Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
In Vivo ; 24(1): 9-19, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20133970

RESUMEN

Antibiotic and antifungal agents used in supportive care regimens for bone marrow transplantation recipients contribute to a significant dose-modifying effect of otherwise lethal total body irradiation. To determine whether drugs used in supportive care and other commonly used antibiotics such as tetracycline function as radiation protectors or damage mitigators in vitro, 13 drugs were tested for radiation protection and radiation damage mitigation of 32D cl 3 hematopoietic progenitor cells in clonagenic survival curves in vitro. Antibiotic/Antifungal agents including cilastatin, amikacin, ceftazidine, vancomycin, tetracycline, doxycycline, ciprofloxacin, metronidazole, methacycline, minocycline, meclocycline, oxytetracycline and rolitetracycline were added in 1, 10, or 100 micromolar concentrations to murine interleukin-3-dependent hematopoietic progenitor cell line 32D cl 3 cells either before or after irradiation of 0 to 8 Gy. Control irradiated 32D cl 3 cells showed radiosensitivity comparable to freshly explanted mouse marrow hematopoietic progenitor cells (D(0) 1.1+/-0.1 Gy, N 1.5+/-0.4). Positive control GS-nitroxide JP4-039 (known radiation mitigator) treated 32D cl 3 cells were radioresistant (D(0) 1.2+/-0.1, N 5.8+/-2.4 (p=0.009)). Of the 13 drugs tested, tetracycline was found to be a significant radiation mitigator (D(0) 0.9+/-0.1, N 13.9+/-0.4 (p=0.0027)). Thus, the radiation dose-modifying effect of some antibiotics, but not those currently used in the supportive care (antibiotic/antifungal regimens) for marrow transplant patients, may act as radiation damage mitigators for hematopoietic cells as well as decreasing the growth and inflammatory response to microbial pathogens.


Asunto(s)
Antibacterianos/farmacología , Antifúngicos/farmacología , Trasplante de Médula Ósea , Células Madre Hematopoyéticas/efectos de la radiación , Traumatismos Experimentales por Radiación/prevención & control , Protectores contra Radiación/farmacología , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/patología , Ratones , Traumatismos Experimentales por Radiación/patología
2.
In Vivo ; 24(1): 1-8, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20133969

RESUMEN

Hypoxic regions limit the radiocontrollability of head and neck carcinomas. Whether or not combinations of plasmid/liposome mediated overexpression of normal tissue protective manganese superoxide dismutase (MnSOD), cetuximab (C225), and the hypoxic cytotoxin tirapazamine (TPZ) enhanced radiotherapeutic effects was tested in a CAL-33 orthotopic mouse cheek tumor model. The tumor volume continued to increase in the control (untreated) mice, with a ninefold increase by 10 days when the tumors exceeded 2 cm(3). The mice receiving 14 Gy only showed reduced tumor growth to 3.1+/-0.1 fold at day 10. The mice receiving MnSOD-PL, C225, TPZ plus 14 Gy had the best outcome with 0.7+/-0.1 fold increase in tumor volume by 10 days (p=0.015) compared to irradiation only. The addition of MnSOD-PL, TPZ, and C225 to irradiation optimized the therapeutic ratio for the local control of hypoxic region-containing CAL-33 orthotopic tumors.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Células Escamosas/terapia , Terapia Genética , Neoplasias de la Boca/terapia , Superóxido Dismutasa/genética , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales Humanizados , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Cetuximab , Relación Dosis-Respuesta en la Radiación , Humanos , Liposomas , Ratones , Ratones Desnudos , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Óxido Nítrico/metabolismo , Fármacos Sensibilizantes a Radiaciones/administración & dosificación , Radioterapia Adyuvante , Tirapazamina , Triazinas/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Radiat Res ; 172(4): 414-22, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19772462

RESUMEN

To assist in screening existing drugs for use as potential radioprotectors, we used a human unbiased 16,560 short interfering RNA (siRNA) library targeting the druggable genome. We performed a synthetic protection screen that was designed to identify genes that, when silenced, protected human glioblastoma T98G cells from gamma-radiation-induced cell death. We identified 116 candidate protective genes, then identified 10 small molecule inhibitors of 13 of these candidate gene products and tested their radioprotective effects. Glyburide, a clinically used second-generation hypoglycemic drug, effectively decreased radiation-induced cell death in several cell lines including T98G, glioblastoma U-87 MG, and normal lung epithelial BEAS-2B and in primary cultures of astrocytes. Glyburide significantly increased the survival of 32D cl3 murine hematopoietic progenitor cells when administrated before irradiation. Glyburide was radioprotective in vivo (90% of C57BL/6NHsd female mice pretreated with 10 mg/kg glyburide survived 9.5 Gy total-body irradiation compared to 42% of irradiated controls, P = 0.0249). These results demonstrate the power of unbiased siRNA synthetic protection screening with a druggable genome library to identify new radioprotectors.


Asunto(s)
Gliburida/farmacología , Hipoglucemiantes/farmacología , ARN Interferente Pequeño , Protectores contra Radiación/farmacología , Animales , Secuencia de Bases , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Ensayo de Unidades Formadoras de Colonias , Evaluación Preclínica de Medicamentos , Femenino , Biblioteca Genómica , Hematopoyesis/efectos de los fármacos , Hematopoyesis/efectos de la radiación , Humanos , Ratones , Fenotipo , ARN Interferente Pequeño/genética , Radiación Ionizante , Irradiación Corporal Total
4.
Radiat Res ; 171(5): 588-95, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19580494

RESUMEN

To determine whether increased mitochondrially localized catalase was radioprotective, a human catalase transgene was cloned into a small pSVZeo plasmid and localized to the mitochondria of 32D cl 3 cells by adding the mitochondrial localization sequence of MnSOD (mt-catalase). The cell lines 32D-Cat and 32D-mt-Cat had increased catalase biochemical activity as confirmed by Western blot analysis compared to the 32D cl 3 parent cells. The MnSOD-overexpressing 32D cl 3 cell line, 2C6, had decreased baseline catalase activity that was increased in 2C6-Cat and 2C6-mt-Cat subclonal cell lines. 32D-mt-Cat cells were more radioresistant than 32D-Cat cells, but both were radioresistant relative to 32D cl 3 cells. 2C6-mt-Cat cells but not 2C6-Cat cells were radioresistant compared to 2C6 cells. Intratracheal injection of the mt-catalase-plasmid liposome complex (mt-Cat-PL) but not the catalase-plasmid liposome complex (Cat-PL) increased the resistance of C57BL/6NHsd female mice to 20 Gy thoracic irradiation compared to MnSOD-plasmid liposomes. Thus mitochondrially targeted overexpression of the catalase transgene is radioprotective in vitro and in vivo.


Asunto(s)
Catalasa/fisiología , Mitocondrias/enzimología , Tolerancia a Radiación , Animales , Catalasa/genética , Línea Celular , Supervivencia Celular/efectos de la radiación , Femenino , Glutatión/análisis , Glutatión Peroxidasa/análisis , Humanos , Liposomas , Ratones , Ratones Endogámicos C57BL , Plásmidos , Superóxido Dismutasa/fisiología , Transgenes
5.
In Vivo ; 23(5): 669-77, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19779099

RESUMEN

Fluorescent yellow direct repeat (FYDR) mice carry a transgenic reporter for homologous recombination (HR) and have been used to reveal an age-dependent increase in HR in the pancreas. An established in vitro model system for accelerated aging of the marrow is the mouse long-term bone marrow culture (LTBMC) system. To determine whether the FYDR system, in which an HR event can lead to a fluorescent cell, can be used to study the effects of aging in LTBMCs, clonally expanded hematopoietic and marrow stromal cells in FYDR, positive control FYDR-Recombined (FYDR-Rec), and negative control wild-type C57BL/6NHsd (WT) LTBMCs were analysed. All groups of cultures demonstrated equivalent parameters of continuous hematopoiesis including generation of multilineage colony forming CFU-GM progenitor cells for over 22 weeks and age associated senescence of hematopoiesis. Results indicate that low expression of the FYDR transgene in bone marrow cells in vivo and in vitro prevents the use of the FYDR mice to study rare combination events in bone marrow. Using an alternative approach for detecting HR, namely the sister chromatid exchange (SCE) assay, a statistically significant increase in the number of SCEs per chromosome was observed in adherent cells subcultured from 20-week-compared to 4-week-old LTBMCs. These data suggest that adherent marrow stromal cells from LTBMCs become increasingly susceptible to HR events during aging.


Asunto(s)
Células de la Médula Ósea/citología , Senescencia Celular/genética , Hematopoyesis/genética , Recombinación Genética , Animales , Células de la Médula Ósea/fisiología , Células Cultivadas , Células Clonales , Femenino , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Ratones , Intercambio de Cromátides Hermanas , Células del Estroma/citología , Células del Estroma/fisiología
6.
In Vivo ; 23(5): 717-26, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19779106

RESUMEN

It was unknown if a mitochondria-targeted nitroxide (JP4-039) could augment potentially lethal damage repair (PLDR) of cells in quiescence. We evaluated 32D cl 3 murine hematopoietic progenitor cells which were irradiated and then either centrifuged to pellets (to simulate PLDR conditions) or left in exponential growth for 0, 24, 48 or 72 h. Pelleted cells demonstrated cell cycle arrest with a greater percentage in the G(1)-phase than did exponentially growing cells. Irradiation survival curves demonstrated a significant radiation damage mitigation effect of JP4-039 over untreated cells in cells pelleted for 24 h. No significant radiation mitigation was detected if drugs were added 48 or 72 h after irradiation. Electron paramagnetic resonance spectroscopy demonstrated a greater concentration of JP4-039 in mitochondria of 24 h-pelleted cells than in exponentially growing cells. These results establish a potential role of mitochondria-targeted nitroxide drugs as mitigators of radiation damage to quiescent cells including stem cells.


Asunto(s)
Antioxidantes/farmacología , Células Madre Hematopoyéticas/patología , Células Madre Hematopoyéticas/efectos de la radiación , Óxidos de Nitrógeno/farmacología , Traumatismos Experimentales por Radiación/patología , Traumatismos Experimentales por Radiación/prevención & control , Protectores contra Radiación/farmacología , Animales , Ciclo Celular/efectos de los fármacos , Ciclo Celular/efectos de la radiación , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Ratones , Ratones Endogámicos C3H , Mitocondrias/química , Mitocondrias/efectos de los fármacos , Mitocondrias/efectos de la radiación , Óxidos de Nitrógeno/análisis
7.
In Vivo ; 33(2): 303-311, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30804107

RESUMEN

AIM: To demonstrate that Fanconi anemia complementation group D2-deficient (Fancd2-/-) hematopoietic progenitor cell lines can be transformed by transfection with a plasmid containing either the E6 or E7 oncogene of human papillomavirus (HPV) to generate malignant plasmacytoma-inducing cell lines. MATERIALS AND METHODS: In order to determine whether a single HPV type 16 (HPV16) oncogene induced malignant transformation, Fancd2-/- and Fancd2+/+ interleukin 3 (IL3)-dependent hematopoietic progenitor cell lines were transfected with plasmids containing E6 or E7 oncogene, or control empty plasmid. RESULTS: Fancd2-/- but not Fancd2+/+ cells were transformed into malignant IL3-independent cells by both E6, and E7 oncogenes, but not by empty plasmid. Hematopoietic cell lines and tumors induced by Fancd2-/- E6 and Fancd2-/- E7 cell lines were positive for each respective HPV RNA and protein. CONCLUSION: A single HPV16 oncogene is adequate to produce malignant transformation of Fancd2-/- hematopoietic cells.


Asunto(s)
Transformación Celular Neoplásica/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Células Madre Hematopoyéticas/virología , Interleucina-3/genética , Línea Celular Tumoral , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/patogenicidad , Humanos , Proteínas Oncogénicas Virales/genética , Oncogenes/genética , Proteínas E7 de Papillomavirus/genética , Plásmidos/genética , Proteínas Represoras/genética , Transfección/métodos
8.
Radiat Res ; 191(2): 139-153, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30499383

RESUMEN

We quantitated age-related accumulation of senescent cells in irradiated Fanconi anemia (FA) (Fanca-/- mouse cell lines in vitro, and monitored the effect of continuous administration (via drinking water) of the water-soluble radiation mitigator, MMS350, on tissues in vivo over one year after 7.5 Gy total-body irradiation (TBI). Irradiated Fanca-/- mouse bone marrow stromal cell lines showed increased numbers of beta-galactosidase- and p21-positive senescent cells compared to Fanca+/+ cell lines, which was reduced by MMS350. One week after 7.5 Gy TBI, Fanca-/- mice showed increased numbers of senescent cells in spleen compared to Fanca+/+ controls, decreased bone marrow cellularity, failure of explanted bone marrow to proliferate in vitro to form a hematopoietic microenvironment and no detectable single stromal cell cloning capacity. There was no detectable amelioration by MMS350 administration at one week. In contrast, one year post-TBI, Fanca-/- mice demonstrated fewer senescent cells in brain and spleen compared to Fanca+/+ controls. While Fanca-/- mouse bone marrow stromal cells explanted one year post-TBI still failed to proliferate in vitro, continuous oral administration of 400 µ M, MMS350 in drinking water restored explanted stromal cell proliferation. The data indicate that continuous administration of MMS350 modulated several properties of TBI-accelerated aging in Fanca-/- mice as well as control mice, and support further study of MMS350 as a modulator of radiation late effects.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Éteres Cíclicos/administración & dosificación , Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Anemia de Fanconi/patología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de la radiación , Protectores contra Radiación/administración & dosificación , Sulfóxidos/administración & dosificación , Irradiación Corporal Total , Administración Oral , Animales , Éteres Cíclicos/farmacología , Femenino , Masculino , Células Madre Mesenquimatosas/citología , Ratones , Ratones Noqueados , Protectores contra Radiación/farmacología , Sulfóxidos/farmacología , Microambiente Tumoral
9.
In Vivo ; 33(3): 675-688, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31028184

RESUMEN

Background/Aim: The cause of fatal neuromuscular amyotrophic lateral sclerosis (ALS) is not known. Materials and Methods: Ninety-day-old superoxide-dismutase-1 G93A (SOD1 G93A ) mice demonstrating level 1 paralysis, received 9.0 Gy total body irradiation (TBI) from a cesium source at 340 cGy per minute, and intravenous transplantation with 1×10 6 C57BL/6 green fluorescent protein (GFP)+ donor bone marrow cells. Results: Paralysis-free survival was prolonged in TBI and bone marrow-transplanted SOD1 G93A mice from 100 to over 250 days (p=0.0018). Other mice transplanted with SOD1 G93A marrow or marrow treated with the free-radical scavenger MMS350 showed no therapeutic effect. GFP+ macrophage-2 (M2) microglial cells of bone marrow origin, were seen at sites of degenerating anterior horn motor neurons. SOD1 G93A mice had a disruption in the blood-brain barrier permeability which was reversed by marrow transplant from C57BL/6 mice. SOD1 G93A marrow showed unexpected robust hematopoiesis in vitro, and radioresistance. Conclusion: After TBI, M2 microglial cells from transplanted donor marrow extended the paralysis-free interval in SOD1 G93A mice.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Microglía/metabolismo , Mutación , Superóxido Dismutasa-1/genética , Sustitución de Aminoácidos , Esclerosis Amiotrófica Lateral/etiología , Esclerosis Amiotrófica Lateral/fisiopatología , Esclerosis Amiotrófica Lateral/terapia , Animales , Barrera Hematoencefálica/metabolismo , Trasplante de Médula Ósea , Diferenciación Celular/genética , Modelos Animales de Enfermedad , Expresión Génica , Supervivencia de Injerto , Hematopoyesis/genética , Ratones , Ratones Transgénicos , Microglía/inmunología , Tolerancia a Radiación/genética , Superóxido Dismutasa-1/metabolismo , Quimera por Trasplante
10.
In Vivo ; 33(6): 1757-1766, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31662500

RESUMEN

BACKGROUND/AIM: We tested JP4-039, a GS-nitroxide radiation damage mitigator in proton therapy of Fanconi anemia (FA) mice. MATERIALS AND METHODS: Fanca-/- and Fanca+/+ bone marrow stromal cells were pre-treated with JP4-039 and irradiated with either protons or photons (0-10 GyRBE) followed by clonogenic survival and ß-Galactosidase senescence analysis. Fanca-/- and Fanca+/+ mice were pretreated with JP4-039 for 10 min prior to oropharyngeal irradiation with either protons or photons (0 or 30 GyRBE) followed by sacrifice and measurement of oral cavity ulceration, distant hematopoietic suppression, and real-time polymerase chain reaction analysis. RESULTS: JP4-039 reduced oral cavity ulceration in Fanca-/- mice, transcripts Nfkb, Ap1, Sp1, and Nrf2, and proton therapy induced distant marrow suppression. CONCLUSION: JP4-039 protected Fanca-/- and Fanca+/+ cells and mouse oral cavity from both proton and photon radiation.


Asunto(s)
Anemia de Fanconi/radioterapia , Mucositis/tratamiento farmacológico , Óxidos de Nitrógeno/farmacología , Terapia de Protones/efectos adversos , Protectores contra Radiación/farmacología , Animales , Línea Celular , Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación A de la Anemia de Fanconi/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/efectos de la radiación , Ratones , Mucositis/metabolismo , Tolerancia a Radiación/efectos de los fármacos
11.
Radiat Res ; 170(4): 437-43, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19024650

RESUMEN

To determine whether systemic administration of MnSOD-PL protected mice from the acute hematopoietic syndrome and delayed death after total-body irradiation (TBI), C57BL/ 6J mice were injected intravenously with 100 microl liposomes containing 100 microg of human MnSOD-transgene plasmid 24 h prior to irradiation with 9.5 Gy or 1.0 Gy. The dose of 9.5 Gy was lethal to 42% of irradiated control female mice and 74% of irradiated control male mice at 30 days, with bone marrow hypocellularity consistent with the hematopoietic syndrome. A statistically significant increase in survival was observed in MnSOD-PL-treated female mice out to 400 days and in male mice out to 340 days. The incidence of tumors was similar between surviving groups. Between 350 and 600 days, the outcome was similar for both MnSOD-PL-treated and control irradiated groups, consistent with aging, with no difference in gross or microscopic pathological evidence of tumors. Male and female mice receiving 1.0 Gy TBI showed radiation-induced life shortening after 120 days that was decreased by MnSOD-PL administration and that was not associated with an increase in rate of tumor-associated death. Therefore, systemic MnSOD-PL radioprotective gene therapy is not associated with a detectably higher incidence of late carcinogenesis.


Asunto(s)
Enfermedades Hematológicas/prevención & control , Traumatismos Experimentales por Radiación/prevención & control , Superóxido Dismutasa/genética , Enfermedad Aguda , Animales , Femenino , Terapia Genética , Enfermedades Hematológicas/genética , Enfermedades Hematológicas/mortalidad , Hematopoyesis/efectos de la radiación , Humanos , Liposomas , Masculino , Ratones , Ratones Endogámicos C57BL , Plásmidos , Traumatismos Experimentales por Radiación/genética , Traumatismos Experimentales por Radiación/mortalidad , Síndrome , Irradiación Corporal Total
12.
Exp Hematol ; 35(1): 137-45, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17198882

RESUMEN

OBJECTIVE: Neuronal nitric oxide synthase (NOS1, mitochondrial NOS, neuronal NOS) homozygous deletion recombinant negative mice demonstrate ionizing irradiation resistance in vivo, attributable to the decrease in mitochondrial-localized production of peroxynitrite, a potent lipid toxic free radical species resulting from the combination of nitric oxide and superoxide. The present studies were designed to determine whether reduced mitochondrial generation of toxic radical oxygen species in NOS1-/- mice also increased the longevity of hematopoiesis in continuous bone marrow cultures and conferred radioresistance to cells in vitro. MATERIALS AND METHODS: Long-term bone marrow cultures (LTBMCs) were established from NOS1-/- and NOS1+/+ littermate mice. Radiation resistance of hematopoietic and marrow stromal cells was measured. Cell cycle analysis and measurement of glutathione and glutathione peroxidase were carried out on irradiated clonal bone marrow stromal cell lines. RESULTS: A significant increase in longevity of hematopoiesis was detected in NOS1-/- mouse LTBMCs for over 64 weeks in culture compared to 20 weeks for NOS1+/+ mouse LTBMCs (p < 0.001). Permanent bone marrow stromal cell lines derived from NOS1-/- mouse LTBMCs demonstrated increased radioresistance in vitro reflected by an increased shoulder on the survival curve with n = 32.15 +/- 1.21 compared to NOS1+/+ cells n = 10.47 +/- 3.2 (p = 0.0026), interleukin-3-dependent NOS1-/- hematopoietic progenitor cell lines also demonstrated decreased apoptosis after 10 Gy irradiation. Both pre- and postirradiation stabilization of the cellular antioxidant pool was detected in NOS1-/- cells. NOS1-/- cells showed a prolonged G1 cell cycle arrest after 10 Gy. CONCLUSIONS: Prolonged hematopoiesis in LTBMCs correlates with intrinsic radioresistance of hematopoietic and marrow stromal cells from NOS1-/- mice. The data confirm the importance to hematopoiesis of mitochondrial localized nitric oxide in both radioresistance and longevity of hematopoiesis in continuous bone marrow cultures.


Asunto(s)
Células de la Médula Ósea/citología , Hematopoyesis , Células Madre Hematopoyéticas/efectos de la radiación , Óxido Nítrico Sintasa de Tipo I/genética , Células del Estroma/efectos de la radiación , Animales , Células de la Médula Ósea/enzimología , Técnicas de Cultivo de Célula , Células Cultivadas , Homocigoto , Ratones , Ratones Mutantes , Mitocondrias/metabolismo , Mutación , Óxido Nítrico/metabolismo , Óxido Nítrico/fisiología , Efectos de la Radiación , Factores de Tiempo
13.
In Vivo ; 32(5): 1009-1023, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30150422

RESUMEN

BACKGROUND/AIM: The mitochondrial targeted GS-nitroxide, JP4-039, is an effective total body irradiation (TBI) mitigator when delivered intravenously (IV) up to 72 h after exposure. Effective systemic and localized administration to oral cavity/oropharynx and esophagus has been demonstrated. The objective of the study was to establish alternatives to IV administration suitable for JP4-039 delivery to mass casualties. MATERIALS AND METHODS: JP4-039 was administered to C57BL/6 mice by topically applied carboxy-methyl-cellulose microneedle arrays (MNAs) or by intramuscular (IM) injection. Three different formulations that have passed Food and Drug Administration review, namely Captisol, 2-hydroxypropyl-ß-cyclodextrin (cyclodextrin), and Miglyol-812-N, were used for drug delivery. Intraoral (IO) administration with each formulation was also evaluated. RESULTS: All tested formulations and MNAs successfully delivered JP4-039. However, IM delivery of the Miglyol-812-N displayed very efficient and highly reproducible radiation mitigation. CONCLUSION: Effective IM delivery of JP4-039 in animal models after TBI or partial-body irradiation suggested the use of the Miglyol-812-N formulation in both medical indications and radiation countermeasures.


Asunto(s)
Vías de Administración de Medicamentos , Composición de Medicamentos , Óxidos de Nitrógeno/administración & dosificación , Óxidos de Nitrógeno/química , Protectores contra Radiación/administración & dosificación , Protectores contra Radiación/química , Administración Intravenosa , Administración Oral , Animales , Apoptosis/efectos de los fármacos , Daño del ADN/efectos de la radiación , Reparación del ADN/efectos de los fármacos , Estabilidad de Medicamentos , Femenino , Inyecciones Intramusculares , Ratones , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Mitocondrias/metabolismo , Estructura Molecular , Óxidos de Nitrógeno/farmacocinética , Traumatismos Experimentales por Radiación , Radiación Ionizante , Protectores contra Radiación/farmacocinética , Reproducibilidad de los Resultados , Tasa de Supervivencia , Irradiación Corporal Total
14.
Radiat Res ; 168(5): 560-6, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17973554

RESUMEN

We determined whether manganese superoxide dismutase (MnSOD)-plasmid liposome (PL) transfection of C57BL/ 6NHsd mouse bone marrow protected cells irradiated at room temperature (24 degrees C) or in the cryopreserved state. MnSOD-overexpressing hematopoietic progenitor 2C6 cells were radioresistant compared to the parent 32D cl 3 cells when irradiated frozen or at 24 degrees C. Fresh whole marrow from mice injected intravenously with MnSOD-PL prior to explant as well as explanted marrow single cell suspensions transfected in vitro were irradiated at 24 degrees C or -80 degrees C. In vivo or in vitro transfection of marrow with MnSOD-PL produced significant radiation protection of irradiated marrow progenitor cells compared to controls at 24 degrees C or -80 degrees C. (in vivo transfection D(0) 2.19 +/- 0.21 at 24 degrees C, D(0) 2.10 +/- 0.07 at -80 degrees C compared to control D(0) 1.56 +/- 0.06 or 1.66 +/- 0.04, P = 0.047 and 0.017 respectively; in vitro transfection D(0) 2.35 +/- 0.11 at 24 degrees C, D(0) 3.42 +/- 0.13 at -80 degrees C compared to D(0) 1.81 +/- 0.01 or 2.53 +/- 0.05, P = 0.0087 and 0.0026, respectively). Thus the MnSOD transgene product protects frozen marrow cells as well as marrow cells irradiated at 24 degrees C.


Asunto(s)
Células de la Médula Ósea/fisiología , Células de la Médula Ósea/efectos de la radiación , Criopreservación/métodos , Células Madre Hematopoyéticas/fisiología , Células Madre Hematopoyéticas/efectos de la radiación , Superóxido Dismutasa/metabolismo , Animales , Células de la Médula Ósea/citología , Células Cultivadas , Citoprotección/fisiología , Citoprotección/efectos de la radiación , Femenino , Células Madre Hematopoyéticas/citología , Ratones , Ratones Endogámicos C57BL , Radiación Ionizante , Superóxido Dismutasa/genética , Transfección/métodos , Transgenes/fisiología , Regulación hacia Arriba/fisiología
15.
Radiat Res ; 168(5): 552-9, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17973549

RESUMEN

Ethyl pyruvate (EP), a simple aliphatic ester of pyruvic acid, has been shown to improve survival and ameliorate organ damage in animal models of sepsis, ischemia/reperfusion injury and hemorrhagic shock. Incubating IL3-dependent mouse hematopoietic progenitor cell 32Dcl3 cells before or after irradiation with 10 mM EP increased resistance to radiation as assessed by clonogenic radiation survival curves, decreased release of mitochondrial cytochrome C into the cytoplasm, and decreased apoptosis. EP inhibited radiation-induced caspase 3 activation and poly(ADP-ribose) polymerase (PARP) cleavage in 32Dcl3 cells in a concentration-dependent fashion. EP was given i.p. to C57BL/6NHsd mice irradiated with 9.75 Gy total-body irradiation (TBI). This treatment significantly improved survival. The survival benefit was apparent irrespective of whether treatment with EP was started 1 h before TBI and continued for 5 consecutive days after TBI or the compound was injected only 1 h before or only for 5 days after TBI. In all of the in vitro and in vivo experiments, ethyl lactate, an inactive analogue of EP, had no detectable radioprotective or mitigating effects. EP may be an effective radioprotector and mitigator of the hematopoietic syndrome induced by TBI.


Asunto(s)
Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de la radiación , Piruvatos/administración & dosificación , Protectores contra Radiación/administración & dosificación , Tasa de Supervivencia , Irradiación Corporal Total , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Femenino , Células Madre Hematopoyéticas/citología , Ratones , Ratones Endogámicos C57BL , Análisis de Supervivencia
16.
In Vivo ; 31(3): 313-320, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28438857

RESUMEN

BACKGROUND/AIM: The polycythemia form of Friend leukemia virus (FVP) causes splenomegaly and lethal erythroleukemia in Fv-2s-susceptible mouse strains. We sought to determine whether the hematopoietic stem cell (HSC) pool was expanded in Fv-2r-resistant mice. MATERIALS AND METHODS: The 120-day bone marrow transplantation competitive repopulation assay was used to determine whether FVP-infected Fv-2r C57BL/6 mice demonstrated expansion of the HSC pool compared to the pool of committed hematopoietic progenitor cells in the same marrow assayed in vitro. RESULTS: There was a significant expansion of committed hematopoietic progenitors observed in virus-infected Fv-2s FVB mice, but not Fv-2r C57BL/6 mice. Furthermore, Fv-2r mice showed no detectable expansion of either committed hematopoietic progenitor cells or the multipotential stem cell pool by competitive repopulation assay. CONCLUSION: Friend virus disease in Fv-2s mice is associated with expansion of committed hematopoietic progenitors. Fv-2r mice show no expansion of either committed progenitor or pluripotential stem cell numbers.


Asunto(s)
Médula Ósea/patología , Células Madre Hematopoyéticas/patología , Infecciones por Retroviridae/patología , Infecciones Tumorales por Virus/patología , Animales , Médula Ósea/virología , Femenino , Virus de la Leucemia Murina de Friend/patogenicidad , Células Madre Hematopoyéticas/virología , Leucemia Eritroblástica Aguda/patología , Leucemia Eritroblástica Aguda/virología , Leucemia Experimental/patología , Leucemia Experimental/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Infecciones por Retroviridae/virología , Bazo/patología , Bazo/virología , Esplenomegalia/patología , Esplenomegalia/virología , Infecciones Tumorales por Virus/virología
17.
In Vivo ; 31(2): 159-168, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28358695

RESUMEN

BACKGROUND/AIM: Total-body irradiation and/or administration of chemotherapy drugs in bone marrow transplantation induce cytokines that can suppress engraftment. Fanconi Anemia (FA) patients have a hyperactive responsiveness to the inhibitory cytokine, transforming growth factor-beta (TGF-ß). Small molecule radiation mitigator drugs, JP4-039 and MMS350, were evaluated for suppression of irradiation or drug-induced TGF-ß. MATERIALS AND METHODS: In vivo induction of TGF-ß by total-body ionizing irradiation (TBI), L-phenylalanine mustard (L-PAM), busulfan or fludarabine, was quantified. In parallel, mitigator drug amelioration of TGF-ß induction in FA D2-/- (FANCD2-/-) mouse bone marrow, was studied in vitro. Tissue culture medium, cell lysates, and mouse plasma were analyzed for TGF-ß levels. RESULTS: Induction of TGF-ß levels in FANCD2-/- and FANCD2+/+ mice and in mouse bone marrow were modulated by both JP4-039 and MMS350. CONCLUSION: Bone marrow transplantation in FA recipients may benefit from administration of small molecule agents that suppress TGF-ß induction.


Asunto(s)
Médula Ósea/efectos de los fármacos , Éteres Cíclicos/farmacología , Anemia de Fanconi/tratamiento farmacológico , Anemia de Fanconi/radioterapia , Óxidos de Nitrógeno/farmacología , Sulfóxidos/farmacología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Western Blotting , Médula Ósea/metabolismo , Busulfano/farmacología , Línea Celular , Células Cultivadas , Quimioterapia/métodos , Anemia de Fanconi/metabolismo , Melfalán/farmacología , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Agonistas Mieloablativos/farmacología , Protectores contra Radiación/farmacología , Técnicas de Cultivo de Tejidos , Factor de Crecimiento Transformador beta/sangre , Factor de Crecimiento Transformador beta/genética , Vidarabina/análogos & derivados , Vidarabina/farmacología , Irradiación Corporal Total/métodos
18.
Radiat Res ; 165(6): 671-7, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16802867

RESUMEN

Smad3 protein is a prominent member of the Tgfb receptor signaling pathway. Smad3(-/-) mice display decreased radiation-induced skin fibrosis, suggesting a defect in both Tgfb-mediated fibroblast proliferation and migration. We established bone marrow stromal cell lines from Smad3(-/-) mice and homozygous littermate(+/+) mice. Smad3(-/-) cells displayed a significant increase in radiation resistance with a D(0)=2.25+/- 0.14 Gy compared to Smad3(+/+) cells with a D(0)=1.75+/- 0.03 (P=0.023). Radioresistance was abrogated by reinsertion of the human SMAD3 transgene, resulting in a D(0)=1.49 0.10 (P=0.028) for Smad3(-/-)(3) cells. More Smad3(-/-) cells than Smad3(+/+) cells were in the G(2)/M phase; Smad3(-/-)(3) cells were similar to Smad3(+/+) cells. Smad3(+/+) cells exhibited increased apoptosis 24 h after 5 Gy (15%) or 8 Gy (43%) compared to less than 1% in Smad3(-/-) cells exposed to either dose. The movement of Smad3(-/-) cells, measured in an automated cell tracking system, was slower than that of Smad3(+/+) cells. Smad3(-/-)(3) cells resembled Smad3(+/+) cells. These studies establish concordance of a defective Tgfb signal transduction pathway, an increased proportion of G(2)/M cells, and radioresistance. The decreased migratory capacity of Smad3(-/-) cells in vitro correlates with decreased radiation fibrosis in vivo in mice deficient in Tgfb signaling.


Asunto(s)
Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteína smad3/metabolismo , Animales , Apoptosis/efectos de la radiación , Células de la Médula Ósea/efectos de la radiación , Línea Celular , Movimiento Celular/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Genes cdc/fisiología , Genes cdc/efectos de la radiación , Ratones , Tolerancia a Radiación/fisiología , Células del Estroma/citología , Células del Estroma/fisiología , Células del Estroma/efectos de la radiación
19.
In Vivo ; 20(6B): 791-6, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17203769

RESUMEN

Radiation therapy of tumors of the head and neck region is compromised by dose limiting toxicity of normal tissues including the oral cavity and oropharyngeal mucosa. MnSOD-Plasmid Liposome (MnSOD-PL) intraoral gene therapy has been demonstrated to decrease normal tissue toxicity and also improve survival in mice with orthotopic SCC-VII squamous cell tumors on the floor of the mouth. Furthermore, intravenous administration of MnSOD-PL in mice with orthotopic tumors, or addition of MnSOD-PL to tumor cell lines in vitro produces a radiosensitizing effect attributable to differences in antioxidant pool responses of tumor cells compared to normal tissues following irradiation. To determine whether EGF receptor (EGFR) antagonists Iressa, or Cetuximab provided further improvement of radiation killing of squamous cell tumors, MnSOD-PL transfected or control SCCVII tumor cells were irradiated in vitro, and then the effect of EGFR receptor antagonists was tested. Cells transfected with MnSOD-PL were relatively radiosensitive D0 = 1.244 +/- 0.126 Gy compared to control D0 = 3.246 +/- 0.087 (p < 0.0001). Clonogenic radiation survival curves of SCCVII cells demonstrated radiosensitization by Iressa D0 = 2.770 +/- 0.134 Gy (p = 0.0264), but no significant radiosensitizing effect of Cetuximab D0 = 3.193 +/- 0.309 (p = 0.7338). The combination of MnSOD-PL plus Iressa further increased radiosensitivity of SCC-VII cells in vitro D0 = 0.785 +/- 0.01064 (p < 0.0001). The results suggest some synergy of the effectiveness of the EGFR antagonist Iressa on increasing the radiation killing of SCC-VII cells that supplements MnSOD-PL tumor radiosensitization.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Receptores ErbB/antagonistas & inhibidores , Quinazolinas/farmacología , Superóxido Dismutasa/genética , Animales , Anticuerpos Monoclonales Humanizados , Antineoplásicos/farmacología , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Supervivencia Celular/efectos de la radiación , Cetuximab , Relación Dosis-Respuesta en la Radiación , Gefitinib , Liposomas , Ratones , Transfección , Transgenes/genética
20.
In Vivo ; 20(3): 333-9, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16724666

RESUMEN

BACKGROUND: Thalidomide (TL), due to its antiangiogenic effects, has been postulated to be a potential radiosensitizer of multiple myeloma and squamous tumors in vivo. MATERIALS AND METHODS: To determine whether TL was a radiosensitizer, 32D cl 3 cells (hematopoietic progenitor) as well as SCC-VII (squamous cell carcinoma), OPM1 or OPM2 (multiple myeloma) tumor cells were irradiated to doses ranging from 0 to 8 Gy and then plated in 0, 50 or 150 microM TL in each of three protocols: i) 1 hour before irradiation; ii) 1 hour before irradiation and also in medium following irradiation; or iii) placed in TL containing medium following irradiation. RESULTS: Using 150 microM TL (which did not stimulate cell growth) the 32D cl 3 cells had increased radiation sensitivity compared to the control irradiated cells. In contrast, the SCC-VII, OPMI or OPM2 cells showed no detectable radiosensitization when incubated in TL before, during or after irradiation compared to the control irradiated cells. CONCLUSION: These results demonstrated that TL may be a selective radiosensitizer.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Células Madre Hematopoyéticas/efectos de la radiación , Mieloma Múltiple/metabolismo , Fármacos Sensibilizantes a Radiaciones/farmacología , Talidomida/farmacología , Animales , Carcinoma de Células Escamosas/patología , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Supervivencia Celular/efectos de la radiación , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Células Madre Hematopoyéticas/efectos de los fármacos , Ratones , Mieloma Múltiple/patología , Tolerancia a Radiación , Radiación Ionizante
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA