) is expected to occur in astronauts during large solar particle events (SPEs). One parameter associated with ARS is the hematopoietic syndrome, which can result from decreased numbers of circulating blood cells in those exposed to radiation. The peripheral blood cells are critical for an adequate immune response, and low blood cell counts can result in an increased susceptibility to infection. In this study, Yucatan minipigs were exposed to proton radiation within a range of skin dose levels expected for an SPE (estimated from previous SPEs). The proton-radiation exposure resulted in significant decreases in total white blood cell count (WBC) within 1 day of exposure, 60% below baseline control value or preirradiation values. At the lowest level of the blood cell counts, lymphocytes, neutrophils, monocytes and eosinophils were decreased up to 89.5%, 60.4%, 73.2% and 75.5%, respectively, from the preirradiation values. Monocytes and lymphocytes were decreased by an average of 70% (compared to preirradiation values) as early as 4 h after radiation exposure. Skin doses greater than 5 Gy resulted in decreased blood cell counts up to 90 days after exposure. The results reported here are similar to studies of ARS using the nonhuman primate model, supporting the use of the Yucatan minipig as an alternative. In addition, the high prevalence of hematologic abnormalities resulting from exposure to acute, whole-body SPE-like proton radiation warrants the development of appropriate countermeasures to prevent or treat ARS occurring in astronauts during space travel.


Subject(s)
Acute Radiation Syndrome/blood , Leukocytes/radiation effects , Solar Activity , Animals , Astronauts , Dose-Response Relationship, Radiation , Hematopoietic System/radiation effects , Humans , Leukocyte Count , Protons , Radiation, Ionizing , Swine , Swine, Miniature/blood
6.
Technol Cancer Res Treat ; 11(5): 447-57, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22475066

ABSTRACT

Due to radiation-induced immune depression and development of pathologies such as cancer, there is increasing urgency to identify radiomitigators that are effective when administered after radiation exposure. The main goal of this study was to determine the radiomitigation capacity of MnTE-2-PyP[Mn(III) tetrakis (N-ethylpyridinium-2-yl) porphyrin], a superoxide dismutase (SOD) mimetic, and evaluate leukocyte parameters in spleen and blood. C57BL/6 mice were total-body exposed to 2 Gy γ-rays (Co-60), i.e., well below a lethal dose, followed by subcutaneous implantation of 5 × 10(5) RM-9 prostate tumor cells and initiation of MnTE-2-PyP treatment (day 0); interval between each procedure was 1-2 h. The drug was administered daily (12 times). Tumor progression was monitored and immunological analyses were performed on a subset per group on day 12. Animals treated with MnTE-2-PyP alone had significantly slower tumor growth compared to mice that did not receive the drug (P < 0.05), while radiation alone had no effect. Treatment of tumor-bearing mice with MnTE-2-PyP alone significantly increased spleen mass relative to body mass; the numbers of splenic white blood cells (WBC) and lymphocytes (B and T), as well as circulating WBC, granulocytes, and platelets, were high compared to one of more of the other groups (P < 0.05). The results show that MnTE-2-PyP slowed RM-9 tumor progression and up-regulated immune parameters, but mitigation of the effects of 2 Gy total-body irradiation were minimal.


Subject(s)
Antioxidants/administration & dosage , Metalloporphyrins/administration & dosage , Prostatic Neoplasms/radiotherapy , Radiation-Protective Agents/administration & dosage , Animals , Blood Cell Count , Male , Metalloporphyrins/chemistry , Mice , Mice, Inbred C57BL , Molecular Mimicry , Organ Size , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Radiation Injuries, Experimental/prevention & control , Spleen/drug effects , Spleen/pathology , Spleen/radiation effects , Superoxide Dismutase/chemistry , Tumor Burden/drug effects , Tumor Burden/radiation effects , Whole-Body Irradiation , Xenograft Model Antitumor Assays
7.
Curr Eye Res ; 34(1): 62-72, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19172472

ABSTRACT

PURPOSE: The purpose of this study was to evaluate the efficacy of the antioxidant Mn (III) tetrakis (N-ethylpyridinium-2-yl) porphyrin (MnTE-2-PyP) in protecting ocular tissue and retinal microvasculature from radiation damage. MATERIALS AND METHODS: 75 rats were treated with Mn TE-2-PyP at 2.5 micro g/injection into one eye an hour before proton irradiation. The radiation was delivered in a single fraction to total doses of 8 Gray (Gy) or 28 Gy; Rats were sacrificed 3 days and 3, 6, 9, and 12 months thereafter for histology and quantification of photoreceptor cell populations and retinal capillary changes. RESULTS: By 6 months following radiation, there was significant loss of retinal outer and inner nuclear layers in eyes receiving radiation only (8 and 28 Gy) (p < 0.05) compared to their controls and to the eyes of rats treated with radiation plus metalloporphyrin. Retinal microvessel length density decreased significantly 6 months following 28 Gy (p < 0.05) compared to their controls and to MnTE-2-PyP treated rats. By 12 months following irradiation, irradiated eyes showed extensive damage to the photoreceptor layer, whereas the eyes of animals receiving radiation plus MnTE-2-PyP showed almost no morphological damage. MnTE-2-PyP treatment also suppressed radiation-induced apoptosis in our study. CONCLUSIONS: These results demonstrated that MnTE-2-PyP protected both photoreceptors and retinal capillaries from radiation damage, suggesting that this metalloporphyrin antioxidant is effective in regulating the damage induced by proton radiation.


Subject(s)
Antioxidants/therapeutic use , Metalloporphyrins/therapeutic use , Radiation Injuries, Experimental/prevention & control , Radiation-Protective Agents/therapeutic use , Retinal Diseases/prevention & control , Animals , Apoptosis , Caspase 3/metabolism , Cataract/classification , Cataract/etiology , Cataract/prevention & control , Fluorescent Antibody Technique, Indirect , Lens, Crystalline/radiation effects , Male , Photoreceptor Cells, Vertebrate/enzymology , Photoreceptor Cells, Vertebrate/pathology , Photoreceptor Cells, Vertebrate/radiation effects , Protons , Radiation Dosage , Radiation Injuries, Experimental/etiology , Radiation Injuries, Experimental/pathology , Rats , Rats, Sprague-Dawley , Retina/radiation effects , Retinal Diseases/etiology , Retinal Diseases/pathology , Retinal Vessels/pathology , Retinal Vessels/radiation effects , Treatment Outcome
8.
Radiat Res ; 170(2): 201-7, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18666808

ABSTRACT

High-energy, high-charge (HZE) radiation, including iron ions ((56)Fe(26+)), is a component of the space environment. We recently observed a profound loss of trabecular bone in mice after whole-body HZE irradiation. The goal of this study was to examine morphology in bones that were excluded from a (56)Fe(26+) beam used to irradiate the body. Using 10-week-old male Sprague-Dawley rats and excluding the hind limbs and pelvis, we irradiated animals with 0, 1, 2 and 4 Gy (56)Fe(26+) ions and killed them humanely after 9 months. Animals grew throughout the experiment. Trabecular bone volume, connectivity and thickness within the proximal tibiae were significantly lower than control in a dose-dependent manner. Irradiated animals generally had less body mass than controls, which largely accounted for the variability in bone parameters as determined by ANCOVA. Likewise, lower cortical parameters were associated with reduced mass. However, lesser trabecular thickness in the 4-Gy group could not be attributed to body mass alone. Indicators of bone metabolism were generally unchanged, suggesting stabilized turnover. Exposure to (56)Fe(26+) ions can alter trabecular microarchitecture in shielded bones. Reduced body mass seems to be correlated with these deficits of trabecular and cortical bone.


Subject(s)
Body Weight/physiology , Body Weight/radiation effects , Iron Radioisotopes , Tibia/physiology , Tibia/radiation effects , Whole-Body Irradiation , Animals , Dose-Response Relationship, Radiation , Heavy Ions , Male , Radiation Dosage , Radiography , Rats , Rats, Sprague-Dawley , Tibia/diagnostic imaging
9.
J Appl Physiol (1985) ; 101(3): 789-93, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16741258

ABSTRACT

Cancer patients receiving radiation therapy are exposed to photon (gamma/X-ray), electron, and less commonly proton radiation. Similarly, astronauts on exploratory missions will be exposed to extended periods of lower-dose radiation from multiple sources and of multiple types, including heavy ions. Therapeutic doses of radiation have been shown to have deleterious consequences on bone health, occasionally causing osteoradionecrosis and spontaneous fractures. However, no animal model exists to study the cause of radiation-induced osteoporosis. Additionally, the effect of lower doses of ionizing radiation, including heavy ions, on general bone quality has not been investigated. This study presents data developing a murine model for radiation-induced bone loss. Female C57BL/6 mice were exposed to gamma, proton, carbon, or iron radiation at 2-Gray doses, representing both a clinical treatment fraction and spaceflight exposure for an exploratory mission. Mice were euthanized 110 days after irradiation. The proximal tibiae and femur diaphyses were analyzed using microcomputed tomography. Results demonstrate profound changes in trabecular architecture. Significant losses in trabecular bone volume fraction were observed for all radiation species: gamma, (-29%), proton (-35%), carbon (-39%), and iron (-34%). Trabecular connectivity density, thickness, spacing, and number were also affected. These data have clear implications for clinical radiotherapy in that bone loss in an animal model has been demonstrated at low doses. Additionally, these data suggest that space radiation has the potential to exacerbate the bone loss caused by microgravity, although lower doses and dose rates need to be studied.


Subject(s)
Cosmic Radiation/adverse effects , Disease Models, Animal , Heavy Ions/adverse effects , Osteoradionecrosis/etiology , Osteoradionecrosis/physiopathology , Radiotherapy/adverse effects , Animals , Calcification, Physiologic/radiation effects , Dose-Response Relationship, Drug , Environmental Exposure/adverse effects , Female , Heavy Ion Radiotherapy , Mice , Mice, Inbred C57BL , Osteoporosis/etiology , Osteoporosis/physiopathology , Radiation Dosage , Radiation Injuries, Experimental/etiology , Radiation Injuries, Experimental/physiopathology , Risk Assessment/methods , Risk Factors
10.
Phys Med Biol ; 49(16): 3817-30, 2004 Aug 21.
Article in English | MEDLINE | ID: mdl-15446807

ABSTRACT

Standard methods for risk assessments resulting from human exposures to mixed radiation fields in Space consisting of different particle types and energies rely upon quality factors. These are generally defined as a function of linear energy transfer (LET) and are assumed to be proportional to the risk. In this approach, it is further assumed that the risks for single exposures from each of the radiation types add linearly. Although risks of cancer from acute exposures to photon radiations have been measured in humans, quality factors for protons and ions of heavier atomic mass are generally inferred from animal and/or cellular data. Because only a small amount of data exists for such particles, this group has been examining tumourigenesis initiated by energetic protons and iron ions. In this study, 741 female Sprague-Dawley rats were irradiated or sham irradiated at approximately 60 days of age with 250 MeV protons, 1 GeV/nucleon iron ions or both protons and iron ions. The results suggest that the risk of mammary tumours in the rats sequentially irradiated with 1 GeV/nucleon 56Fe ions and 250 MeV protons is less than additive. These data in conjunction with earlier results further suggest that risk assessments in terms of only mean LETs of the primary cosmic rays may be insufficient to accurately evaluate the relative risks of each type of particle in a radiation field of mixed radiation qualities.


Subject(s)
Harderian Gland/pathology , Mammary Neoplasms, Animal/etiology , Neoplasms, Radiation-Induced , Radiometry , Animals , Dose-Response Relationship, Radiation , Female , Harderian Gland/radiation effects , Ions , Linear Energy Transfer , Models, Statistical , Photons , Protons , Rats , Rats, Sprague-Dawley , Risk , Time Factors
11.
J Interferon Cytokine Res ; 21(11): 885-97, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11747620

ABSTRACT

Recent reports have shown that tumor necrosis factor-alpha (TNF-alpha) can augment the effects of radiation against certain tumor types. However, the high concentrations of intravenous infusion of TNF-alpha needed to cause tumor regression can induce many systemic side effects. The aims of this study were to determine if TNF-alpha encapsulated in sterically stabilized (Stealth, ALZA Corporation, Mountain View, CA), PEGylated liposomes (SL) augments the antitumor effects of radiation and to compare its efficacy and possible toxicity with free TNF-alpha in the LS174T human colon tumor xenograft model. Nude mice were injected subcutaneously (s.c.) with LS174T cells and treated intravenously (i.v.) with Stealth-liposomal TNF-alpha (SL-TNF-alpha) with and without radiation or TNF-alpha with or without radiation when tumor size was approximately 200 mm(3). In phase 1, a significant decrease (p = 0.047) in tumor growth was observed with radiation at day 21 but not with SL-TNF-alpha or free TNF-alpha alone. By the end of phase 1 (day 27) with continued treatments, the SL-TNF-alpha plus radiation group had significantly smaller tumors (p = 0.044) than those in the free TNF-alpha plus radiation group. In phase 2, where a similar tumor growth reduction pattern was observed, the addition of TNF-alpha to radiation, either as free protein or within SL, increased lymphocyte activation and natural killer (NK) cell numbers in both blood and spleen. The effect was generally more pronounced with SL-TNF-alpha. Systemic toxicity, based on hematologic analyses and body weight, was absent or minimal. Collectively, the data show that pretreatment with SL-TNF-alpha can enhance more effectively, and possibly more safely, the effects of radiation against human colon tumor xenografts than can free TNF-alpha and that the increased antitumor action may involve upregulation of lymphocytes.


Subject(s)
Colonic Neoplasms/drug therapy , Colonic Neoplasms/radiotherapy , Radiation-Sensitizing Agents/therapeutic use , Tumor Necrosis Factor-alpha/therapeutic use , Animals , Blood Cell Count , Body Weight/drug effects , Body Weight/radiation effects , Cell Division/drug effects , Cell Division/radiation effects , Chemotherapy, Adjuvant , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Humans , Immunophenotyping , Kinetics , Liposomes , Lymphocyte Activation/drug effects , Lymphocyte Activation/radiation effects , Lymphocyte Subsets/classification , Lymphocyte Subsets/drug effects , Lymphocyte Subsets/radiation effects , Male , Mice , Mice, Nude , Organ Size/drug effects , Organ Size/radiation effects , Radiation-Sensitizing Agents/administration & dosage , Radiation-Sensitizing Agents/adverse effects , Spleen/immunology , Spleen/pathology , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/adverse effects , Xenograft Model Antitumor Assays
12.
Cytokine ; 15(6): 305-14, 2001 Sep 21.
Article in English | MEDLINE | ID: mdl-11594797

ABSTRACT

Single intratumoural treatment of nude mice with a vaccinia virus (VV)-expressing interleukin-1 (IL-2) or IL-12 induced significant tumour growth inhibition associated with clear signs of toxicity. At a low virus dose, only some treated animals showed signs of toxicity. We characterized and compared the activity of NK and B cells and major pro-inflammatory factors (IFN-gamma, TNF-alpha) in treated animals with and without toxicity. One week after treatment animals exhibiting signs of cytokine-related toxicity showed dramatic increases in several measured parameters. High leukocyte and lymphocyte counts in blood and marked increases in NK and CD25(+)cells in both blood and spleen were associated with IL-2-induced toxicity, while IL-12-induced toxicity was related to a great elevation of CD25(+)cells in blood and CD71(+)cells in the spleen. In contrast, immune activation in animals free of toxicity was observed on day 2 after the treatment, which drastically declined by day 7. Thus, immune responses induced by IL-2 and IL-12 therapy appear to play important roles in both tumour inhibition and the accompanying toxicity. Short-term effects induced by IL-2 and IL-12 could be critical for antitumour therapy that prolongs survival and protects from adverse side effects.


Subject(s)
Cytokines/toxicity , Genetic Therapy/methods , Interleukin-12/genetics , Interleukin-2/genetics , Neoplasms/therapy , Vaccinia virus/genetics , Animals , Antigens, CD/biosynthesis , Antigens, Differentiation, B-Lymphocyte/biosynthesis , B-Lymphocytes/metabolism , Cell Line , Cells, Cultured , Cytokines/genetics , Fibroblasts/metabolism , Flow Cytometry , Glioma/therapy , Interferon-gamma/metabolism , Killer Cells, Natural/metabolism , Leukocytes/metabolism , Lymphocytes/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , Organ Size , Rats , Receptors, Interleukin-2/biosynthesis , Receptors, Transferrin , Spleen/metabolism , Time Factors , Transgenes , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism
13.
Oncology ; 61(3): 243-53, 2001.
Article in English | MEDLINE | ID: mdl-11574781

ABSTRACT

Long-term control of high-grade brain tumors is rarely achieved with current therapeutic regimens. The major goal of this study was to determine whether polysaccharopeptide (PSP), a crude polysaccharide peptide extract derived from Coriolus versicolor, a fungus, could enhance the effects of radiation against glioma cells in culture and in xenografted tumors in vivo. PSP significantly augmented radiation-induced damage to C6 rat glioma cells in vitro. Nude mice injected subcutaneously with the C6 cells were treated with PSP (injected intraperitoneally at 2 mg/injection) and radiation (2 Gy/fraction, 8 Gy in total) using three different time-dose protocols. Tumor volumes were consistently smaller in all treated groups compared to the non-treated tumor-bearing controls except in one group which received PSP prior to tumor implantation. The administration of radiation alone resulted in the slowest tumor progression, whereas PSP alone had no effect. Furthermore, PSP in combination with radiation treatment did not increase radiation efficacy. Natural killer cell, lymphocyte and granulocyte counts in blood and spleen were significantly higher in PSP-treated animals, demonstrating that PSP has protective effects on immunological function. Collectively, these results warrant further investigation to determine if PSP can be effectively utilized to upregulate immune responsiveness in case of neoplasia and other diseases in which immunosuppression is a prominent feature.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Antineoplastic Agents/therapeutic use , Brain Neoplasms/radiotherapy , Glioma/radiotherapy , Proteoglycans/therapeutic use , Radiation-Protective Agents/therapeutic use , Radiation-Sensitizing Agents/therapeutic use , Radioisotope Teletherapy , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/toxicity , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Body Weight/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Combined Modality Therapy , DNA Replication/drug effects , Disease Progression , Glioma/drug therapy , Glioma/pathology , Glioma/therapy , Hematologic Diseases/etiology , Lymphocyte Count , Lymphocyte Subsets/drug effects , Mice , Mice, Nude , Neoplasm Transplantation , Oxidative Stress , Proteoglycans/pharmacology , Radiation-Protective Agents/pharmacology , Radiation-Sensitizing Agents/pharmacology , Radioisotope Teletherapy/adverse effects , Rats , Spleen/drug effects , Spleen/pathology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/radiation effects
14.
In Vivo ; 15(3): 209-16, 2001.
Article in English | MEDLINE | ID: mdl-11491015

ABSTRACT

The goal of part II of this study was to evaluate the effects of gamma-radiation on circulating blood cells, functional characteristics of splenocytes, and cytokine expression after whole-body irradiation at varying total doses and at low- and high-dose-rates (LDR, HDR). Young adult C57BL/6 mice (n = 75) were irradiated with either 1 cGy/min or 80 cGy/min photons from a 60Co source to cumulative doses of 0.5, 1.5, and 3.0 Gy. The animals were euthanized at 4 days post-exposure for in vitro assays. Significant dose- (but not dose-rate-) dependent decreases were observed in erythrocyte and blood leukocyte counts, hemoglobin, hematocrit, lipopolysaccharide (LPS)-induced 3H-thymidine incorporation, and interleukin-2 (IL-2) secretion by activated spleen cells when compared to sham-irradiated controls (p < 0.05). Basal proliferation of leukocytes in the blood and spleen increased significantly with increasing dose (p < 0.05). Significant dose rate effects were observed only in thrombocyte counts. Plasma levels of transforming growth factor-beta 1 (TGF-beta 1) and splenocyte secretion of tumor necrosis factor-alpha (TNF-alpha) were not affected by either the dose or dose rate of radiation. The data demonstrate that the responses of blood and spleen were largely dependent upon the total dose of radiation employed and that an 80-fold difference in the dose rate was not a significant factor in the great majority of measurements.


Subject(s)
Blood Platelets/radiation effects , Cytokines/blood , Erythrocytes/radiation effects , Leukocytes/radiation effects , Whole-Body Irradiation , Animals , Blood Platelets/cytology , Cytokines/metabolism , Dose-Response Relationship, Radiation , Erythrocyte Count , Erythrocytes/cytology , Female , Hematocrit , Hemoglobins , Interleukin-2/blood , Interleukin-2/metabolism , Leukocyte Count , Leukocytes/drug effects , Leukocytes/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mitogens/pharmacology , Phytohemagglutinins/pharmacology , Platelet Count , Spleen/cytology , Spleen/metabolism , Spleen/radiation effects , Transforming Growth Factor beta/blood , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/metabolism
15.
In Vivo ; 15(3): 195-208, 2001.
Article in English | MEDLINE | ID: mdl-11491014

ABSTRACT

The major goal of part I of this study was to compare varying doses and dose rates of whole-body gamma-radiation on lymphoid cells and organs. C57BL/6 mice (n = 75) were exposed to 0, 0.5, 1.5, and 3.0 Gy gamma-rays (60Co) at 1 cGy/min (low-dose rate, LDR) and 80 cGy/min (high-dose rate, HDR) and euthanized 4 days later. A significant dose-dependent loss of spleen mass was observed with both LDR and HDR irradiation; for the thymus this was true only with HDR. Decreasing leukocyte and lymphocyte numbers occurred with increasing dose in blood and spleen at both dose rates. The numbers (not percentages) of CD3+ T lymphocytes decreased in the blood in a dose-dependent manner at both HDR and LDR. Splenic T cell counts decreased with dose only in HDR groups; percentages increased with dose at both dose rates. Dose-dependent decreases occurred in CD4+ T helper and CD8+ T cytotoxic cell counts at HDR and LDR. In the blood the percentages of CD4+ cells increased with increasing dose at both dose rates, whereas in the spleen the counts decreased only in the HDR groups. The percentages of the CD8+ population remained stable in both blood and spleen. CD19+ B cell counts and percentages in both compartments declined markedly with increasing HDR and LDR radiation. NK1.1+ natural killer cell numbers and proportions remained relatively stable. Overall, these data indicate that the observed changes were highly dependent on the dose, but not dose rate, and that cells in the spleen are more affected by dose rate than those in blood. The results also suggest that the response of lymphocytes in different body compartments may be variable.


Subject(s)
Lymphocytes/radiation effects , Spleen/radiation effects , Whole-Body Irradiation , Animals , CD4-CD8 Ratio , CD4-Positive T-Lymphocytes/radiation effects , CD8-Positive T-Lymphocytes/radiation effects , Dose-Response Relationship, Radiation , Killer Cells, Natural/radiation effects , Linear Models , Mice , Mice, Inbred C57BL , Models, Animal , Spleen/cytology
16.
J Immunother ; 24(1): 46-57, 2001.
Article in English | MEDLINE | ID: mdl-11211148

ABSTRACT

Recombinant viruses can produce cytokines in tumors mobilizing an immune response to tumor cells. In this study, the authors investigated gene expression, in vivo antitumor efficacy, and safety of attenuated recombinant vaccinia virus (rVV) carrying murine cytokine genes interleukin (IL)-2 (rVV-mIL-2), IL-12 (rVV-mIL-12), and both IL-2 and IL-12 (rVV-2-12) in an athymic nude mice model. Significant tumor inhibition (p < 0.05) was observed in a preestablished subcutaneously implanted C6 glioma model using rVVs at doses ranging from 10(2) to 10(7) plaque forming units (PFU). An antitumor effect did not depend on the dose of the rVV-mIL-2 and rVV-mIL-12 viruses. All constructed rVVs induced a high level of cytokine expression in vitro and in vivo. Most groups injected with high doses of recombinant viruses encoding cytokine genes (10(5) to 10(7) PFU) showed signs of cytokine toxicity, whereas in the low-dose treatment groups (10(2) to 10(3) PFU) toxicity was greatly reduced. The antitumor activity of rVV-mIL-12 was associated with increases in both the percentage and number of natural killer T cells in the spleen. Local detection of interferon-y and tumor necrosis factor-alpha was also correlated with tumor growth arrest induced by the treatment. High-dose VV control vector per se induced tumor inhibition by activating Mac-1+ cells in blood, but the antitumor effect was less pronounced compared with rVV-carrying cytokine genes (p < 0.05). These results suggest that attenuated recombinant strains of VV at low doses may potentially be efficient vectors for cancer immunotherapy.


Subject(s)
Cytokines/genetics , Cytokines/therapeutic use , Genetic Therapy , Glioma/genetics , Glioma/therapy , Vaccinia virus/genetics , Vaccinia virus/immunology , Animals , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Brain Neoplasms/virology , Cell Line , Chlorocebus aethiops , Disease Models, Animal , Dose-Response Relationship, Immunologic , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Glioma/immunology , Glioma/virology , Injections, Intralesional , Interferon-gamma/biosynthesis , Interleukin-12/biosynthesis , Interleukin-12/genetics , Interleukin-2/biosynthesis , Interleukin-2/genetics , Lymphocyte Activation/genetics , Lymphocyte Subsets/immunology , Lymphocyte Subsets/virology , Male , Mice , Mice, Nude , Organ Specificity/genetics , Organ Specificity/immunology , Rats , Vaccinia virus/physiology , Virus Replication/genetics , Virus Replication/immunology
17.
Phys Med ; 17 Suppl 1: 30-2, 2001.
Article in English | MEDLINE | ID: mdl-11770533

ABSTRACT

The Loma Linda University (LLU) Radiobiology Program coordinates basic research and proton beam service activities for the university and extramural communities. The current focus of the program is on the biological and physical properties of protons and the operation of radiobiology facilities for NASA-sponsored projects. The current accelerator, supporting facilities and operations are described along with a brief review of extramural research projects supported by the program. These include space craft electronic parts and shielding testing as well as tumorigenesis and animal behavior experiments. An overview of research projects currently underway at LLU is also described. These include: 1) acute responses of the C57Bl/6 mouse immune system, 2) modulation of gene expression in the nematode C. elegans and rat thyroid cells, 3) quantitation of dose tolerance in rat CNS microvasculature, 4) behavioral screening of whole body proton and iron ion-irradiated C57Bl/6 mice, and 5) investigation of the role of cell integration into epithelial structures on responses to radiation.


Subject(s)
Heavy Ions , Protons , Radiobiology/instrumentation , Synchrotrons , Universities , Animals , Behavior, Animal/radiation effects , Caenorhabditis elegans , California , Gene Expression/radiation effects , Humans , Immune System/radiation effects , Mice , Radiotherapy, High-Energy/instrumentation , Rats , Research , United States , United States National Aeronautics and Space Administration
18.
Cancer Gene Ther ; 7(11): 1437-47, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11129286

ABSTRACT

Our previous studies have shown that vaccinia virus (VV) expressing p53, interleukin-2 (IL-2), and interleukin-12 (IL-12) results in an effective inhibition of subcutaneous glioma growth in mice. We propose that combination therapy of tumors with virus-mediated p53 and cytokine genes offers the prospect of synergistic antitumor response. In this work, the antitumor efficacy of VV-mediated combination of p53, IL-2, and IL-12 genes was evaluated in a nude mouse model. To minimize cytokine-associated toxicity, a virus dose as low as 10 plaque-forming units of VV expressing IL-2 and IL-12 per animal was used alone and together with 2 x 10(7) plaque-forming units of VV expressing p53. Intratumoral treatment of established C6 glioma with recombinant viruses rVV-p53, rVV-mIL2, rVV-mIL12, and rVV-2-12 induced the prolonged expression of p53, IL-2, IL-12, and both cytokines simultaneously. The combination of rVV-p53/rVV-mIL 2 or rVV-p53/rVV-2-12 resulted in significant tumor inhibition compared to single modality treatment (P<.05). rVV-p53/rVV-2-12 therapy was associated with significant elevation of natural killer, Mac-1+, and NKT cells in blood and interferon-gamma, and tumor necrosis factor-alpha expression in tumors. The difference in the inhibition of tumor growth between the rVV-p53/rVV-mIL2 combination and rVV-p53 was statistically insignificant. These data demonstrate that gene therapy based on VV-mediated combination of p53, IL-2, and IL-12 treatment may be a promising adjunctive strategy for glioma treatment.


Subject(s)
Brain Neoplasms/genetics , Genes, p53/genetics , Genetic Therapy/methods , Glioma/therapy , Interleukin-12/genetics , Interleukin-2/genetics , Vaccinia virus/genetics , Analysis of Variance , Animals , Apoptosis , Cell Line , Flow Cytometry , Haplorhini , In Situ Nick-End Labeling , Interferon-gamma/metabolism , Interleukin-12/biosynthesis , Interleukin-2/biosynthesis , Killer Cells, Natural/metabolism , Leukocytes/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , Rats , Recombinant Proteins/metabolism , Spleen/cytology , Time Factors , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/biosynthesis
19.
Anticancer Res ; 20(3A): 1337-42, 2000.
Article in English | MEDLINE | ID: mdl-10928041

ABSTRACT

The use of interleukin-2 (IL-2) and p53 for immunotherapy and gene therapy for cancer has shown promising results. In this study, we examined the efficacy of plasmid gene therapy utilizing murine IL-2, the wild-type (wt) human p53 gene, the combination of these genes, and the murine bax gene, which are under the control of the cytomegalovirus (CMV) immediate-early promoter, in nude mice bearing established subcutaneous C6 glioma. In vitro assays and immunocytochemical analysis for therapeutic genes demonstrated expression of the proteins in C6 transfected cells. In animal studies, significant antitumor activity was observed for the IL-2, p53/IL-2, and bax treated groups. However, no synergistic effect was observed in the p53/IL-2 combination group. Demonstrating for the first time, bax showed a significant reduction of tumor volume when compared to p53 (p < 0.02). Thus, our in vivo studies show that delivery of naked therapeutic genes is safe and results in significantly slower progression of glioma in athymic rodents.


Subject(s)
Glioma/therapy , Interleukin-2/therapeutic use , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/therapeutic use , Tumor Suppressor Protein p53/therapeutic use , Analysis of Variance , Animals , Disease Models, Animal , Gene Expression , Gene Transfer Techniques , Genetic Therapy , Glioma/genetics , Glioma/pathology , Immunotherapy , Interleukin-2/genetics , Mice , Mice, Nude , Neoplasm Transplantation , Proto-Oncogene Proteins/genetics , Rats , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , bcl-2-Associated X Protein
SELECTION OF CITATIONS
SEARCH DETAIL