Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Hyperthermia ; 39(1): 405-413, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35236209

RESUMEN

BACKGROUND: Enediynes are anti-cancer agents that are highly cytotoxic due to their propensity for low thermal activation of radical generation. The diradical intermediate produced from Bergman cyclization of the enediyne moiety may induce DNA damage and cell lethality. The cytotoxicity of enediynes and difficulties in controlling their thermal cyclization has limited their clinical use. We recently showed that enediyne toxicity at 37 °C can be mitigated by metallation, but cytotoxic effects of 'metalloenediynes' on cultured tumor cells are potentiated by hyperthermia. Reduction of cytotoxicity at normothermia suggests metalloenediynes will have a large therapeutic margin, with cell death occurring primarily in the heated tumor. Based on our previous in vitro findings, FeSO4-PyED, an Fe co-factor complex of (Z)-N,N'-bis[1-pyridin-2-yl-meth-(E)-ylidene]oct-4-ene-2,6-diyne-1,8-diamine, was prioritized for further in vitro and in vivo testing in normal human melanocytes and melanoma cells. METHODS: Clonogenic survival, apopotosis and DNA binding assays were used to determine mechanisms of enhancement of FeSO4-PyED cytotoxicity by hyperthermia. A murine human melanoma xenograft model was used to assess in vivo efficacy of FeSO4-PyED at 37 or 42.5 °C. RESULTS: FeSO4-PyED is a DNA-binding compound. Enhancement of FeSO4-PyED cytotoxicity by hyperthermia in melanoma cells was due to Bergman cyclization, diradical formation, and increased apoptosis. Thermal enhancement, however, was not observed in melanocytes. FeSO4-PyED inhibited tumor growth when melanomas were heated during drug treatment, without inducing normal tissue damage. CONCLUSION: By leveraging the unique thermal activation properties of metalloenediynes, we propose that localized moderate hyperthermia can be used to confine the cytotoxicity of these compounds to tumors, while sparing normal tissue.


Asunto(s)
Antineoplásicos , Hipertermia Inducida , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Ciclización , Enediinos/química , Enediinos/farmacología , Enediinos/uso terapéutico , Calor , Humanos , Ratones
2.
Nucleic Acids Res ; 48(20): 11536-11550, 2020 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-33119767

RESUMEN

DNA-dependent protein kinase (DNA-PK) plays a critical role in the non-homologous end joining (NHEJ) repair pathway and the DNA damage response (DDR). DNA-PK has therefore been pursued for the development of anti-cancer therapeutics in combination with ionizing radiation (IR). We report the discovery of a new class of DNA-PK inhibitors that act via a novel mechanism of action, inhibition of the Ku-DNA interaction. We have developed a series of highly potent and specific Ku-DNA binding inhibitors (Ku-DBi's) that block the Ku-DNA interaction and inhibit DNA-PK kinase activity. Ku-DBi's directly interact with the Ku and inhibit in vitro NHEJ, cellular NHEJ, and potentiate the cellular activity of radiomimetic agents and IR. Analysis of Ku-null cells demonstrates that Ku-DBi's cellular activity is a direct result of Ku inhibition, as Ku-null cells are insensitive to Ku-DBi's. The utility of Ku-DBi's was also revealed in a CRISPR gene-editing model where we demonstrate that the efficiency of gene insertion events was increased in cells pre-treated with Ku-DBi's, consistent with inhibition of NHEJ and activation of homologous recombination to facilitate gene insertion. These data demonstrate the discovery and application of new series of compounds that modulate DNA repair pathways via a unique mechanism of action.


Asunto(s)
Reparación del ADN por Unión de Extremidades/efectos de los fármacos , Proteína Quinasa Activada por ADN/antagonistas & inhibidores , Autoantígeno Ku/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Animales , Células Cultivadas , ADN/química , Roturas del ADN de Doble Cadena , Edición Génica , Humanos , Autoantígeno Ku/química , Ratones , Inhibidores de Proteínas Quinasas/química
3.
NAR Cancer ; 5(1): zcad003, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36755959

RESUMEN

The DNA-dependent protein kinase (DNA-PK) plays a critical role in the DNA damage response (DDR) and non-homologous end joining (NHEJ) double-strand break (DSB) repair pathways. Consequently, DNA-PK is a validated therapeutic target for cancer treatment in certain DNA repair-deficient cancers and in combination with ionizing radiation (IR). We have previously reported the discovery and development of a novel class of DNA-PK inhibitors with a unique mechanism of action, blocking the Ku 70/80 heterodimer interaction with DNA. These Ku-DNA binding inhibitors (Ku-DBi's) display nanomolar activity in vitro, inhibit cellular DNA-PK, NHEJ-catalyzed DSB repair and sensitize non-small cell lung cancer (NSCLC) cells to DSB-inducing agents. In this study, we demonstrate that chemical inhibition of the Ku-DNA interaction potentiates the cellular effects of bleomycin and IR via p53 phosphorylation through the activation of the ATM pathway. This response is concomitant with a reduction of DNA-PK catalytic subunit (DNA-PKcs) autophosphorylation at S2056 and a time-dependent increase in H2AX phosphorylation at S139. These results are consistent with Ku-DBi's abrogating DNA-PKcs autophosphorylation to impact DSB repair and DDR signaling through a novel mechanism of action, and thus represent a promising anticancer therapeutic strategy in combination with DNA DSB-inducing agents.

4.
Radiat Res ; 197(1): 1-6, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34788470

RESUMEN

Recent epidemiological and experimental animal data, as well as reanalyses of data previously accumulated, indicate that the lens of the eye is more radiosensitive than was previously thought. This has resulted in a reduction of the occupational lens dose limit within the European Union countries, Japan and elsewhere. This Commentary introduces the work done by the LDLensRad Consortium contained within this Focus Issue, towards advancement of understanding of the mechanisms of low dose radiation cataract.


Asunto(s)
Catarata/etnología , Cristalino/efectos de la radiación , Animales , Relación Dosis-Respuesta en la Radiación , Europa (Continente) , Humanos , Japón , Ratones Endogámicos C57BL , Exposición Profesional , Dosis de Radiación , Tolerancia a Radiación
5.
Int J Hyperthermia ; 27(5): 435-44, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21756041

RESUMEN

PURPOSE: Enediynes are potent inducers of DNA damage, but their clinical usefulness has been limited. Here we report the thermal enhancement of cytotoxicity of two novel metalloenediyne compounds at concentrations that are either not or minimally cytotoxic at 37°C, and present evidence regarding possible mechanisms for enhanced cytotoxicity. MATERIALS AND METHODS: HeLa cells were exposed to (Z)-N,N'-bis[1-pyridyl-2-yl-meth-(E)-ylidene]octa-4-ene-2,6-diyne-1,8-diamine (PyED) (which becomes metallated in culture medium) or ((Z)-N,N'-bis[quinolin-2-yl-meth-(E)-ylidene]octa-4-ene-2,6-diyne-1,8-diamine)zinc(II) chloride (QuinED · ZnCl(2)) at 37°C or 42.5°C for 1 h, and clonogenic survival was compared after treatment at each temperature. Analyses of cell cycle progression and mode of death were performed after treatments. RESULTS: Treatment with PyED or QuinED · ZnCl(2) resulted in a significant decrease in cell survival when cells were treated with drug at 42.5°C compared to 37°C. Enhanced cytotoxicity was attributed to increased apoptosis. However, perturbation of the cell cycle may also play a role. Cells which were only heated or exposed to PyED at 37°C experienced significant G(2)/M blocks that were eliminated when PyED and heat were administered simultaneously, suggesting that combined treatments override cell cycle arrests normally observed with each agent individually. Conversely, cells heated during treatment with QuinED · ZnCl(2) displayed an increased G(2)/M arrest compared to treatment at 37°C. CONCLUSIONS: With improvements in site-specific heat delivery to tumours, systemic administration of non-toxic metalloenediynes coupled with localised hyperthermia may improve selective enediyne activation/targeting. Therefore PyED and QuinED · ZnCl(2), which show significantly enhanced cytotoxicity at elevated temperatures, may represent viable candidates for thermochemotherapy.


Asunto(s)
Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Complejos de Coordinación/toxicidad , Enediinos/toxicidad , Hipertermia Inducida , Compuestos Organometálicos/toxicidad , Piridinas/toxicidad , Supervivencia Celular , Terapia Combinada , Sinergismo Farmacológico , Células HeLa , Calor , Humanos
6.
Cytometry A ; 77(10): 940-52, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21290468

RESUMEN

Radiosensitization of mammalian cells by heat is believed to involve the inhibition of repair of DNA double-strand breaks (DSBs). The Mre11 complex (composed of Mre11, Rad50, and Nbs1) is involved in DSB repair and forms foci at sites of radiation-induced DSBs. Heat induces the translocation of a significant amount of Mre11, Rad50, and Nbs1 from the nucleus to the cytoplasm, but little is known about how heat affects the integrity of the proteins still remaining in nuclei, or alters kinetics of formation/disappearance of DNA repair foci in heated, irradiated cells. Here, we show that hyperthermia alters the interaction between proteins of the Mre11 complex in irradiated human melanoma cells and inhibits the formation of repair foci. At various times after X-irradiation and/or heating (2 h at 41.5 or 42.5 °C), the cells were fixed and stained for Mre11, Rad50, and Nbs1. Colocalization of proteins and formation and disappearance of nuclear foci in heated and/or irradiated cells, determined using confocal microscopy, were compared. In heated, irradiated cells, focus formation was inhibited for 2-8 h, and colocalization of the proteins of the Mre11 complex was reduced for 12-24 h post-treatment. Colocalization was recovered in irradiated cells within 24 h after heating at 41.5 °C, but was inhibited longer after heating at 42.5 °C. The decreased colocalization in heated, irradiated cells suggests that there is a decrease in protein interaction, and Mre11 complexes in nuclei disassemble after heating. Such changes could be involved, at least in part, in heat radiosensitization and inhibition of DSB repair. Also, the kinetics of disassembly and reassembly of Mre11 complexes appears to be dependent upon treatment temperature.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Calor , Tolerancia a Radiación , Ácido Anhídrido Hidrolasas , Línea Celular Tumoral , Núcleo Celular/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN/fisiología , Reparación del ADN/efectos de la radiación , Enzimas Reparadoras del ADN/metabolismo , Humanos , Cinética , Proteína Homóloga de MRE11 , Microscopía Confocal
7.
Radiat Res ; 194(5): 557-565, 2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-33045089

RESUMEN

Astronauts participating in prolonged space missions constitute a population of individuals who are at an increased risk for cataractogenesis due to exposure to densely ionizing charged particles. Using a rat model, we have previously shown that after irradiation of eyes with either low-linear energy transfer (LET) 60Co γ rays or high-LET 56Fe particles, the rate of progression of anterior and posterior subcapsular cataracts was significantly greater in ovariectomized females implanted with 17-ß-estradiol (E2) compared to ovariectomized or intact rats. However, our additional low-LET studies indicated that cataractogenesis may be a modifiable late effect, since we have shown that the modulation of cataractogenesis is dependent upon the timing of administration of E2. Interestingly, we found that E2 protected against cataractogenesis induced by low-LET radiation, but only if administered after the exposure; if administered prior to and after irradiation, for the entire period of observation, then E2 enhanced progression and incidence of cataracts. Since most radioprotectors tested to date are unsuccessful in protecting against the effects of high-LET radiation, we wished to determine whether the protection mediated by E2 against radiation cataractogenesis induced by low-LET radiation would also be observed after high-LET irradiation. Female 56-day-old Sprague-Dawley rats were treated with E2 at various times relative to the time of single-eye irradiation with 2 Gy of 56Fe ions. We found that administration of E2 before irradiation and throughout the lifetime of the rat enhanced cataractogenesis compared to ovariectomized animals. The enhancing effect was slightly reduced when estrogen was removed after irradiation. However, in contrast to what we observed after γ-ray irradiation, there was no inhibition of cataractogenesis if E2 was administered only after 56Fe-ion irradiation. We conclude that protection against cataractogenesis by estrogen is dependent upon the type and ionization density of radiation that the lens was exposed to. The lack of inhibition of radiation cataractogenesis in rats that receive E2 treatment after high-LET irradiation may be attributed to the qualitative differences in the types of DNA damage induced with high-LET radiation compared to low-LET radiation or how damage may be modified at the DNA or tissue level after irradiation.


Asunto(s)
Catarata/prevención & control , Radioisótopos de Cobalto , Estradiol/uso terapéutico , Rayos gamma/efectos adversos , Iones Pesados/efectos adversos , Hierro , Traumatismos Experimentales por Radiación/prevención & control , Medicina Aeroespacial , Animales , Catarata/etiología , Esquema de Medicación , Implantes de Medicamentos , Estradiol/administración & dosificación , Incidencia , Transferencia Lineal de Energía , Ovariectomía , Ratas , Ratas Sprague-Dawley
8.
Radiat Res ; 193(2): 107-118, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31800359

RESUMEN

Enediyne natural products are a class of compounds that were recognized for their potential as chemotherapeutic agents many years ago, but found to be highly cytotoxic due to their propensity for low thermal activation. Bergman cyclization of the enediyne moiety produces a diradical intermediate, and may subsequently induce DNA damage and account for the extreme cytotoxicity. While difficulties in controlling the thermal cyclization reaction have limited the clinical use of cyclic enediynes, we have previously shown that enediyne activity, and thus toxicity at physiological temperatures can be modulated by metallation of acyclic enediynes. Furthermore, the cytotoxicity of "metalloenediynes" can be potentiated by hyperthermia. In this study, we characterized a suite of novel metallated enediyne motifs that usually induced little or no cytotoxicity when two different human cancer cell lines were treated with the compounds at 37°C, but showed a significant enhancement of cytotoxicity after cells were exposed to moderate hyperthermia during drug treatment. Cultured U-1 melanoma or MDA-231 breast cancer cells were treated with various concentrations of Cu, Fe and Zn complexes of the enediyne (Z)-N,N'-bis[1-pyridyl-2-yl-meth-(E)-ylidene]octa-4-ene-2,6-diyne-1,8-diamine (PyED) and clonogenic survival was assessed to determine the effects of the drugs at 37°C and 42.5°C. Toxicity at 37°C varied for each compound, but hyperthermia potentiated the cytotoxicity of each compound in both cell lines. Cytotoxicity was concentration-, time- and temperature-dependent. Heating cells during drug treatment resulted in enhanced apoptosis, but the role of cell cycle perturbation in the response of the cells to the drugs was less clear. Lastly, we showed that hyperthermia enhanced the number of DNA double-strand breaks (DSBs) induced by the compounds, and inhibited their repair after drug treatment. Thus, thermal enhancement of cytotoxicity may be due, at least in part, to the propensity of the enediyne moiety to induce DSBs, and/or a reduction in DSB repair efficiency. We propose that "tuning" of metalloenediyne toxicity through better-controlled reactivity could have potential clinical utility, since we envision that such compounds could be administered systemically as relatively non-toxic agents, but cytotoxicity could be enhanced in, and confined to a tumor volume when subjected to localized heating.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Complejos de Coordinación/química , Complejos de Coordinación/farmacología , Enediinos/química , Hipertermia Inducida , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Humanos
9.
Cytometry A ; 75(3): 245-52, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18836995

RESUMEN

Upon induction of DNA double-strand breaks (DSBs), Mre11 and Rad50 proteins of the Mre11 DNA repair complex accumulate at the sites of DSBs and form discrete nuclear foci. Precision in scoring of Mre11/Rad50-containing foci depends upon detection of those foci, some of which have a fluorescence staining intensity that is too close to the fluorescence staining intensity of the remaining Mre11 and Rad50 proteins that have not been incorporated into foci. Human U-1 melanoma cells in exponential growth were irradiated with various doses of X-rays (0-12 Gy) to induce the formation of repair foci. Four hours after irradiation, cells were simultaneously labeled for Mre11 and Rad50 proteins, using a two-color immunofluorescence staining technique. Laser scanning confocal microscopy was used to collect the composite images of randomly selected cell nuclei. Intensity correlation analysis (ICA) of equally intense fluorescence signals from Mre11 and Rad50 proteins was performed to obtain the regions with correlated pixels. ICA permitted enhanced detection of low level fluorescence of Mre11/Rad50 foci ("hidden" foci) that can be barely detected upon imaging of only one protein. For example, while imaging of only one protein (either Mre11 or Rad50) in the nucleus of a 6 Gy-irradiated cell revealed 9 foci, imaging of two proteins with ICA revealed 11 foci. ICA permitted an evaluation of the dose dependence of nuclear foci in cells irradiated with various doses of X-rays, with focus formation increasing up to a dose of 6 Gy. Our data accumulated using two-color immunofluorescence staining of Mre11 and Rad50 proteins and ICA of these two target proteins provide a basis for enhanced detection and accuracy in the scoring of DNA repair foci.


Asunto(s)
Núcleo Celular/metabolismo , Reparación del ADN , Técnica del Anticuerpo Fluorescente/métodos , Ácido Anhídrido Hidrolasas , Línea Celular Tumoral , Roturas del ADN de Doble Cadena , Enzimas Reparadoras del ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Humanos , Proteína Homóloga de MRE11 , Microscopía Confocal
10.
Radiat Res ; 171(1): 9-21, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19138055

RESUMEN

The molecular mechanisms governing acquired tumor resistance during radiotherapy remain to be elucidated. In breast cancer patients, overexpression of HER2 (human epidermal growth factor receptor 2) is correlated with aggressive tumor growth and increased recurrence. In the present study, we demonstrate that HER2 expression can be induced by radiation in breast cancer cells with a low basal level of HER2. Furthermore, HER2-postive tumors occur at a much higher frequency in recurrent invasive breast cancer (59%) compared to the primary tumors (41%). Interestingly, NF-kappaB is required for radiation-induced HER2 transactivation. HER2 was found to be co-activated with basal and radiation-induced NF-kappaB activity in radioresistant but not radiosensitive breast cancer cell lines after long-term radiation exposure, indicating that NF-kappaB-mediated HER2 overexpression is involved in radiation-induced repopulation in heterogeneous tumors. Finally, we found that inhibition of HER2 resensitizes the resistant cell lines to radiation. Since HER2 is shown to activate NF-kappaB, our data suggest a loop-like HER2-NF-kappaB-HER2 pathway in radiation-induced adaptive resistance in breast cancer cells.


Asunto(s)
Adaptación Fisiológica/genética , Neoplasias de la Mama/patología , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Genes erbB-2/genética , Genes erbB-2/efectos de la radiación , FN-kappa B/metabolismo , Tolerancia a Radiación/genética , Animales , Benzamidas/farmacología , Neoplasias de la Mama/genética , Neoplasias de la Mama/prevención & control , Neoplasias de la Mama/radioterapia , Línea Celular Tumoral , Rayos gamma/efectos adversos , Humanos , Ratones , FN-kappa B/antagonistas & inhibidores , Fenotipo , Regiones Promotoras Genéticas/genética , Regiones Promotoras Genéticas/efectos de la radiación , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Recurrencia , Transducción de Señal/genética , Transducción de Señal/efectos de la radiación , Regulación hacia Arriba/efectos de la radiación
11.
Radiat Res ; 172(1): 129-33, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19580515

RESUMEN

Radiation cataractogenesis is an important consideration for radiotherapy patients and for astronauts. Data in the literature suggest that gender and/or estrogen may play a role in the incidence of age-related cataracts. However, few data exist on the effect of gender on radiation-induced cataractogenesis. We compared the incidence and rate of progression of cataracts induced by ionizing radiation in male and female Sprague-Dawley rats. Male rats were implanted with either an empty silastic capsule or a capsule containing 17-beta-estradiol. Ovary-intact female rats were implanted with empty capsules. All rats received a single dose of 10 Gy (60Co gamma rays) to the right eye only. Lens opacification was measured at 2-4-week intervals with a slit lamp. The incidence of radiation-induced cataracts was significantly increased in male rats compared to female rats (P=0.034). There was no difference in the rate of cataract progression between the three groups. Our data suggest there is a gender-related difference in radiation-induced cataractogenesis, but the increased incidence of radiation cataractogenesis in male rats compared to female rats cannot be attributed to estrogen levels, since there was no difference in cataract incidence between male rats implanted with empty capsules and those implanted with capsules containing 17-beta-estradiol.


Asunto(s)
Catarata/etiología , Catarata/patología , Estradiol/metabolismo , Traumatismos Experimentales por Radiación/complicaciones , Traumatismos Experimentales por Radiación/patología , Caracteres Sexuales , Animales , Progresión de la Enfermedad , Femenino , Rayos gamma/efectos adversos , Cristalino/patología , Cristalino/efectos de la radiación , Modelos Lineales , Masculino , Ratas , Ratas Sprague-Dawley
12.
Anticancer Res ; 29(4): 1319-25, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19414382

RESUMEN

BACKGROUND: The mechanism by which heat sensitizes mammalian cells to ionizing radiation remains to be elucidated. We determined whether base excision repair (BER) is involved in heat-radiosensitization and report novel findings that provide insight regarding the role of BER in the radiation response of HeLa cells. MATERIALS AND METHODS: An siRNA approach was utilized to suppress expression of AP endonuclease (Ape1), a critical enzyme of BER. Clonogenic survival curves were obtained for HeLa cells expressing normal or reduced Ape1 content and which had been irradiated, and these were compared to survival curves from cells that were irradiated prior to hyperthermia treatment. RESULTS: The amount of heat-radiosensitization observed in Ape1-suppressed cells was similar to or slightly greater than that observed in cells expressing near-normal levels of Ape1. Interestingly, we also found that for unheated HeLa cells, suppressed expression of Ape1 resulted in enhanced resistance to X-rays. CONCLUSION: The data suggest that Ape1, and therefore BER, is not involved in heat-radiosensitization. However, the observation that suppressed expression of Ape1 results in enhanced radioresistance supports the notion that BER may be detrimental to the survival of irradiated cells.


Asunto(s)
Reparación del ADN/efectos de la radiación , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Calor , Tolerancia a Radiación , Western Blotting , ADN-(Sitio Apurínico o Apirimidínico) Liasa/antagonistas & inhibidores , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Células HeLa , Humanos , ARN Interferente Pequeño/farmacología , Rayos X
13.
Radiat Res ; 191(1): 31-42, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30339056

RESUMEN

Several investigators performing bone marrow transplantation studies have previously reported sporadic increases in mortality that were associated with pronounced swelling in the face, head and neck of mice. Over the past few years, we and others have noted an increasing number of experiments in which mice that have received total-body irradiation (TBI) or partial-body irradiation (PBI) develop swollen muzzles, drastic thickening of the upper lip and redness, bruising and/or swelling around the nose and muzzle and sometimes over the top of the head. We refer to this rapid and extreme swelling after irradiation as swollen muzzle syndrome (SMS). The development of SMS postirradiation is associated with morbidity that occurs earlier than would be expected from the traditional hematopoietic acute radiation syndrome (H-ARS), and has impeded studies in several laboratories attempting to evaluate medical countermeasures (MCM) against radiation. However, little has been done to characterize this somewhat unpredictable radiation effect. To investigate the cause and etiology of SMS, data from three different laboratories collected over a seven-year period from 100 MCM 30-day survival studies using mice from different vendors were retrospectively analyzed to determine the time of onset, progression and incidence of SMS in male and female mice exposed to various doses of ionizing radiation. An additional study compared incidence and etiology of SMS in mice from two different vendors (identified as vendors A and B) after exposure to the LD50/30 (X rays). Mice presenting with SMS, as well as non-SMS (irradiated) control mice, were necropsied to determine microbial status of the blood, heart, spleen, liver, kidney and muzzle tissue. Only mice from vendor A (20%) developed SMS. While the number of bacterial species isolated from various tissues of SMS and non-SMS mice was not different, the number of tissues positive for bacteria was significantly greater in SMS mice. At least one tissue in 83% of SMS mice from vendor A tested positive for Streptococcus agalactiae [group B beta Streptococcus (GBS)], compared to 25% of non-SMS mice from vendor A, and 0% of non-SMS mice from vendor B. In addition, all mice from vendor A with SMS had at least one tissue with >104 CFU/g, with GBS as the predominant bacterium, compared to only 25% of non-SMS mice from vendor A, and 0% of non-SMS mice from vendor B. The incidence and magnitude of GBS growth in cultures correlated with the onset of SMS; the earliest and heaviest infections occurred in mice presenting with SMS on days 5-6 postirradiation. The majority of SMS mice (5 out of 6) had positive blood cultures, with the same bacterial strain isolated from other tissues, suggesting systemic translocation via the bloodstream. We propose that testing of mice and the identification of the microorganisms frequently associated with SMS may provide guidance for selection of antimicrobials for use by other investigators in studies evaluating potential MCM, and for the ordering, handling and care of immunodeficient mice or mice that are to be rendered immunodeficient after acute irradiation.


Asunto(s)
Edema/etiología , Cara/efectos de la radiación , Cabeza/efectos de la radiación , Cuello/efectos de la radiación , Traumatismos Experimentales por Radiación/etiología , Síndrome de Radiación Aguda/etiología , Síndrome de Radiación Aguda/patología , Animales , Edema/patología , Cara/patología , Femenino , Cabeza/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Cuello/patología , Traumatismos Experimentales por Radiación/patología , Estudios Retrospectivos , Irradiación Corporal Total/efectos adversos
14.
Radiat Res ; 170(6): 758-64, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19138041

RESUMEN

Cataractogenesis is a complication of radiotherapy when the eye is included in the treatment field. Low doses of densely ionizing space radiation may also result in an increased risk of cataracts in astronauts. We previously reported that estrogen (17-beta-estradiol), when administered to ovariectomized rats commencing 1 week before gamma irradiation of the eye and continuously thereafter, results in a significant increase in the rate and incidence of cataract formation and a decreased latent period compared to an ovariectomized control group. We therefore concluded that estrogen accelerates progression of radiation-induced opacification. We now show that estrogen, if administered continuously, but commencing after irradiation, protects against radiation cataractogenesis. Both the rate of progression and incidence of cataracts were greatly reduced in ovariectomized rats that received estrogen treatment after irradiation compared to ovariectomized rats. As in our previous study, estradiol administered 1 week prior to irradiation at the time of ovariectomy and throughout the period of observation produced an enhanced rate of cataract progression. Estrogen administered for only 1 week prior to irradiation had no effect on the rate of progression but resulted in a slight reduction in the incidence. We conclude that estrogen may enhance or protect against radiation cataractogenesis, depending on when it is administered relative to the time of irradiation, and may differentially modulate the initiation and progression phases of cataractogenesis. These data have important implications for astronauts and radiotherapy patients.


Asunto(s)
Catarata/etiología , Catarata/prevención & control , Estrógenos/farmacología , Traumatismos Experimentales por Radiación/prevención & control , Animales , Catarata/patología , Estrógenos/administración & dosificación , Ojo/efectos de los fármacos , Ojo/patología , Ojo/efectos de la radiación , Femenino , Dosis de Radiación , Traumatismos Experimentales por Radiación/patología , Oncología por Radiación , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
15.
Radiat Res ; 190(2): 107-116, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29763378

RESUMEN

Enediynes are a highly cytotoxic class of compounds. However, metallation of these compounds may modulate their activation, and thus their cytotoxicity. We previously demonstrated that cytotoxicity of two different metalloenediynes, including (Z)-N,N'-bis[1-pyridyl-2-yl-meth-(E)-ylidene]octa-4-ene-2,6-diyne-1,8-diamine] (PyED), is potentiated when the compounds are administered to HeLa cells during hyperthermia treatment at concentrations that are minimally or not cytotoxic at 37°C. In this study, we further characterized the concentration, time and temperature dependence of cytotoxicity of PyED on human U-1 melanoma cells. We also investigated the potential mechanisms by which PyED cytotoxicity is enhanced during hyperthermia treatment. Cell killing with PyED was dependent on concentration, temperature during treatment and time of exposure. Potentiation of cytotoxicity was observed when cells were treated with PyED at temperatures ≥39.5°C, and enhancement of cell killing increased with temperature and with increasing time at a given temperature. All cells treated with PyED were shown to have DNA damage, but substantially more damage was observed in cells treated with PyED during heating. DNA repair was also inhibited in cells treated with the drug during hyperthermia. Thus, potentiation of PyED cytotoxicity by hyperthermia may be due to enhancement of drug-induced DNA lesions, and/or the inhibition of repair of sublethal DNA damage. While the selective thermal activation of PyED supports the potential clinical utility of metalloenediynes as cancer thermochemotherapeutic agents, therapeutic gain could be optimized by identifying compounds that produce minimal toxicity at 37°C but which become activated and show enhancement of cytotoxicity within a tumor subjected to localized hyperthermic or thermal ablative treatment, or which might act as bifunctional agents. We thus also describe the development and initial characterization of a novel cofactor complex of PyED, platinated PyED (Pt-PyED). Pt-PyED binds to DNA-like cisplatin, and much like PyED, cytotoxicity is greatly enhanced after treatment with the drug at elevated temperatures. However, in contrast to PyED, Pt-PyED is only minimally cytotoxic at 37°C, at concentrations at which cytotoxicity is enhanced by hyperthermia. Further development of cisplatin-based enediynes may result in compounds which, when activated, will possess multiple DNA binding modalities similar to cisplatin, but produce less side effects in tissues at normothermic temperatures.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Enediinos/química , Melanoma/patología , Compuestos Organometálicos/química , Compuestos Organometálicos/farmacología , Temperatura , Antineoplásicos/metabolismo , Línea Celular Tumoral , Daño del ADN , Reparación del ADN/efectos de los fármacos , Humanos , Hipertermia Inducida , Compuestos Organometálicos/metabolismo , Factores de Tiempo
16.
Radiat Res ; 168(2): 262-5, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17722362

RESUMEN

Nearly all residents from accredited radiation oncology residency programs in the United States are required to take the American College of Radiology (ACR) In-Training examination each year. The test is comprised of three sections: Clinical Radiation Oncology, Radiological Physics, and Radiation (and Cancer) Biology. Here we provide an update on changes to the biology portion of the ACR exam. We also discuss the availability and use of the ACR and biology practice exams as assessment and teaching tools for both the instructors of radiation and cancer biology and the residents they teach.


Asunto(s)
Internado y Residencia , Oncología por Radiación/educación , Radiobiología/educación , Biología/educación , Humanos , Oncología Médica/educación
17.
Radiat Res ; 187(6): 732-742, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28437188

RESUMEN

There continues to be a major effort in the United States to develop mitigators for the treatment of mass casualties that received high-intensity acute ionizing radiation exposures from the detonation of an improvised nuclear device during a radiological terrorist attack. The ideal countermeasure should be effective when administered after exposure, and over a wide range of absorbed doses. We have previously shown that the administration of a subcutaneous incision of a defined length, if administered within minutes after irradiation, protected young adult female C57BL/6 mice against radiation-induced lethality, and increased survival after total-body exposure to an LD50/30 X-ray dose from 50% to over 90%. We refer to this approach as "protective wounding". In this article, we report on our efforts to further optimize, characterize and demonstrate the validity of the protective wounding response by comparing the response of female and male mice, varying the radiation dose, the size of the wound, and the timing of wounding with respect to administration of the radiation dose. Both male and female mice that received a subcutaneous incision after irradiation were significantly protected from radiation lethality. We observed that the extent of protection against lethality after an LD50/30 X-ray dose was independent of the size of the subcutaneous cut, and that a 3 mm subcutaneous incision is effective at enhancing the survival of mice exposed to a broad range of radiation doses (LD15-LD100). Over the range of 6.2-6.7 Gy, the increase in survival observed in mice that received an incision was associated with an enhanced recovery of hematopoiesis. The enhanced rate of recovery of hematopoiesis was preceded by an increase in the production of a select group of cytokines. Thus, a thorough knowledge of the timing of the cytokine cascade after wounding could aid in the development of novel pharmacological radiation countermeasures that can be administered several days after the actual radiation exposure.


Asunto(s)
Síndrome de Radiación Aguda/fisiopatología , Síndrome de Radiación Aguda/terapia , Hematopoyesis/efectos de la radiación , Punciones/métodos , Herida Quirúrgica/fisiopatología , Tasa de Supervivencia , Síndrome de Radiación Aguda/etiología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Dosis de Radiación , Resultado del Tratamiento , Irradiación Corporal Total/efectos adversos
18.
Radiat Res ; 165(1): 9-15, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16392957

RESUMEN

Cataractogenesis is a widely reported late effect that is observed in patients receiving total-body irradiation (TBI) prior to bone marrow transplantation or radiotherapy for ocular or head and neck cancers. Recent studies indicate that estrogens may protect against age-related and drug-induced cataracts. Moreover, other reports suggest that estrogen possesses antioxidant properties. Since the effect of estrogen on radiation cataractogenesis is unknown, we wished to determine whether estrogen modulates radiation-induced opacification of the lens. Intact or ovariectomized Sprague-Dawley rats were treated with either 17-beta-estradiol or an empty silastic capsule. The right orbit was then irradiated with either 10 or 15 Gy of (60)Co gamma rays using a Leksell Gamma Knife, and lenses were examined at various times postirradiation with a slit lamp or evaluated for light transmission. We found that for ovariectomized rats irradiated with 15 Gy, the lens opacity and the incidence of cataract formation in the estradiol-treated group were significantly increased compared to the control group at the end of the 25-week period of observation. Cataract incidence was also high in irradiated eyes of ovary-intact animals at 25 weeks postirradiation but was greatly reduced in the ovariectomized control group, with less than half of irradiated eyes showing evidence of cataractogenesis. Thus, after irradiation with 15 Gy of gamma rays, estrogen increased the incidence of cataract formation. We also observed that although the incidence of cataract formation in rats irradiated with 10 Gy and receiving continuous estrogen treatment was not altered compared to rats in the control group that did not receive estrogen, the latent period for posterior subcapsular cataract formation decreased and the severity of the anterior cataract increased. Taken together, our data suggest that estrogen accelerates progression of radiation-induced opacification.


Asunto(s)
Catarata/patología , Catarata/prevención & control , Estrógenos/administración & dosificación , Traumatismos por Radiación/patología , Traumatismos por Radiación/prevención & control , Radiocirugia/efectos adversos , Animales , Catarata/etiología , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Estrógenos/metabolismo , Femenino , Rayos gamma/efectos adversos , Ovariectomía , Traumatismos por Radiación/etiología , Tolerancia a Radiación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
19.
Mutat Res Rev Mutat Res ; 770(Pt B): 238-261, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27919334

RESUMEN

The lens of the eye has long been considered as a radiosensitive tissue, but recent research has suggested that the radiosensitivity is even greater than previously thought. The 2012 recommendation of the International Commission on Radiological Protection (ICRP) to substantially reduce the annual occupational equivalent dose limit for the ocular lens has now been adopted in the European Union and is under consideration around the rest of the world. However, ICRP clearly states that the recommendations are chiefly based on epidemiological evidence because there are a very small number of studies that provide explicit biological, mechanistic evidence at doses <2Gy. This paper aims to present a review of recently published information on the biological and mechanistic aspects of cataracts induced by exposure to ionizing radiation (IR). The data were compiled by assessing the pertinent literature in several distinct areas which contribute to the understanding of IR induced cataracts, information regarding lens biology and general processes of cataractogenesis. Results from cellular and tissue level studies and animal models, and relevant human studies, were examined. The main focus was the biological effects of low linear energy transfer IR, but dosimetry issues and a number of other confounding factors were also considered. The results of this review clearly highlight a number of gaps in current knowledge. Overall, while there have been a number of recent advances in understanding, it remains unknown exactly how IR exposure contributes to opacification. A fuller understanding of how exposure to relatively low doses of IR promotes induction and/or progression of IR-induced cataracts will have important implications for prevention and treatment of this disease, as well as for the field of radiation protection.


Asunto(s)
Catarata/etiología , Radiación Ionizante , Animales , Catarata/patología , Opacidad de la Córnea/etiología , Humanos , Cristalino/metabolismo , Cristalino/fisiología , Cristalino/efectos de la radiación , Modelos Animales
20.
Oncotarget ; 7(15): 20788-800, 2016 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-26959112

RESUMEN

Pro-oxidative stressors can suppress host immunity due to their ability to generate oxidized lipid agonists of the platelet-activating factor-receptor (PAF-R). As radiation therapy also induces reactive oxygen species, the present studies were designed to define whether ionizing radiation could generate PAF-R agonists and if these lipids could subvert host immunity. We demonstrate that radiation exposure of multiple tumor cell lines in-vitro, tumors in-vivo, and human subjects undergoing radiation therapy for skin tumors all generate PAF-R agonists. Structural characterization of radiation-induced PAF-R agonistic activity revealed PAF and multiple oxidized glycerophosphocholines that are produced non-enzymatically. In a murine melanoma tumor model, irradiation of one tumor augmented the growth of the other (non-treated) tumor in a PAF-R-dependent process blocked by a cyclooxygenase-2 inhibitor. These results indicate a novel pathway by which PAF-R agonists produced as a byproduct of radiation therapy could result in tumor treatment failure, and offer important insights into potential therapeutic strategies that could improve the overall antitumor effectiveness of radiation therapy regimens.


Asunto(s)
Antioxidantes/farmacología , Melanoma/terapia , Factor de Activación Plaquetaria/agonistas , Glicoproteínas de Membrana Plaquetaria/agonistas , Receptores Acoplados a Proteínas G/agonistas , Neoplasias Cutáneas/terapia , Rayos Ultravioleta , Animales , Femenino , Humanos , Melanoma/inmunología , Melanoma/metabolismo , Melanoma/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Estrés Oxidativo , Glicoproteínas de Membrana Plaquetaria/fisiología , Receptores Acoplados a Proteínas G/fisiología , Transducción de Señal , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/secundario , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA