RESUMO
Prostate cancer (PCa) prevalence is higher in older men and poorer coping with psychosocial stressors effect prognosis. Yet, interactions between age, stress and PCa progression are underexplored. Therefore, we characterized the effects of age and isolation combined with restraint (2 h/day) for 14 days post-tumor inoculation on behavior, tumor growth and host defense in the immunocompetent, orthotopic RM-9 murine PCa model. All mice were tumor inoculated. Isolation/restraint increased sympathetic and hypothalamic-pituitary-adrenal cortical activation, based on elevated serum 3-methoxy-4-hydroxyphenylglycol/norepinephrine ratios and corticosterone levels, respectively. Elevated zero maze testing revealed age-related differences in naïve C57Bl/6 mice, and increased anxiety-like behavior in tumor-bearing mice. In open field testing, old stressed mice were less active throughout the 30-min test than young non-stressed and stressed, and old non-stressed mice, suggesting greater anxiety in old stressed mice. Old (18 month) mice demonstrated more depression-like behavior than young mice with tail suspension testing, without effects of isolation/restraint stress. Old mice developed larger tumors, despite similar tumor expression of tumor vascular endothelial growth factor or transforming growth factor-beta1 across age. Tumor chemokine/cytokine expression, commonly prognostic for poorer outcomes, were uniquely age- and stress-dependent, underscoring the need for PCa research in old animals. Macrophages predominated in RM-9 tumors. Macrophages, and CD4+ and CD4+FoxP3+ T-cell tumor infiltration were greater in young mice than in old mice. Stress increased macrophage infiltration in old mice. Conversely, stress reduced intratumoral CD4+ and CD4+FoxP3+ T-cell numbers in young mice. CD8+ T-cell infiltration was similar across treatment groups. Our findings support that age- and psychological stress interacts to affect PCa outcomes by interfering with neural-immune mechanisms and affecting behavioral responses.
RESUMO
Energetic protons are the most abundant particle type in space and can pose serious health risks to astronauts during long-duration missions. The health effects of proton exposure are also a concern for cancer patients undergoing radiation treatment with accelerated protons. To investigate the damage induced by energetic protons in vivo to radiosensitive organs, 6-week-old BALB/c male mice were subjected to 250 MeV proton radiation at whole-body doses of 0.1, 1, and 2 Gy. The gastrointestinal (GI) tract of each exposed animal was dissected 4 h post-irradiation, and the isolated small intestinal tissue was analyzed for histopathological and gene expression changes. Histopathologic observation of the tissue using standard hematoxylin and eosin (H&E) staining methods to screen for morphologic changes showed a marked increase in apoptotic lesions for even the lowest dose of 0.1 Gy, similar to X- or γ rays. The percentage of apoptotic cells increased dose-dependently, but the dose response appeared supralinear, indicating hypersensitivity at low doses. A significant decrease in surviving crypts and mucosal surface area, as well as in cell proliferation, was also observed in irradiated mice. Gene expression analysis of 84 genes involved in the apoptotic process showed that most of the genes affected by protons were common between the low (0.1 Gy) and high (1 and 2 Gy) doses. However, the genes that were distinctively responsive to the low or high doses suggest that high doses of protons may cause apoptosis in the small intestine by direct damage to the DNA, whereas low doses of protons may trigger apoptosis through a different stress response mechanism.
Assuntos
Apoptose/efeitos da radiação , Dano ao DNA , Mucosa Intestinal/metabolismo , Prótons/efeitos adversos , Irradiação Corporal Total/efeitos adversos , Animais , Relação Dose-Resposta à Radiação , Intestinos/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Lesões Experimentais por RadiaçãoRESUMO
High-energy particle radiation could have a considerable impact on health during space missions. This study evaluated C57BL/6 mice on Day 40 after total-body 56Fe26+ irradiation at 0, 1, 2 and 3 gray (Gy). Radiation consistently increased thymus mass (one-way ANOVA: P < 0.005); spleen, liver and lung masses were similar among all groups. In the blood, there was no radiation effect on the white blood cell (WBC) count or major leukocyte types. However, the red blood cell count, hemoglobin, hematocrit and the CD8+ T cytotoxic (Tc) cell count and percentage all decreased, while both the CD4:CD8 (Th:Tc) cell ratio and spontaneous blastogenesis increased, in one or more irradiated groups compared with unirradiated controls (P < 0.05 vs 0 Gy). In contrast, splenic WBC, lymphocyte, B cell and T helper (Th) counts, %B cells and the CD4:CD8 ratio were all significantly elevated, while Tc percentages decreased, in one or more of the irradiated groups compared with controls (P < 0.05 vs 0 Gy). Although there were trends for minor, radiation-induced increases in %CD11b+ granulocytes in the spleen, cells double-labeled with adhesion markers (CD11b+CD54+, CD11b+CD62E+) were normal. Splenocyte spontaneous blastogenesis and that induced by mitogens (PHA, ConA, LPS) was equivalent to normal. In bone marrow, the percentage of cells expressing stem cell markers, Sca-1 and CD34/Sca-1, were low in one or more of the irradiated groups (P < 0.05 vs 0 Gy). Collectively, the data indicate that significant immunological abnormalities still exist more than a month after 56Fe irradiation and that there are differences dependent upon body compartment.
Assuntos
Ferro/farmacologia , Leucócitos/metabolismo , Irradiação Corporal Total , Animais , Biomarcadores/metabolismo , Contagem de Células Sanguíneas , Peso Corporal/efeitos dos fármacos , Peso Corporal/efeitos da radiação , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/efeitos da radiação , Adesão Celular/efeitos dos fármacos , Adesão Celular/efeitos da radiação , Feminino , Íons , Leucócitos/efeitos dos fármacos , Leucócitos/efeitos da radiação , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/efeitos da radiação , Subpopulações de Linfócitos/efeitos dos fármacos , Subpopulações de Linfócitos/efeitos da radiação , Camundongos Endogâmicos C57BL , Mitógenos/farmacologia , Tamanho do Órgão/efeitos dos fármacos , Tamanho do Órgão/efeitos da radiação , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/efeitos da radiaçãoRESUMO
Microgravity and radiation are stressors unique to the spaceflight environment that can have an impact on the central nervous system (CNS). These stressors could potentially lead to significant health risks to astronauts, both acutely during the course of a mission or chronically, leading to long-term, post-mission decrements in quality of life. The CNS is sensitive to oxidative injury due to high concentrations of oxidizable, unsaturated lipids and low levels of antioxidant defenses. The purpose of this study was to evaluate oxidative damage in the brain cortex and hippocampus in a ground-based model for spaceflight, which includes prolonged unloading and low-dose radiation. Whole-body low-dose/low-dose-rate (LDR) gamma radiation using (57)Co plates (0.04 Gy at 0.01 cGy/h) was delivered to 6 months old, mature, female C57BL/6 mice (n = 4-6/group) to simulate the radiation component. Anti-orthostatic tail suspension was used to model the unloading, fluid shift and physiological stress aspects of the microgravity component. Mice were hindlimb suspended and/or irradiated for 21 days. Brains were isolated 7 days or 9 months after irradiation and hindlimb unloading (HLU) for characterization of oxidative stress markers and microvessel changes. The level of 4-hydroxynonenal (4-HNE) protein, an oxidative specific marker for lipid peroxidation, was significantly elevated in the cortex and hippocampus after LDR + HLU compared to controls (P < 0.05). The combination group also had the highest level of nicotinamide adenine dinucleotide phosphate oxidase 2 (NOX2) expression compared to controls (P < 0.05). There was a significant decrease in superoxide dismutase (SOD) expression in the animals that received HLU only or combined LDR + HLU compared to control (P < 0.05). In addition, 9 months after LDR and HLU exposure, microvessel densities were the lowest in the combination group, compared to age-matched controls in the cortex (P < 0.05). Our data provide the first evidence that prolonged exposure to simulated microgravity and LDR radiation is associated with increased oxidative stress biomarkers that may increase the likelihood of brain injury and reduced antioxidant defense. NOX2-containing nicotinamide adenosine dinucleotide phosphate (NADPH oxidase) may contribute to spaceflight environment-induced oxidative stress.
Assuntos
Encéfalo/metabolismo , Encéfalo/efeitos da radiação , Estresse Oxidativo/efeitos da radiação , Ausência de Peso/efeitos adversos , Aldeídos/metabolismo , Animais , Antioxidantes/metabolismo , Apoptose/efeitos da radiação , Encéfalo/irrigação sanguínea , Encéfalo/citologia , Relação Dose-Resposta à Radiação , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Microvasos/metabolismo , Microvasos/efeitos da radiação , NADPH Oxidases/metabolismoRESUMO
AIM: To determine whether proton radiation can be used to treat chronic intractable pain. The focus of this study was on the biological effects of spinal cord irradiation. MATERIALS AND METHODS: Proton radiation (0-25 Gy, single fraction) was applied to the spinal cord within L3-L5 of Yucatan mini-pigs (n=20). Skin reaction, body mass and behavior were monitored. At euthanasia, blood and spinal cord were analyzed. RESULTS: Skin morbidity was mild and overall health for the 5-20 Gy-treated groups was good based on behavior and weight gain up to 8.5-9 months post-exposure. The 25 Gy-treated animals developed hind limb weakness at 2.5-3 months and were euthanized. Radiation had a significant effect on white blood cell count (p<0.05), with the 25 Gy-treated mini-pigs having the highest number of all three major leukocyte populations. A few differences were also noted for erythrocyte parameters, but the blood chemistry panel was normal. Apoptosis in the targeted portion of the spinal cord was elevated in the 20- and 25 Gy-treated groups versus 0 Gy (p<0.05) based on the terminal deoxynucleotidyl transferase dUTP nick-end labeling assay. There was a trend (p<0.1) for a radiation effect on glial fibrillary acidic protein expression, with the highest value being found after 25 Gy. Histology showed no difference between 0 versus 25 Gy. CONCLUSION: The data demonstrated that a small segment of the spinal cord can be readily targeted using proton radiation; doses ranging from 5-20 Gy were well-tolerated in an animal model with radiosensitivity similar to humans. Future studies with a pain model should use ≤15 Gy.
Assuntos
Dor Crônica/radioterapia , Dor Intratável/radioterapia , Traumatismos da Medula Espinal/radioterapia , Medula Espinal/patologia , Animais , Apoptose/efeitos da radiação , Dor Crônica/patologia , Relação Dose-Resposta à Radiação , Humanos , Contagem de Leucócitos , Dor Intratável/patologia , Projetos Piloto , Terapia com Prótons , Traumatismos da Medula Espinal/patologia , Suínos , Porco MiniaturaRESUMO
The space environment exposes astronauts to risks of acute and chronic exposure to ionizing radiation. Of particular concern is possible exposure to ionizing radiation from a solar particle event (SPE). During an SPE, magnetic disturbances in specific regions of the Sun result in the release of intense bursts of ionizing radiation, primarily consisting of protons that have a highly variable energy spectrum. Thus, SPE events can lead to significant total body radiation exposures to astronauts in space vehicles and especially while performing extravehicular activities. Simulated energy profiles suggest that SPE radiation exposures are likely to be highest in the skin. In the current report, we have used our established miniature pig model system to evaluate the skin toxicity of simulated SPE radiation exposures that closely resemble the energy and fluence profile of the September, 1989 SPE using either conventional radiation (electrons) or proton simulated SPE radiation. Exposure of animals to electron or proton radiation led to dose-dependent increases in epidermal pigmentation, the presence of necrotic keratinocytes at the dermal-epidermal boundary and pigment incontinence, manifested by the presence of melanophages in the derm is upon histological examination. We also observed epidermal hyperplasia and a reduction in vascular density at 30 days following exposure to electron or proton simulated SPE radiation. These results suggest that the doses of electron or proton simulated SPE radiation results in significant skin toxicity that is quantitatively and qualitatively similar. Radiation-induced skin damage is often one of the first clinical signs of both acute and non-acute radiation injury where infection may occur, if not treated. In this report, histopathology analyses of acute radiation-induced skin injury are discussed.
Assuntos
Meio Ambiente Extraterreno , Prótons/efeitos adversos , Exposição à Radiação/efeitos adversos , Radiação Ionizante , Pele/lesões , Pele/efeitos da radiação , Animais , Astronautas , Relação Dose-Resposta à Radiação , Exposição Ambiental/efeitos adversos , Modelos Animais , Doses de Radiação , Pigmentação da Pele/efeitos da radiação , Atividade Solar , Sistema Solar , Suínos , Porco Miniatura , Irradiação Corporal Total/efeitos adversosRESUMO
BACKGROUND/AIM: Fisher rat thyroid cells (FRTL-5) display increased proliferation, reduced follicularization and decreased thyroxin release with repeated sub-culturing. These changes occur earlier and more rapidly following exposure to ionizing radiation. We hypothesized that altered transforming growth factor-ß1 (TGF-ß1) signaling contributes to these differences. MATERIALS AND METHODS: Assessments included FRTL-5 cell growth rate and quantification of TGF-ß1 ligand and receptors. The levels and activity of Smads2, 3 and 4 were measured by western blotting and the ability of TGF-ß1 to regulate cyclin A and plasminogen activator inhibitor type 1 (PAI-1) activity was assessed using transfection assays. RESULTS: TGF-ß1 production increased after radiation but returned to control levels after repeated sub-culturing. There was no difference in TGF-ß1 levels between un-irradiated cells at low versus high-passage number. TGF-ß1 receptors and basal levels of Smads2, 3 and 4 remained unchanged. However, there were significant changes in cell proliferation, TGF-ß1-mediated Smads2 and 3 activation and in TGF-ß1's ability to regulate cyclin A and PAI-1 transcription in irradiated and repeatedly sub-cultured cells (p<0.05). CONCLUSION: Collectively, these results support the conclusion that alterations in the TGF-ß1 pathway contribute to phenotypic changes in FRTL-5 cells as a function of passage number and radiation.
Assuntos
Transdução de Sinais/efeitos da radiação , Fator de Crescimento Transformador beta1/metabolismo , Animais , Técnicas de Cultura de Células , Linhagem Celular , Proliferação de Células/efeitos da radiação , Células Cultivadas , Relação Dose-Resposta à Radiação , Raios gama , Expressão Gênica , Fosforilação , Doses de Radiação , Ratos , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteínas Smad/metabolismo , Glândula Tireoide/citologia , Glândula Tireoide/metabolismo , Glândula Tireoide/efeitos da radiaçãoRESUMO
The goal was to characterize differences in cell response after exposure to active beam scanning (ABS) protons compared to a passive delivery system. Human lung epithelial (HLE) cells were evaluated at various locations along the proton depth dose profile. The dose delivered at the Bragg peak position was essentially identical (â¼4 Gy) with the two techniques, but depth dose data showed that ABS resulted in lower doses at entry and more rapid drop-off after the peak. Average dose rates for the passive and ABS beams were 1.1 Gy/min and 5.1 Gy/min, respectively; instantaneous dose rates were 19.2 Gy/min and 2,300 Gy/min (to a 0.5 ×â 0.5 mm(2) voxel). Analysis of DNA synthesis was based on (3)H-TdR incorporation. Quantitative real-time polymerase chain reaction (RT-PCR) was done to determine expression of genes related to p53 signaling and DNA damage; a total of 152 genes were assessed. Spectral karyotyping and analyses of the Golgi apparatus and cytokines produced by the HLE cells were also performed. At or near the Bragg peak position, ABS protons resulted in a greater decrease in DNA synthesis compared to passively delivered protons. Genes with >2-fold change (P <â 0.05 vs. 0 Gy) after passive proton irradiation at one or more locations within the Bragg curve were BTG2, CDKN1A, IFNB1 and SIAH1. In contrast, many more genes had >2-fold difference with ABS protons: BRCA1, BRCA2, CDC25A, CDC25C, CCNB2, CDK1, DMC1, DNMT1, E2F1, EXO1, FEN1, GADD45A, GTSE1, IL-6, JUN, KRAS, MDM4, PRC1, PTTG1, RAD51, RPA1, TNF, WT1, XRCC2, XRCC3 and XRCC6BP1. Spectral karyotyping revealed numerous differences in chromosomal abnormalities between the two delivery systems, especially at or near the Bragg peak. Percentage of cells staining for the Golgi apparatus was low after exposure to passive and active proton beams. Studies such as this are needed to ensure patient safety and make modifications in ABS delivery, if necessary.
Assuntos
Células Epiteliais Alveolares/efeitos da radiação , Prótons/efeitos adversos , Células Epiteliais Alveolares/metabolismo , Aberrações Cromossômicas/efeitos da radiação , Citocinas/genética , Citocinas/metabolismo , Dano ao DNA/genética , Replicação do DNA/efeitos da radiação , Relação Dose-Resposta à Radiação , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos da radiação , Complexo de Golgi/metabolismo , Complexo de Golgi/efeitos da radiação , Humanos , Cariótipo , Pessoa de Meia-Idade , Terapia com Prótons , Doses de Radiação , Radiação Ionizante , Eficiência Biológica Relativa , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismoRESUMO
PURPOSE: Concern regarding radiation effects on human health continues to increase worldwide. Given that infection is a major cause of morbidity and mortality after exposure, the aim of this study was to evaluate decrements in immune cell populations using a mammalian model subjected to a live bacterial infection. MATERIALS AND METHODS: C57BL/6 mice were exposed to total-body irradiation (TBI) with 3 Gy protons (70 cGy/min). One, 2, 4, 8 or 16 days later, subsets of mice were injected intraperitoneally with live Escherichia coli [055:K59(B5)]. Control groups received no radiation and vehicle (no bacteria). The mice were euthanized for analyses 90-120 min after injection of the bacteria. RESULTS: There were no unexpected effects of radiation or E. coli alone. Despite dramatic radiation-induced decreases in all leukocyte populations in both the blood and spleen, irradiated mice were still able to respond to an immune challenge based on capacity to generate an oxidative burst and secrete inflammatory cytokines, i.e., tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6). However, these responses were generally elevated above control values. CONCLUSIONS: Together, these results suggest the possibility for enhanced inflammation-associated tissue injury and increased risk for chronic inflammation.
Assuntos
Escherichia coli/fisiologia , Viabilidade Microbiana , Irradiação Corporal Total/efeitos adversos , Animais , Tamanho Corporal/imunologia , Tamanho Corporal/efeitos da radiação , Citocinas/metabolismo , Relação Dose-Resposta à Radiação , Contagem de Eritrócitos , Feminino , Regulação da Expressão Gênica/imunologia , Regulação da Expressão Gênica/efeitos da radiação , Antígenos de Histocompatibilidade Classe II/metabolismo , Leucócitos/citologia , Leucócitos/imunologia , Leucócitos/efeitos da radiação , Camundongos , Camundongos Endogâmicos C57BL , Tamanho do Órgão/imunologia , Tamanho do Órgão/efeitos da radiação , Contagem de Plaquetas , Explosão Respiratória/imunologia , Explosão Respiratória/efeitos da radiação , Baço/citologia , Baço/imunologia , Baço/efeitos da radiaçãoRESUMO
An effective countermeasure against radiation damage to normal tissues is urgently needed. The major goal of the present study was to determine if minocycline could modify the immunomodulatory effects of radiation on the brain. C57BL/6 mice were treated with minocycline intraperitoneally for 5 days beginning immediately before total-body exposure to 0, 1, 2 and 3 Gray (Gy) (60)Co γ-rays. Brains were collected on days 4 and 32 post-irradiation for cytokine and gene analyses. Minocycline treatment significantly increased the levels of interleukin (IL)-10, IL-15 and vascular endothelial growth factor (VEGF) in the brain on day 4 in one or more irradiated groups compared to radiation-alone (p<0.05). IL-10 is anti-inflammatory, IL-15 can prevent apoptosis and VEGF is nuroprotective. On day 32, the drug decreased IL-1ß in the 2- Gy group (p<0.05 vs. 2-Gy alone); this cytokine is implicated in immune-related central nervous system pathologies. Microarray analysis of brains on day 32 showed that while radiation increased expression of inflammatory genes such as Il1f10, Il17, Tnfrsf11b, Tnfsf12, Il12b and Il1f8, these were no longer up-regulated in the minocycline-treated groups. Similarly, the pro-apoptotic gene Bik and nitric oxide synthase producer (Nostrin) were no longer up-regulated in the drug-treated groups. Pathway analysis based on gene data suggested that catenin-ß1 and tumor suppressor-related transcription regulation were significantly activated by radiation and/or minocycline (activation z-score >2.0). Overall, the data warrant further testing of minocycline as a potential neuroprotectant against radiation-induced damage.
Assuntos
Encéfalo/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Minociclina/administração & dosagem , Irradiação Corporal Total , Animais , Encéfalo/efeitos da radiação , Raios gama , Regulação da Expressão Gênica/efeitos da radiação , Interleucina-10/biossíntese , Interleucina-15/biossíntese , Camundongos , Fármacos Neuroprotetores/administração & dosagem , Doses de Radiação , Protetores contra Radiação/administração & dosagem , Transcriptoma/efeitos dos fármacos , Transcriptoma/efeitos da radiação , Fator A de Crescimento do Endotélio Vascular/biossínteseRESUMO
The goal of the present study was to obtain pilot data on the effects of protracted low-dose/low-dose-rate (LDR) γ-rays on the skin, both with and without acute gamma or proton irradiation (IR). Six groups of C57BL/6 mice were examined: a) 0 Gy control, b) LDR, c) Gamma, d) LDR+Gamma, e) Proton, and f) LDR+Proton. LDR radiation was delivered to a total dose of 0.01 Gy (0.03 cGy/h), whereas the Gamma and Proton groups received 2 Gy (0.9 Gy/min and 1.0 Gy/min, respectively). Assays were performed 56 days after exposure. Skin samples from all irradiated groups had activated caspase-3, indicative of apoptosis. The significant (p<0.05) increases in immunoreactivity in the Gamma and Proton groups were not present when LDR pre-exposure was included. However, the terminal deoxynucleotidyl transferase dUTP nick-end labeling assay for DNA fragmentation and histological examination of hematoxylin and eosin-stained sections revealed no significant differences among groups, regardless of radiation regimen. The data demonstrate that caspase-3 activation initially triggered by both forms of acute radiation was greatly elevated in the skin nearly two months after whole-body exposure. In addition, LDR γ-ray priming ameliorated this response.
Assuntos
Raios gama , Prótons , Lesões Experimentais por Radiação/enzimologia , Pele/efeitos da radiação , Animais , Apoptose , Caspase 3/metabolismo , Fragmentação do DNA , Relação Dose-Resposta à Radiação , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Tolerância a Radiação/efeitos da radiação , Pele/enzimologia , Pele/patologia , Irradiação Corporal TotalRESUMO
Our previous results with flight (FLT) mice showed abnormalities in thymuses and spleens that have potential to compromise immune defense mechanisms. In this study, the organs were further evaluated in C57BL/6 mice after Space Shuttle Atlantis returned from a 13-day mission. Thymuses and spleens were harvested from FLT mice and ground controls housed in similar animal enclosure modules (AEM). Organ and body mass, DNA fragmentation and expression of genes related to T cells and cancer were determined. Although significance was not obtained for thymus mass, DNA fragmentation was greater in the FLT group (P<0.01). Spleen mass alone and relative to body mass was significantly decreased in FLT mice (P<0.05). In FLT thymuses, 6/84 T cell-related genes were affected versus the AEM control group (P<0.05; up: IL10, Il18bp, Il18r1, Spp1; down: Ccl7, IL6); 15/84 cancer-related genes had altered expression (P<0.05; up: Casp8, FGFR2, Figf, Hgf, IGF1, Itga4, Ncam1, Pdgfa, Pik3r1, Serpinb2, Sykb; down: Cdc25a, E2F1, Mmp9, Myc). In the spleen, 8/84 cancer-related genes were affected in FLT mice compared to AEM controls (P<0.05; up: Cdkn2a; down: Birc5, Casp8, Ctnnb1, Map2k1, Mdm2, NFkB1, Pdgfa). Pathway analysis (apoptosis signaling and checkpoint regulation) was used to map relationships among the cancer-related genes. The results showed that a relatively short mission in space had a significant impact on both organs. The findings also indicate that immune system aberrations due to stressors associated with space travel should be included when estimating risk for pathologies such as cancer and infection and in designing appropriate countermeasures. Although this was the historic last flight of NASA's Space Shuttle Program, exploration of space will undoubtedly continue.
Assuntos
Voo Espacial , Baço/fisiologia , Timo/fisiologia , Animais , Fragmentação do DNA , Comportamento Alimentar , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Camundongos , Modelos Biológicos , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Tamanho do Órgão , Transdução de Sinais , Baço/anatomia & histologia , Timo/anatomia & histologia , Ausência de PesoRESUMO
The ability to efficiently generate integration-free induced pluripotent stem cells (iPSCs) from the most readily available source-peripheral blood-has the potential to expedite the advances of iPSC-based therapies. We have successfully generated integration-free iPSCs from cord blood (CB) CD34(+) cells with improved oriP/EBNA1-based episomal vectors (EV) using a strong spleen focus forming virus (SFFV) long terminal repeat (LTR) promoter. Here we show that Yamanaka factors (OCT4, SOX2, MYC, and KLF4)-expressing EV can also reprogram adult peripheral blood mononuclear cells (PBMNCs) into pluripotency, yet at a very low efficiency. We found that inclusion of BCL-XL increases the reprogramming efficiency by approximately 10-fold. Furthermore, culture of CD3(-)/CD19(-) cells or T/B cell-depleted MNCs for 4-6 days led to the generation of 20-30 iPSC colonies from 1 ml PB, an efficiency that is substantially higher than previously reported. PB iPSCs express pluripotency markers, form teratomas, and can be induced to differentiate in vitro into mesenchymal stem cells, cardiomyocytes, and hepatocytes. Used together, our optimized factor combination and reprogramming strategy lead to efficient generation of integration-free iPSCs from adult PB. This discovery has potential applications in iPSC banking, disease modeling and regenerative medicine.
Assuntos
Células Sanguíneas/citologia , Técnicas de Cultura de Células/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Fatores de Transcrição/metabolismo , Proteína bcl-X/metabolismo , Adulto , Animais , Antígenos CD34/metabolismo , Células Sanguíneas/metabolismo , Diferenciação Celular , Linhagem da Célula , Reprogramação Celular , Sangue Fetal/citologia , Vetores Genéticos/genética , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Fator 4 Semelhante a Kruppel , Lentivirus/genética , Camundongos , Plasmídeos/metabolismoRESUMO
OBJECTIVES: Novel targeted therapeutic strategies to overcome radio-resistance of cancer cells traditionally treated with radiation may improve patient survival with the added benefit of reduced systemic toxicity. Herein, we tested the feasibility of Linifanib (ABT-869), a multi-receptor tyrosine kinase inhibitor of members of vascular endothelial growth factor (VEGF) and platelet derived growth factor (PDGF) receptor families, on radio-sensitization of Head and Neck Squamous Cell Carcinoma (HNSCC). MATERIALS AND METHODS: UMSCC-22A and UMSCC-22B cells were treated with Linifanib and γ-radiation response was determined. Cell viability, cytotoxicity, apoptosis induction and cell cycle distribution were examined by MTT assay, colony formation assay and flow cytometry. In addition, expression of STAT3 and downstream signaling proteins were assessed using western immunoblotting. RESULTS: Treatment with Linifanib resulted in cell growth inhibition, G2/M cell cycle arrest, induction of cell death via apoptosis, reduced phosphorylation of STAT3, which has been linked to radio-resistance, lower expression of cyclin D1, survivin and increased PARP cleavage. In addition, Linifanib overcame the radio-resistance of the cell lines and significantly enhanced radiation-induced cytotoxicity (p<0.05). CONCLUSION: These data suggest the possibility of combining targeted therapeutic such as Linifanib with radiation to enhance inhibition of cell growth and apoptosis in HNSCC cells. Thus, it may provide a novel therapeutic strategy and improve efficacy of radiation against HNSCC in the future.
Assuntos
Carcinoma de Células Escamosas/radioterapia , Neoplasias de Cabeça e Pescoço/radioterapia , Indazóis/farmacologia , Compostos de Fenilureia/farmacologia , Tolerância a Radiação/efeitos dos fármacos , Radiossensibilizantes/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma de Células Escamosas/patologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Fator de Transcrição STAT3/metabolismo , Transdução de SinaisRESUMO
The direct conversion of skin cells into somatic stem cells has opened new therapeutic possibilities in regenerative medicine. Here, we show that human induced mesenchymal stem cells (iMSCs) can be efficiently generated from cord blood (CB)- or adult peripheral blood (PB)-CD34(+) cells by direct reprogramming with a single factor, OCT4. In the presence of a GSK3 inhibitor, 16% of the OCT4-transduced CD34(+) cells are converted into iMSCs within 2 weeks. Efficient direct reprogramming is achieved with both episomal vector-mediated transient OCT4 expression and lentiviral vector-mediated OCT4 transduction. The iMSCs express MSC markers, resemble bone marrow (BM)-MSCs in morphology, and possess in vitro multilineage differentiation capacity, yet have a greater proliferative capacity compared with BM-MSCs. Similar to BM-MSCs, the implanted iMSCs form bone and connective tissues, and are non-tumorigenic in mice. However, BM-MSCs do not, whereas iMSCs do form muscle fibers, indicating a potential functional advantage of iMSCs. In addition, we observed that a high level of OCT4 expression is required for the initial reprogramming and the optimal iMSC self-renewal, while a reduction of OCT4 expression is required for multilineage differentiation. Our method will contribute to the generation of patient-specific iMSCs, which could have applications in regenerative medicine. This discovery may also facilitate the development of strategies for direct conversion of blood cells into other types of cells of clinical importance.
Assuntos
Antígenos CD34/metabolismo , Células Sanguíneas/citologia , Sangue Fetal/citologia , Células-Tronco Mesenquimais/citologia , Fator 3 de Transcrição de Octâmero/metabolismo , Animais , Células Sanguíneas/metabolismo , Osso e Ossos/patologia , Diferenciação Celular , Células Cultivadas , Reprogramação Celular , Tecido Conjuntivo/patologia , Sangue Fetal/metabolismo , Vetores Genéticos/metabolismo , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/metabolismo , Células-Tronco Hematopoéticas/citologia , Humanos , Cariotipagem , Lentivirus/genética , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Camundongos , Fator 3 de Transcrição de Octâmero/genética , Transdução GenéticaRESUMO
BACKGROUND: Development of mammary tumors is an age-associated phenomenon that is likely due to deficits in the neuroendocrine-immune interactions. Previously, we demonstrated that L-deprenyl, a monoamine oxidase-B (MAO-B) inhibitor, can enhance immune responses and restore noradrenergic (NA) innervation in the spleens of rats with carcinogen-induced and spontaneously developing mammary tumors. OBJECTIVES: To investigate whether (1) treatment of early middle-aged female rats would prevent the spontaneous development of mammary tumors accompanied by restoration of immunity in the spleen and draining lymph nodes (DLN) and sympathetic NA innervation in the spleen and (2) deprenyl can influence the proliferation of estrogen receptor (ER)-positive (MCF-7 and T47D) and ER-negative (MDA-MB-231 and Hs 578T) human breast cancer cells. METHODS: Early middle-aged (8- to 9-month-old) female Sprague-Dawley rats were treated with 0, 1.0 or 2.5 mg of deprenyl/kg body weight (BW) daily i.p. for 12 months. Cells of ER-positive (ER+) and ER-negative (ER-) human breast cancer cell lines were incubated with media or 10(-3) to 10(-8) M deprenyl for 1, 2, 4 or 6 days to examine the proliferation of cells. RESULTS: Tumor incidence increased in saline-treated old female rats, while deprenyl treatment significantly reduced the incidence of mammary tumors in these rats. Saline-treated tumor-bearing rats exhibited reduced splenic NA innervation and norepinephrine (NE) content, splenic interleukin (IL)-2 and interferon (IFN)-γ levels and NK cell activity as well as DLN IL-2 and IFN-γ levels compared to young female rats without tumors. In contrast, treatment with 2.5 mg/kg of deprenyl enhanced IL-2 and IFN-γ production in both the spleen and DLN as well as splenic natural killer (NK) cell activity. Deprenyl treatment also increased concanavalin A (Con A)-induced proliferation of T lymphocytes in the DLN. Deprenyl-induced changes in immune responses were accompanied by enhanced NA innervation and NE content in the spleen. In vitro incubation of various concentrations of deprenyl with ER+ human breast cancer cell lines partly inhibited the proliferation of cells, while it had no effect on the ER- breast cancer cells. CONCLUSIONS: These results suggest that (1) development of mammary tumors is mediated through the loss of immunity and sympathetic NA nerve fibers accompanied by reduced NE levels in the spleen, (2) the prevention of mammary tumor development by deprenyl may involve the reversal of the tumor-associated decline in sympathetic NA activity and cell-mediated immune responses in the spleen and DLN and (3) the antitumor effects of deprenyl may be partially mediated through ER-dependent intracellular signaling pathways.
Assuntos
Neoplasias da Mama/patologia , Neoplasias da Mama/prevenção & controle , Linfonodos , Neuroimunomodulação/efeitos dos fármacos , Fármacos Neuroprotetores/administração & dosagem , Selegilina/administração & dosagem , Baço , Fatores Etários , Análise de Variância , Animais , Proliferação de Células/efeitos dos fármacos , Concanavalina A/farmacologia , Citocinas/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Citometria de Fluxo , Humanos , Células Matadoras Naturais/efeitos dos fármacos , Linfonodos/efeitos dos fármacos , Linfonodos/imunologia , Linfonodos/metabolismo , Transplante de Neoplasias , Ratos , Ratos Sprague-Dawley , Baço/efeitos dos fármacos , Baço/imunologia , Baço/metabolismoRESUMO
Exposure to total-body radiation induces hematological changes, which can detriment one's immune response to wounds and infection. Here, the decreases in blood cell counts after acute radiation doses of γ-ray or proton radiation exposure, at the doses and dose-rates expected during a solar particle event (SPE), are reported in the ferret model system. Following the exposure to γ-ray or proton radiation, the ferret peripheral total white blood cell (WBC) and lymphocyte counts decreased whereas neutrophil count increased within 3 hours. At 48 hours after irradiation, the WBC, neutrophil, and lymphocyte counts decreased in a dose-dependent manner but were not significantly affected by the radiation type (γ-rays verses protons) or dose rate (0.5 Gy/minute verses 0.5 Gy/hour). The loss of these blood cells could accompany and contribute to the physiological symptoms of the acute radiation syndrome (ARS).
RESUMO
PURPOSE: The major goal was to evaluate effects of various radiation regimens on leukocyte populations relatively long-term after whole-body irradiation. MATERIALS AND METHODS: C57BL/6 mice were exposed to-low-dose/low-dose rate (LDR) (57)Co γ-rays (0.01 Gy, 0.03 cGy/h), with and without acute 2 Gy proton (1 Gy/min) or γ-ray (0.9 Gy/min) irradiation; analyses were done on days 21 and 56 post-exposure. RESULTS: Numerous radiation-induced changes were noted at one or both time points. Among the most striking differences (P < 0.05) were: (i) High percentage of CD4(+)CD25(+)Foxp3(+) T cells in spleens from the Proton vs. LDR, Gamma and LDR + Proton groups (day 56); (ii) high interleukin-2 (IL-2) in spleen supernatants from the LDR and LDR + Proton groups vs. 0 Gy (day 56), whereas IL-10 was high in the LDR + Gamma group vs. 0 Gy (day 56); (iii) difference in transforming growth factor-ß1 (TGF-ß1) in spleen supernatants from Proton and LDR + Proton groups vs. Gamma and LDR + Gamma groups (both days); (iv) low TGF-ß1 in blood from LDR + Proton vs. LDR + Gamma group (day 21); and (v) high level of activated cJun N-terminal kinase (JNK) in CD4(+) T cells from LDR + Proton vs. LDR + Gamma group (day 21). CONCLUSIONS: The findings demonstrate that at least some immune responses to acute 2 Gy radiation were dependent on radiation quality time of assessment, and pre-exposure to LDR γ-rays.
Assuntos
Citocinas/metabolismo , Meio Ambiente Extraterreno , Fatores de Transcrição Forkhead/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/efeitos da radiação , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Peso Corporal/efeitos da radiação , Citocinas/biossíntese , Feminino , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Leucócitos/citologia , Leucócitos/efeitos da radiação , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/efeitos da radiação , Baço/citologia , Linfócitos T Reguladores/citologia , Fator de Crescimento Transformador beta1/sangue , Fator A de Crescimento do Endotélio Vascular/sangueRESUMO
BACKGROUND/AIM: We previously found that minocycline enhanced the levels of several leukocyte populations and had the capacity to induce secretion of certain cytokines early after irradiation. In the current study we further determined the drug's effect on hematopoietic recovery. MATERIALS AND METHODS: Minocycline was injected intraperitoneally into C57BL/6 mice for 5 days, beginning immediately before exposure to (60)Co γ-rays (1, 2, 3 Gy). Thirty-two days post-irradiation, spleen and blood were collected to quantify cell populations, cytokines in splenic T-cell supernatants after anti-CD3 activation, and chromosomic status based on spectral karyotyping. RESULTS: While radiation resulted in significantly lower B-cell counts at 3 Gy in both blood and spleen, minocycline treatment increased the counts and/or percentages of splenic B-cells at 2 Gy and 3 Gy. In spleen supernatants, the drug-alone increased the levels of cytokines, including interleukin-1α (IL-1α) and IL-6 that are radioprotective, as well as granulocyte-macrophage colony-stimulating factor (GM-CSF) and G-CSF that accelerate neutrophil recovery. In addition, minocycline suppressed the production of interferon-γ that can prevent hematopoiesis. Dose-dependent radiation-induced chromosomic abnormalities were present in splenic leukocytes. CONCLUSION: The data indicate that minocycline exerts a relatively long-term effect on parameters that influence hematopoietic recovery. Further testing of this drug as a countermeasure for acute radiation syndrome, is necessary to determine its full potential.
Assuntos
Hematopoese/efeitos dos fármacos , Hematopoese/efeitos da radiação , Minociclina/farmacologia , Irradiação Corporal Total/métodos , Animais , Antibacterianos/administração & dosagem , Antibacterianos/farmacologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Linfócitos B/efeitos da radiação , Contagem de Células Sanguíneas , Aberrações Cromossômicas/efeitos dos fármacos , Aberrações Cromossômicas/efeitos da radiação , Citocinas/metabolismo , Relação Dose-Resposta à Radiação , Feminino , Citometria de Fluxo , Fator Estimulador de Colônias de Granulócitos/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Injeções Intraperitoneais , Interleucina-1alfa/metabolismo , Interleucina-6/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Minociclina/administração & dosagem , Cariotipagem Espectral , Baço/efeitos dos fármacos , Baço/metabolismo , Baço/efeitos da radiação , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Linfócitos T/efeitos da radiaçãoRESUMO
BACKGROUND/AIM: To evaluate the impact of an antibiotic, minocycline, on several immune parameters in response to radiation in a mouse model. MATERIALS AND METHODS: C57BL/6 mice were treated with minocycline (i.p.) for 5 days, beginning immediately before radiation with 1-3 Gy (60)Co γ-rays. Spleen and blood were collected on day 4 post-irradiation. Cell populations were determined in the blood and spleen. Splenocytes were activated with anti-CD3 antibody for 48 h and cytokines were quantified. RESULTS: Minocycline increased the counts and/or percentages of splenic macrophages, granulocytes, natural killer, T- and CD8(+) T-cells (p<0.05 versus radiation alone). Minocycline significantly increased the expression of interleukin-1α and ß, which are radioprotective, as well as the ones of granulocyte-macrophage colony-stimulating factor and granulocyte colony-stimulating factor, which accelerate neutrophil recovery (p<0.05 versus radiation alone), while suppressing cytokines that could prevent hematopoiesis, e.g. macrophage inflammatory protein-1α, tumor necrosis factor-α and interferon-γ. CONCLUSION: These data indicate that minocycline should be further tested for use in restoration of the hematopoietic system after radiation exposure.