Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Physiol ; 602(12): 2763-2806, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38761133

RESUMO

Hypoxia-inducible factor (HIF)-1α is continuously synthesized and degraded in normoxia. During hypoxia, HIF1α stabilization restricts cellular/mitochondrial oxygen utilization. Cellular stressors can stabilize HIF1α even during normoxia. However, less is known about HIF1α function(s) and sex-specific effects during normoxia in the basal state. Since skeletal muscle is the largest protein store in mammals and protein homeostasis has high energy demands, we determined HIF1α function at baseline during normoxia in skeletal muscle. Untargeted multiomics data analyses were followed by experimental validation in differentiated murine myotubes with loss/gain of function and skeletal muscle from mice without/with post-natal muscle-specific Hif1a deletion (Hif1amsd). Mitochondrial oxygen consumption studies using substrate, uncoupler, inhibitor, titration protocols; targeted metabolite quantification by gas chromatography-mass spectrometry; and post-mitotic senescence markers using biochemical assays were performed. Multiomics analyses showed enrichment in mitochondrial and cell cycle regulatory pathways in Hif1a deleted cells/tissue. Experimentally, mitochondrial oxidative functions and ATP content were higher with less mitochondrial free radical generation with Hif1a deletion. Deletion of Hif1a also resulted in higher concentrations of TCA cycle intermediates and HIF2α proteins in myotubes. Overall responses to Hif1amsd were similar in male and female mice, but changes in complex II function, maximum respiration, Sirt3 and HIF1ß protein expression and muscle fibre diameter were sex-dependent. Adaptive responses to hypoxia are mediated by stabilization of constantly synthesized HIF1α. Despite rapid degradation, the presence of HIF1α during normoxia contributes to lower mitochondrial oxidative efficiency and greater post-mitotic senescence in skeletal muscle. In vivo responses to HIF1α in skeletal muscle were differentially impacted by sex. KEY POINTS: Hypoxia-inducible factor -1α (HIF1α), a critical transcription factor, undergoes continuous synthesis and proteolysis, enabling rapid adaptive responses to hypoxia by reducing mitochondrial oxygen consumption. In mammals, skeletal muscle is the largest protein store which is determined by a balance between protein synthesis and breakdown and is sensitive to mitochondrial oxidative function. To investigate the functional consequences of transient HIF1α expression during normoxia in the basal state, myotubes and skeletal muscle from male and female mice with HIF1α knockout were studied using complementary multiomics, biochemical and metabolite assays. HIF1α knockout altered the electron transport chain, mitochondrial oxidative function, signalling molecules for protein homeostasis, and post-mitotic senescence markers, some of which were differentially impacted by sex. The cost of rapid adaptive responses mediated by HIF1α is lower mitochondrial oxidative efficiency and post-mitotic senescence during normoxia.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia , Mitocôndrias Musculares , Músculo Esquelético , Animais , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Feminino , Masculino , Músculo Esquelético/metabolismo , Camundongos , Mitocôndrias Musculares/metabolismo , Caracteres Sexuais , Homeostase , Fibras Musculares Esqueléticas/metabolismo , Camundongos Endogâmicos C57BL , Consumo de Oxigênio/fisiologia
2.
Cell ; 133(2): 223-34, 2008 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-18423195

RESUMO

Skin plays an essential role, mediated in part by its remarkable vascular plasticity, in adaptation to environmental stimuli. Certain vertebrates, such as amphibians, respond to hypoxia in part through the skin; but it is unknown whether this tissue can influence mammalian systemic adaptation to low oxygen levels. We have found that epidermal deletion of the hypoxia-responsive transcription factor HIF-1alpha inhibits renal erythropoietin (EPO) synthesis in response to hypoxia. Conversely, mice with an epidermal deletion of the von Hippel-Lindau (VHL) factor, a negative regulator of HIF, have increased EPO synthesis and polycythemia. We show that nitric oxide release induced by the HIF pathway acts on cutaneous vascular flow to increase systemic erythropoietin expression. These results demonstrate that in mice the skin is a critical mediator of systemic responses to environmental oxygen.


Assuntos
Epiderme/fisiologia , Oxigênio/metabolismo , Animais , Análise Química do Sangue , Eritropoetina/metabolismo , Humanos , Fator 1 Induzível por Hipóxia/genética , Fator 1 Induzível por Hipóxia/metabolismo , Queratinócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Óxido Nítrico/sangue , Oxigênio/sangue , Proteína Supressora de Tumor Von Hippel-Lindau/genética , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo
3.
J Neurooncol ; 127(2): 391-400, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26746692

RESUMO

The purpose of this pilot study was to determine whether blood-borne microvesicles from newly diagnosed glioblastoma patients could be used as biomarkers. We collected 2.8 mL blood from 16 post-operative patients at the time that they were being simulated for chemoradiation therapy (radiation with concurrent temozolomide). Two additional samples were collected during chemoradiation therapy and a final sample was collected at the end of chemoradiation therapy. Patients continued with the therapy suggested by their physicians, based on tumor conference consensus and were followed for recurrence and overall survival. Microvesicles were isolated using serial centrifugation and stained for surface markers (Annexin V for phosphotidyl serine, CD41 for platelets, anti-EGFR for tumor cells, and CD235 for red blood cells). Flow cytometry analysis was performed. Our findings provide initial evidence that increases in Annexin V positive microvesicle levels during chemoradiation therapy are associated with earlier recurrence and shorter overall survival in newly diagnosed glioblastoma patients. The effect is dramatic, with over a four-fold increase in the hazard ratio for an individual at the 75th versus the 25th percentile. Moreover the pattern of Annexin V positive microvesicles remain significant after adjustment for confounding clinical variables that have previously been shown to be prognostic for recurrence and survival. Inclusion of neutrophil levels at the start of chemoradiation therapy in the model yielded the largest attenuation of the observed association. Further studies will be needed to verify and further investigate the association between these two entities.


Assuntos
Biomarcadores Tumorais/sangue , Neoplasias Encefálicas/patologia , Micropartículas Derivadas de Células/patologia , Glioblastoma/patologia , Recidiva Local de Neoplasia/patologia , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/terapia , Quimiorradioterapia , Feminino , Seguimentos , Glioblastoma/diagnóstico , Glioblastoma/mortalidade , Glioblastoma/terapia , Humanos , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/diagnóstico , Recidiva Local de Neoplasia/mortalidade , Recidiva Local de Neoplasia/terapia , Estadiamento de Neoplasias , Prognóstico , Taxa de Sobrevida
4.
Int J Mol Sci ; 17(6)2016 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-27322243

RESUMO

Spaceflight occasionally requires multiple extravehicular activities (EVA) that potentially subject astronauts to repeated changes in ambient oxygen superimposed on those of space radiation exposure. We thus developed a novel in vitro model system to test lung cell damage following repeated exposure to radiation and hyperoxia. Non-tumorigenic murine alveolar type II epithelial cells (C10) were exposed to >95% O2 for 8 h only (O2), 0.25 Gy ionizing γ-radiation (IR) only, or a double-hit combination of both challenges (O2 + IR) followed by 16 h of normoxia (ambient air containing 21% O2 and 5% CO2) (1 cycle = 24 h, 2 cycles = 48 h). Cell survival, DNA damage, apoptosis, and indicators of oxidative stress were evaluated after 1 and 2 cycles of exposure. We observed a significant (p < 0.05) decrease in cell survival across all challenge conditions along with an increase in DNA damage, determined by Comet analysis and H2AX phosphorylation, and apoptosis, determined by Annexin-V staining, relative to cells unexposed to hyperoxia or radiation. DNA damage (GADD45α and cleaved-PARP), apoptotic (cleaved caspase-3 and BAX), and antioxidant (HO-1 and Nqo1) proteins were increased following radiation and hyperoxia exposure after 1 and 2 cycles of exposure. Importantly, exposure to combination challenge O2 + IR exacerbated cell death and DNA damage compared to individual exposures O2 or IR alone. Additionally levels of cell cycle proteins phospho-p53 and p21 were significantly increased, while levels of CDK1 and Cyclin B1 were decreased at both time points for all exposure groups. Similarly, proteins involved in cell cycle arrest was more profoundly changed with the combination challenges as compared to each stressor alone. These results correlate with a significant 4- to 6-fold increase in the ratio of cells in G2/G1 after 2 cycles of exposure to hyperoxic conditions. We have characterized a novel in vitro model of double-hit, low-level radiation and hyperoxia exposure that leads to oxidative lung cell injury, DNA damage, apoptosis, and cell cycle arrest.


Assuntos
Dano ao DNA , Hiperóxia , Modelos Biológicos , Estresse Oxidativo , Radiação Ionizante , Voo Espacial , Células Epiteliais Alveolares/metabolismo , Células Epiteliais Alveolares/efeitos da radiação , Animais , Antioxidantes/metabolismo , Apoptose/genética , Apoptose/efeitos da radiação , Ciclo Celular/genética , Ciclo Celular/efeitos da radiação , Sobrevivência Celular/genética , Sobrevivência Celular/efeitos da radiação , Regulação da Expressão Gênica/efeitos da radiação , Regulação Enzimológica da Expressão Gênica , Histonas/metabolismo , Humanos , Camundongos , Oxirredução , Fosforilação , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
5.
Int J Cancer ; 136(5): 1202-9, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25042623

RESUMO

Although of fundamental importance to the treatment of cancer patients, the quantitative study of drug distribution and action in vivo at the single cell level is challenging. We used the recently-developed technique of mass cytometry to measure cisplatin uptake into individual tumor cells (Pt atoms/cell), combined with measurement of the rate of IdU incorporation into DNA (I(127) atoms/cell/min) and tumor hypoxia identified by the 2-nitroimidazole EF5 in cisplatin-treated BxPC-3 and ME-180 xenografts. Pt levels of 10(5) to 10(6) atoms/cell were obtained following a single cisplatin treatment using clinically relevant doses. Cisplatin caused cell cycle arrest in a dose- and time-dependent manner that paralleled effects in vitro, and it readily penetrated into hypoxic tumor regions. Similar levels of Pt/cell were found in xenografts treated with oxaliplatin. Mass cytometry offers the unique capability to study the cellular uptake and anticancer effects of platinum-containing drugs at the single cell level in animal models, and it has the potential for application to samples obtained from cancer patients during treatment.


Assuntos
Antineoplásicos/farmacologia , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Cisplatino/farmacologia , Hipóxia/fisiopatologia , Neoplasias Pancreáticas/tratamento farmacológico , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Animais , Citometria de Fluxo , Humanos , Hipóxia/tratamento farmacológico , Masculino , Camundongos , Camundongos SCID , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Células Tumorais Cultivadas
6.
Prostate ; 74(8): 805-10, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24677180

RESUMO

BACKGROUND: It has been reported that significant hypoxia may occur in the rat prostate following androgen deprivation (AD). It is well known that hypoxia substantially reduces radiation sensitivity of cells both in vitro and in vivo. Given that contemporary management of men with intermediate and high-risk prostate cancer includes the use of neoadjuvant androgen suppression and radiation, AD-induced hypoxia in the prostate could result in suboptimal therapeutic results. Given this concern, we fully investigate possible AD-induced hypoxia in the ventral prostate (VP) of adult rats by two independent methods. METHODS: Tissue pO2 levels in the VP of adult Spraque-Dawley rats were evaluated prior to and at various time points following castration by two independent techniques. First, an Oxylab tissue oxygen monitor with a 240 µm probe was used for quantitative monitoring of global VP oxygenation. Second, fluorescence immunohistochemistry using the hypoxia marker EF5, known to be metabolically activated by hypoxic cells, was used to evaluate cell-to-cell variation in hypoxia at various days post-castration. RESULTS: Neither the oxygen probe nor EF5 method demonstrate any substantive change in pO2 levels in the rat VP at any time point post-castration. CONCLUSIONS: We find no evidence that the rat VP becomes hypoxic at any point following castration using an animal model that closely mimics the human prostate. These data are in contrast to previous reports suggesting prostatic hypoxia occurs following AD and provide assurance that our present therapeutic strategy of neoadjuvant AD followed by radiation is not compromised by AD-induced tissue hypoxia.


Assuntos
Androgênios/metabolismo , Hipóxia Celular , Orquiectomia , Consumo de Oxigênio/fisiologia , Próstata/metabolismo , Androgênios/deficiência , Animais , Hipóxia Celular/fisiologia , Masculino , Próstata/patologia , Ratos , Ratos Sprague-Dawley
7.
Cancers (Basel) ; 16(7)2024 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-38611062

RESUMO

Prostate cancer is a multi-focal disease that can be treated using surgery, radiation, androgen deprivation, and chemotherapy, depending on its presentation. Standard dose-escalated radiation therapy (RT) in the range of 70-80 Gray (GY) is a standard treatment option for prostate cancer. It could be used at different phases of the disease (e.g., as the only primary treatment when the cancer is confined to the prostate gland, combined with other therapies, or as an adjuvant treatment after surgery). Unfortunately, RT for prostate cancer is associated with gastro-intestinal and genitourinary toxicity. We have previously reported that the metabolic modulator lonidamine (LND) produces cancer sensitization through tumor acidification and de-energization in diverse neoplasms. We hypothesized that LND could allow lower RT doses by producing the same effect in prostate cancer, thus reducing the detrimental side effects associated with RT. Using the Seahorse XFe96 and YSI 2300 Stat Plus analyzers, we corroborated the expected LND-induced intracellular acidification and de-energization of isolated human prostate cancer cells using the PC3 cell line. These results were substantiated by non-invasive 31P magnetic resonance spectroscopy (MRS), studying PC3 prostate cancer xenografts treated with LND (100 mg/kg, i.p.). In addition, we found that LND significantly increased tumor lactate levels in the xenografts using 1H MRS non-invasively. Subsequently, LND was combined with radiation therapy in a growth delay experiment, where we found that 150 µM LND followed by 4 GY RT produced a significant growth delay in PC3 prostate cancer xenografts, compared to either control, LND, or RT alone. We conclude that the metabolic modulator LND radio-sensitizes experimental prostate cancer models, allowing the use of lower radiation doses and diminishing the potential side effects of RT. These results suggest the possible clinical translation of LND as a radio-sensitizer in patients with prostate cancer.

9.
Int J Radiat Oncol Biol Phys ; 117(1): 214-222, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37059234

RESUMO

PURPOSE: FLASH (dose rates >40 Gy/s) radiation therapy protects normal tissues from radiation damage, compared with conventional radiation therapy (∼Gy/m). Radiation-chemical oxygen depletion (ROD) occurs when oxygen reacts with radiation-induced free radicals, so a possible mechanism for FLASH involves radioprotection by the decreased oxygen as ROD occurs. High ROD rates would favor this mechanism, but prior studies have reported low ROD values (∼0.35 µM/Gy) in chemical environments such as water and protein/nutrient solutions. We proposed that intracellular ROD might be much larger, possibly promoted by its strongly reducing chemical environment. METHODS AND MATERIALS: ROD was measured, using precision polarographic sensors, from ∼100 µM to zero in solutions containing intracellular reducing agents ± glycerol (1M), to simulate intracellular reducing and hydroxyl-radical-scavenging capacity. Cs irradiators and a research proton beamline allowed dose rates from 0.0085 to 100 Gy/s. RESULTS: Reducing agents significantly altered ROD values. Most greatly increased ROD but some (eg, ascorbate) actually decreased ROD and additionally imposed an oxygen dependence of ROD at low oxygen concentrations. The highest values of ROD were found at low dose rates, but these montonically decreased with increasing dose rate. CONCLUSIONS: ROD was greatly augmented by some intracellular reducing agents but others (eg, ascorbate) effectively reversed this effect. Ascorbate had its greatest effect at low oxygen concentrations. ROD decreased with increasing dose rate in most cases.


Assuntos
Ácido Ascórbico , Substâncias Redutoras , Humanos , Glicerol , Oxigênio , Prótons
10.
Radiat Res ; 198(2): 181-189, 2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35640166

RESUMO

FLASH is a high-dose-rate form of radiation therapy that has the reported ability, compared with conventional dose rates, to spare normal tissues while being equipotent in tumor control, thereby increasing the therapeutic ratio. The mechanism underlying this normal tissue sparing effect is currently unknown, however one possibility is radiochemical oxygen depletion (ROD) during dose delivery in tissue at FLASH dose rates. In order to investigate this possibility, we used the phosphorescence quenching method to measure oxygen partial pressure before, during and after proton radiation delivery in model solutions and in normal muscle and sarcoma tumors in mice, at both conventional (Conv) (∼0.5 Gy/s) and FLASH (∼100 Gy/s) dose rates. Radiation dosimetry was determined by Advanced Markus Chamber and EBT-XL film. For solutions contained in sealed glass vials, phosphorescent probe Oxyphor PtG4 (1 µM) was dissolved in a buffer (10 mM HEPES) containing glycerol (1 M), glucose (5 mM) and glutathione (5 mM), designed to mimic the reducing and free radical-scavenging nature of the intracellular environment. In vivo oxygen measurements were performed 24 h after injection of PtG4 into the interstitial space of either normal thigh muscle or subcutaneous sarcoma tumors in mice. The "g-value" for ROD is reported in mmHg/Gy, which represents a slight modification of the more standard chemical definition (µM/Gy). In solutions, proton irradiation at conventional dose rates resulted in a g-value for ROD of up to 0.55 mmHg/Gy, consistent with earlier studies using X or gamma rays. At FLASH dose rates, the g-value for ROD was ∼25% lower, 0.37 mmHg/Gy. pO2 levels were stable after each dose delivery. For normal muscle in vivo, oxygen depletion during irradiation was counterbalanced by resupply from the vasculature. This process was fast enough to maintain tissue pO2 virtually unchanged at Conv dose rates. However, during FLASH irradiation there was a stepwise decrease in pO2 (g-value ∼0.28 mmHg/Gy), followed by a rebound to the initial level after ∼8 s. The g-values were smaller and recovery times longer in tumor tissue when compared to muscle and may be related to the lower initial endogenous pO2 levels in the former. Considering that the FLASH effect is seen in vivo even at doses as low as 10 Gy, it is difficult to reconcile the amount of protection seen by oxygen depletion alone. However, the phosphorescence probe in our experiments was confined to the extracellular space, and it remains possible that intracellular oxygen depletion was greater than observed herein. In cell-mimicking solutions the oxygen depletion g-vales were indeed significantly higher than observed in vivo.


Assuntos
Prótons , Sarcoma , Animais , Raios gama , Camundongos , Oxigênio , Radiometria/métodos , Dosagem Radioterapêutica , Sarcoma/radioterapia
11.
Int J Radiat Oncol Biol Phys ; 113(3): 624-634, 2022 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-35314293

RESUMO

PURPOSE: Radiation therapy delivered at ultrafast dose rates, known as FLASH RT, has been shown to provide a therapeutic advantage compared with conventional radiation therapy by selectively protecting normal tissues. Radiochemical depletion of oxygen has been proposed to underpin the FLASH effect; however, experimental validation of this hypothesis has been lacking, in part owing to the inability to measure oxygenation at rates compatible with FLASH. METHODS AND MATERIALS: We present a new variant of the phosphorescence quenching method for tracking oxygen dynamics with rates reaching up to ∼3.3 kHz. Using soluble Oxyphor probes we were able to resolve, both in vitro and in vivo, oxygen dynamics during the time of delivery of proton FLASH. RESULTS: In vitro in solutions containing bovine serum albumin the O2 depletion g values (moles/L of O2 depleted per radiation dose, eg, µM/Gy) are higher for conventional irradiation (by ∼13% at 75 µM [O2]) than for FLASH, and in the low-oxygen region (<25 µM [O2]) they decrease with oxygen concentration. In vivo, depletion of oxygen by a single FLASH is insufficient to achieve severe hypoxia in initially well-oxygenated tissue, and the g values measured appear to correlate with baseline oxygen levels. CONCLUSIONS: The developed method should be instrumental in radiobiological studies, such as studies aimed at unraveling the mechanism of the FLASH effect. The FLASH effect could in part originate from the difference in the oxygen dependencies of the oxygen consumption g values for conventional versus FLASH RT.


Assuntos
Terapia com Prótons , Prótons , Humanos , Pulmão , Oxigênio , Terapia com Prótons/métodos , Radiobiologia , Dosagem Radioterapêutica
12.
Front Oncol ; 12: 1004121, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36518319

RESUMO

Introduction: Radiation-induced oxygen depletion in tissue is assumed as a contributor to the FLASH sparing effects. In this study, we simulated the heterogeneous oxygen depletion in the tissue surrounding the vessels and calculated the proton FLASH effective-dose-modifying factor (FEDMF), which could be used for biology-based treatment planning. Methods: The dose and dose-weighted linear energy transfer (LET) of a small animal proton irradiator was simulated with Monte Carlo simulation. We deployed a parabolic partial differential equation to account for the generalized radiation oxygen depletion, tissue oxygen diffusion, and metabolic processes to investigate oxygen distribution in 1D, 2D, and 3D solution space. Dose and dose rates, particle LET, vasculature spacing, and blood oxygen supplies were considered. Using a similar framework for the hypoxic reduction factor (HRF) developed previously, the FEDMF was derived as the ratio of the cumulative normoxic-equivalent dose (CNED) between CONV and UHDR deliveries. Results: Dynamic equilibrium between oxygen diffusion and tissue metabolism can result in tissue hypoxia. The hypoxic region displayed enhanced radio-resistance and resulted in lower CNED under UHDR deliveries. In 1D solution, comparing 15 Gy proton dose delivered at CONV 0.5 and UHDR 125 Gy/s, 61.5% of the tissue exhibited ≥20% FEDMF at 175 µm vasculature spacing and 18.9 µM boundary condition. This percentage reduced to 34.5% and 0% for 8 and 2 Gy deliveries, respectively. Similar trends were observed in the 3D solution space. The FLASH versus CONV differential effect remained at larger vasculature spacings. A higher FLASH dose rate showed an increased region with ≥20% FEDMF. A higher LET near the proton Bragg peak region did not appear to alter the FLASH effect. Conclusion: We developed 1D, 2D, and 3D oxygen depletion simulation process to obtain the dynamic HRF and derive the proton FEDMF related to the dose delivery parameters and the local tissue vasculature information. The phenomenological model can be used to simulate or predict FLASH effects based on tissue vasculature and oxygen concentration data obtained from other experiments.

13.
Int J Radiat Oncol Biol Phys ; 109(5): 1440-1453, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33186615

RESUMO

PURPOSE: To investigate whether the vascular collapse in tumors by conventional dose rate (CONV) irradiation (IR) would also occur by the ultra-high dose rate FLASH IR. METHODS AND MATERIALS: Lewis lung carcinoma (LLC) cells were subcutaneously implanted in mice. This was followed by CONV or FLASH IR at 15 Gy. Tumors were harvested at 6 or 48 hours after IR and stained for CD31, phosphorylated myosin light chain (p-MLC), γH2AX (a surrogate marker for DNA double strand break), intracellular reactive oxygen species (ROS), or immune cells such as myeloid and CD8α T cells. Cell lines were irradiated with CONV IR for Western blot analyses. ML-7 was intraperitoneally administered daily to LLC-bearing mice for 7 days before 15 Gy CONV IR. Tumors were similarly harvested and analyzed. RESULTS: By immunostaining, we observed that CONV IR at 6 hours resulted in constricted vessel morphology, increased expression of p-MLC, and much higher numbers of γH2AX-positive cells in tumors, which were not observed with FLASH IR. Mechanistically, MLC activation by ROS is unlikely, because FLASH IR produced significantly more ROS than CONV IR in tumors. In vitro studies demonstrated that ML-7, an inhibitor of MLC kinase, abrogated IR-induced γH2AX formation and disappearance kinetics. Lastly, we observed that CONV IR when combined with ML-7 produced some effects similar to FLASH IR, including reduction in the vasculature collapse, fewer γH2AX-positive cells, and increased immune cell influx to the tumors. CONCLUSIONS: FLASH IR produced novel changes in the tumor microenvironment that were not observed with CONV IR. We believe that MLC activation in tumors may be responsible for some of the microenvironmental changes differentially regulated between CONV and FLASH IR.


Assuntos
Carcinoma Pulmonar de Lewis/radioterapia , Cadeias Leves de Miosina/efeitos da radiação , Microambiente Tumoral/efeitos da radiação , Animais , Azepinas/administração & dosagem , Vasos Sanguíneos/patologia , Vasos Sanguíneos/efeitos da radiação , Linfócitos T CD8-Positivos/citologia , Carcinoma Pulmonar de Lewis/irrigação sanguínea , Carcinoma Pulmonar de Lewis/metabolismo , Histonas/metabolismo , Histonas/efeitos da radiação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Cadeias Leves de Miosina/antagonistas & inibidores , Cadeias Leves de Miosina/metabolismo , Naftalenos/administração & dosagem , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/efeitos da radiação , Radioterapia/métodos , Dosagem Radioterapêutica , Espécies Reativas de Oxigênio/metabolismo , Espécies Reativas de Oxigênio/efeitos da radiação
14.
NMR Biomed ; 23(6): 624-32, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20661875

RESUMO

The purpose of the study was to investigate the capability of (1)H MRS and MRI methods for detecting early response to radiation therapy in non-Hodgkin's lymphoma (NHL). Studies were performed on the WSU-DLCL2 xenograft model in nude mice of human diffuse large B-cell lymphoma, the most common form of NHL. Radiation treatment was applied as a single 15 Gy dose to the tumor. Tumor lactate, lipids, total choline, T(2) and apparent diffusion coefficients (ADC) were measured before treatment and at 24 h and 72 h after radiation. A Hadamard-encoded slice-selective multiple quantum coherence spectroscopy sequence was used for detecting lactate (Lac) while a stimulated echo acquisition mode sequence was used for detection of total choline (tCho) and lipids. T(2)- and diffusion-weighted imaging sequences were used for measuring T(2) and ADC. Within 24 h after radiation, significant changes were observed in the normalized integrated resonance intensities of Lac and the methylenes of lipids. Lac/H(2)O decreased by 38 +/- 15% (p = 0.03), and lipid (1.3 ppm, CH(2))/H(2)O increased by 57 +/- 14% (p = 0.01). At 72 h after radiation, tCho/H(2)O decreased by 45 +/- 14% (p = 0.01), and lipid (2.8 ppm, polyunsaturated fatty acid)/H(2)O increased by 970 +/- 36% (p = 0.001). ADC increased by 14 +/- 2% (p = 0.003), and T(2) did not change significantly. Tumor growth delay and regression were observed thereafter. This study enabled comparison of the relative sensitivities of various (1)H MRS and MRI indices to radiation and suggests that (1)H MRS/MRI measurements detect early responses to radiation that precede tumor volume changes.


Assuntos
Linfoma não Hodgkin/patologia , Linfoma não Hodgkin/radioterapia , Imageamento por Ressonância Magnética/métodos , Espectroscopia de Ressonância Magnética/métodos , Transplante Heterólogo/patologia , Animais , Colina/metabolismo , Humanos , Ácido Láctico/metabolismo , Metabolismo dos Lipídeos , Linfoma não Hodgkin/metabolismo , Camundongos , Camundongos Nus , Transplante de Neoplasias
15.
Eur J Nucl Med Mol Imaging ; 37(11): 2048-59, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20585774

RESUMO

PURPOSE: The primary purpose of this study was to assess the biodistribution and radiation dose resulting from administration of (18)F-EF5, a lipophilic 2-nitroimidazole hypoxia marker in ten cancer patients. For three of these patients (with glioblastoma) unlabeled EF5 was additionally administered to allow the comparative assessment of (18)F-EF5 tumor uptake with EF5 binding, the latter measured in tumor biopsies by fluorescent anti-EF5 monoclonal antibodies. METHODS: (18)F-EF5 was synthesized by electrophilic addition of (18)F(2) gas, made by deuteron bombardment of a neon/fluorine mixture in a high-pressure gas target, to an allyl precursor in trifluoroacetic acid at 0° then purified and administered by intravenous bolus. Three whole-body images were collected for each of ten patients using an Allegro (Philips) scanner. Gamma counts were determined in blood, drawn during each image, and urine, pooled as a single sample. PET images were analyzed to determine radiotracer uptake in several tissues and the resulting radiation dose calculated using OLINDA software and standard phantom. For three patients, 21 mg/kg unlabeled EF5 was administered after the PET scans, and tissue samples obtained the next day at surgery to determine EF5 binding using immunohistochemistry techniques (IHC). RESULTS: EF5 distributes evenly throughout soft tissue within minutes of injection. Its concentration in blood over the typical time frame of the study (∼3.5 h) was nearly constant, consistent with a previously determined EF5 plasma half-life of ∼13 h. Elimination was primarily via urine and bile. Radiation exposure from labeled EF5 is similar to other (18)F-labeled imaging agents (e.g., FDG and FMISO). In a de novo glioblastoma multiforme patient, focal uptake of (18)F-EF5 was confirmed by IHC. CONCLUSION: These results confirm predictions of biodistribution and safety based on EF5's characteristics (high biological stability, high lipophilicity). EF5 is a novel hypoxia marker with unique pharmacological characteristics allowing both noninvasive and invasive measurements.


Assuntos
Neoplasias Encefálicas/metabolismo , Etanidazol/análogos & derivados , Radioisótopos de Flúor , Glioblastoma/metabolismo , Hidrocarbonetos Fluorados/metabolismo , Hidrocarbonetos Fluorados/farmacocinética , Transporte Biológico , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/patologia , Hipóxia Celular , Etanidazol/metabolismo , Etanidazol/farmacocinética , Feminino , Glioblastoma/diagnóstico por imagem , Glioblastoma/patologia , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Tomografia por Emissão de Pósitrons , Radiometria , Distribuição Tecidual , Imagem Corporal Total
16.
Curr Mol Med ; 9(4): 401-10, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19519397

RESUMO

Hypoxia is a common characteristic of many solid tumors and is associated with poor prognosis. Cells with low oxygen levels can have altered sensitivity to radiotherapy and chemotherapy secondary to changes in the incidence of DNA single- and double-strand breaks (DNA-ssb, DNA-dsb), DNA base damage, DNA-DNA cross-links and DNA-protein cross-links. Recent evidence also supports that cells exposed to chronic hypoxia have a decreased capacity of DNA-dsb repair. This review will examine the influence of short-term and prolonged hypoxia on the two major pathways of DNA-dsb repair: homologous recombination (HR) and non-homologous end-joining (NHEJ). Novel treatment strategies designed to exploit the hypoxic tumor microenvironment are also discussed. Modification of DNA damage sensing and repair due to fluctuating oxygen levels within a dynamic tumor microenvironment may have profound implications for tumor progression and treatment.


Assuntos
Quebras de DNA de Cadeia Dupla , Quebras de DNA de Cadeia Simples , Reparo do DNA/fisiologia , Neoplasias/genética , Animais , Hipóxia Celular/fisiologia , Dano ao DNA , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Recombinação Genética
17.
Immunohorizons ; 3(4): 149-160, 2019 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-31356176

RESUMO

Tumor hypoxia occurs because of an increased demand for oxygen by the rapidly growing tumor cells, together with reduction in the oxygen supply due to malformed and nonfunctional tumor vasculature. The effects of tumor hypoxia on radiotherapy (RT) are well known; however, recent findings suggest it may also suppress immunotherapy, although the mechanisms governing this observation remain undetermined. Our laboratory and others have shown that IFN-γ conditions the tumor milieu and is important for the efficacy of RT. Thus, we hypothesized that hypoxia could inhibit IFN-γ-mediated antitumor responses, resulting in decreased RT efficacy. This inhibition could involve the production and/or the cellular response to IFN-γ. To test this, we used murine tumor cell lines B16F0 and Colon38. We observed that hypoxia inhibited upregulation of IFN-γ-dependent MHC class I expression by tumor cells along with the gene expression of IFN-γ-dependent chemokines CXCL9 and CXCL10, essential for immune cell infiltration. Furthermore, CD8+ T cells, an important source of IFN-γ, which mediate effector antitumor responses, had reduced ability to proliferate and generate IFN-γ under hypoxic conditions in vitro. Interestingly, reoxygenation restored the cytokine-producing capability of these cells. Studies performed in vivo using a mouse tumor model and the hypoxia marker EF5 demonstrated that RT could reverse the hypoxia within treated tumors. This study has identified a unique mechanism of hypoxia-induced immune suppression involving the downregulation of IFN-γ production and cellular responsiveness to this essential cytokine. These results suggest that therapies that target and reduce tumor hypoxia can potentially boost antitumor immune responses.


Assuntos
Antígenos de Histocompatibilidade Classe I/imunologia , Hipóxia/imunologia , Hipóxia/metabolismo , Imunidade , Interferon gama/metabolismo , Neoplasias/imunologia , Neoplasias/metabolismo , Animais , Biomarcadores , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Regulação Neoplásica da Expressão Gênica , Xenoenxertos , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Hipóxia/genética , Imuno-Histoquímica , Masculino , Camundongos , Neoplasias/genética , Neoplasias/patologia , Especificidade do Receptor de Antígeno de Linfócitos T
18.
Radiat Res ; 169(6): 677-88, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18494550

RESUMO

The binding kinetics of the hypoxia marker EF5 can be quantified by uptake of (14)C-labeled drug or calibrated flow cytometry using antibodies specific for drug adducts. Maximum EF5 binding is cell-line dependent and varies directly with drug exposure (area under the curve; concentration integrated over time) but inversely with pO(2) from 0 to >100 mmHg. For pimonidazole, binding is reported to be independent of the cell line and drug AUC, being zero above 10 mmHg, with an easily discriminated increase at lower pO(2). The basis for these kinetic differences is unknown, but the main experimental variable distinguishing the two marker techniques is antibody concentration ([Ab] - pimonidazole << EF5). In this study, EF5 and pimonidazole binding kinetics were compared as a function of pO(2) and antibody concentration in cells of two rat (9L and R3230) and two human (HT1080 and SiHa) cancer cell lines. For both markers, binding varied directly with AUC at all pO(2). The dynamic range of observed binding (maximum change from 0 to 76 mmHg oxygen) decreased with antibody concentration. The pO(2) dependence of binding for pimonidazole, but not EF5, varied dramatically with antibody concentration. Thus the data presented herein do not support the reported binding kinetics of pimonidazole. In particular, it is shown that the common use of antibody concentrations much lower than antigen concentrations can lead to unreliable estimations of adduct level and hence pO(2).


Assuntos
Hipóxia Celular , Etanidazol/análogos & derivados , Citometria de Fluxo/métodos , Hidrocarbonetos Fluorados/farmacologia , Nitroimidazóis/farmacologia , Radiossensibilizantes/farmacologia , Animais , Área Sob a Curva , Calibragem , Linhagem Celular Tumoral , Etanidazol/farmacologia , Humanos , Cinética , Microscopia de Fluorescência/métodos , Oxigênio/química , Oxigênio/metabolismo , Ratos , Fatores de Tempo
19.
Radiat Res ; 170(6): 677-90, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19138031

RESUMO

Tissue hypoxia results from the interaction of cellular respiration, vascular oxygen carrying capacity, and vessel distribution. We studied the relationship between tumor vasculature and regions of low pO(2) using quantitative analysis of binding of the 2-nitroimidazole EF5 given to patients intravenously (21 mg/kg) approximately 24 h preceding surgery. We describe new computer algorithms for determining EF5 binding as a function of radial distance from individual blood vessels and converting this value to tissue pO(2). Tissues from six human brain tumors were assessed. In a hemangiopericytoma, a WHO Grade 2 and WHO Grade 3 glial brain tumor, all tissue pO(2) values calculated by EF5 binding were >20 mmHg (described as "physiologically oxygenated"). In these three tumors, EF5 binding gradients (measured as a function of distance from each observed vessel) were low, with small positive and negative values averaging close to zero. Much lower tissue oxygen levels were found, including near some vessels, in glioblastomas. Gradients of EF5 binding away from vessels were larger in glioblastomas than in the low-grade tumors, but positive and negative values again averaged to near zero. Based on these preliminary data, we hypothesize a new paradigm for tumor blood flow in human brain tumors whereby in-flowing and out-flowing blood patterns may have contrasting effects on average tissue EF5 (and by inference, oxygen) gradients. Our studies also imply that neither distance to the nearest blood vessel nor distance from each observed blood vessel provide reliable estimates of tissue pO(2).


Assuntos
Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/patologia , Hipóxia/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Algoritmos , Animais , Neoplasias Encefálicas/metabolismo , Etanidazol/análogos & derivados , Etanidazol/metabolismo , Glioblastoma/irrigação sanguínea , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Hidrocarbonetos Fluorados/metabolismo , Camundongos , Pessoa de Meia-Idade , Oxigênio/metabolismo
20.
Int J Radiat Biol ; 84(2): 151-64, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18246483

RESUMO

PURPOSE: We describe variations of three radiation-induced endpoints on the basis of cell genotype: Clonogenic survival, expression of apoptosis and cell-cycle redistribution. METHODS: Clonogenic survival, apoptosis and cell-cycle redistribution are measured in multiple cell lines after exposure to radiation between 2 and 16 Gy. Cell lines varied in clonogenic radiosensitivity and expression of specific genes. RESULTS: Clonal radiosensitivity is genotype-dependent, associating with four specific genes: A mutated form of Ataxia telangiectasia mutated (mutATM); with two forms of TP53, the gene that is template for tumor protein p53, wildtype TP53 (wtTP53) and mutated TP53 (mutTP53); and an unidentified gene in radioresistant glioblastoma cells. Apoptosis is also genotype-dependent showing elevated levels in cells that express mutATM and abrogated 14-3-3sigma (an isoform of the 14-3-3 gene) but less variation for different forms of TP53. Cell-cycle redistribution varied in mutATM cells. Kinetics of apoptosis are biphasic for both time and dose; cell lines did not express apoptosis at doses below 5 Gy or times before 24 hours. Kinetics of cell-cycle redistribution changed dynamically in the first 24 hours but showed little change after that time. CONCLUSIONS: Clonogenic survival, radiation-induced apoptosis and radiation-induced redistribution in the cell-cycle vary with cell genotype, but not the same genotypes. There is temporal, not quantitative, correlation between apoptosis and clonal radiosensitivity with apoptosis suppressed by lower, less toxic doses of radiation (<5 Gy) but enabled after larger, more toxic doses. Kinetic patterns for apoptosis and redistribution show a common change at approximately 24 hours.


Assuntos
Apoptose/efeitos da radiação , Ciclo Celular/efeitos da radiação , Glioblastoma/radioterapia , Tolerância a Radiação/genética , Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo , Proteínas 14-3-3/efeitos da radiação , Apoptose/genética , Apoptose/fisiologia , Proteínas Mutadas de Ataxia Telangiectasia , Ciclo Celular/genética , Ciclo Celular/fisiologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/efeitos da radiação , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Sobrevivência Celular/efeitos da radiação , Clonagem Molecular , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/efeitos da radiação , Relação Dose-Resposta à Radiação , Genes p53/genética , Genótipo , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Cinética , Mutação , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/efeitos da radiação , Tolerância a Radiação/fisiologia , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteína Supressora de Tumor p53/efeitos da radiação , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Proteínas Supressoras de Tumor/efeitos da radiação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA