Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
BMC Cancer ; 13: 106, 2013 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-23496909

RESUMEN

BACKGROUND: Glioblastoma is one of the intractable cancers and is highly resistant to ionizing radiation. This radioresistance is partly due to the presence of a hypoxic region which is widely found in advanced malignant gliomas. In the present study, we evaluated the effectiveness of the hypoxic cell sensitizer doranidazole (PR-350) using the C6 rat glioblastoma model, focusing on the status of blood brain barrier (BBB). METHODS: Reproductive cell death in the rat C6 glioma cell line was determined by means of clonogenic assay. An intracranial C6 glioma model was established for the in vivo experiments. To investigate the status of the BBB in C6 glioma bearing brain, we performed the Evans blue extravasation test. Autoradiography with [(14)C]-doranidazole was performed to examine the distribution of doranidazole in the glioma tumor. T2-weighted MRI was employed to examine the effects of X-irradiation and/or doranidazole on tumor growth. RESULTS: Doranidazole significantly enhanced radiation-induced reproductive cell death in vitro under hypoxia, but not under normoxia. The BBB in C6-bearing brain was completely disrupted and [(14)C]-doranidazole specifically penetrated the tumor regions. Combined treatment with X-irradiation and doranidazole significantly inhibited the growth of C6 gliomas. CONCLUSIONS: Our results revealed that BBB disruption in glioma enables BBB-impermeable radiosensitizers to penetrate and distribute in the target region. This study is the first to propose that in malignant glioma the administration of hydrophilic hypoxic radiosensitizers could be a potent strategy for improving the clinical outcome of radiotherapy without side effects.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Neoplasias Encefálicas/radioterapia , Glioblastoma/radioterapia , Imidazoles/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Animales , Neoplasias Encefálicas/patología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Glioblastoma/patología , Estudios Prospectivos , Ratas
2.
Gut ; 59(2): 258-66, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19919948

RESUMEN

BACKGROUND: The pathogenesis of non-alcoholic fatty liver disease (NAFLD) is incompletely understood. Kupffer cells (KCs), phagocytic liver-resident macrophages, provide a protective barrier against egress of endotoxin from the portal to the systemic circulation. It is not known if KC phagocytic function is impaired in NAFLD. Super-paramagnetic iron oxide (SPIO) magnetic resonance imaging is a comparative technology dependent on KC phagocytic function. OBJECTIVE: To evaluate KC uptake function, in patients and experimental animals with NAFLD, using SPIO. METHODS: Abdominal CT and histological examination of liver biopsy specimens were used to estimate the degree of steatosis in patients with NAFLD and controls with chronic hepatitis C. SPIO-MRI was then performed in all patients. Normal rats fed a methionine-choline-deficient diet to induce non-alcoholic steatohepatitis (NASH), the more severe stage of NAFLD, and obese, insulin resistant, Zucker fa/fa rats with steatohepatitis, were also studied with SPIO-MRI and analysed for hepatic uptake of fluorescent microbeads. Immunohistochemical analysis evaluated the numbers of KCs in patients and rat livers. RESULTS: Relative signal enhancement (RSE), inversely proportional to KC function, was higher in patients with NAFLD than in controls and with the degree of steatosis on CT. RSE also positively correlated with the degree of steatosis on histology and was similarly higher in rats with induced severe NAFLD (NASH). On immunohistochemistry, defective phagocytic function was the result of reduced phagocytic uptake and not due to reduced KC numbers in rats or patients with NAFLD. CONCLUSIONS: KC uptake function is significantly impaired in patients with NAFLD and experimental animals with NASH, worsens with the degree of steatosis and is not due to a reduction of KC numbers.


Asunto(s)
Hígado Graso/patología , Macrófagos del Hígado/fisiología , Fagocitosis/fisiología , Animales , Recuento de Células , Medios de Contraste , Dextranos , Modelos Animales de Enfermedad , Hígado Graso/fisiopatología , Femenino , Óxido Ferrosoférrico , Humanos , Circulación Hepática/fisiología , Imagen por Resonancia Magnética/métodos , Nanopartículas de Magnetita , Masculino , Microcirculación/fisiología , Ratas , Ratas Wistar , Ratas Zucker , Índice de Severidad de la Enfermedad
3.
Biochem Biophys Res Commun ; 394(3): 522-8, 2010 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-20206606

RESUMEN

To explore Cu(II) ion coordination by His(186) in the C-terminal domain of full-length prion protein (moPrP), we utilized the magnetic dipolar interaction between a paramagnetic metal, Cu(II) ion, and a spin probe introduced in the neighborhood of the postulated binding site by the spin labeling technique (SDSL technique). Six moPrP mutants, moPrP(D143C), moPrP(Y148C), moPrP(E151C), moPrP(Y156C), moPrP(T189C), and moPrP(Y156C,H186A), were reacted with a methane thiosulfonate spin probe and a nitroxide residue (R1) was created in the binding site of each one. Line broadening of the ESR spectra was induced in the presence of Cu(II) ions in moPrP(Y148R1), moPrP(Y151R1), moPrP(Y156R1), and moPrP(T189R1) but not moPrP(D143R1). This line broadening indicated the presence of electron-electron dipolar interaction between Cu(II) and the nitroxide spin probe, suggesting that each interspin distance was within 20 A. The interspin distance ranges between Cu(II) and the spin probes of moPrP(Y148R1), moPrP(Y151R1), moPrP(Y156R1), and moPrP(T189R1) were estimated to be 12.1 A, 18.1 A, 10.7 A, and 8.4 A, respectively. In moPrP(Y156R1,H186A), line broadening between Cu(II) and the spin probe was not observed. These results suggest that a novel Cu(II) binding site is involved in His186 in the Helix2 region of the C-terminal domain of moPrP(C).


Asunto(s)
Cobre/metabolismo , Histidina/metabolismo , Proteínas PrPC/metabolismo , Animales , Histidina/genética , Interacciones Hidrofóbicas e Hidrofílicas , Ratones , Proteínas PrPC/genética , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
4.
Nucleosides Nucleotides Nucleic Acids ; 39(1-3): 439-452, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31560250

RESUMEN

The combination of low dose of radiation and an anticancer drug is a potent strategy for cancer therapy. Nucleoside analogs are known to have a radiosensitizing effects via the inhibition of DNA damage repair after irradiation. Certain types of nucleoside analogs have the inhibitory effects on RNA synthesis, but not DNA synthesis, with multiple functions in cell cycle modulation and apoptosis. In this review, the most up-to-date findings regarding radiosensitizing nucleoside analogs will be discussed, focusing especially on the mechanisms of action.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Nucleósidos/análogos & derivados , Nucleósidos/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Animales , Línea Celular Tumoral , Daño del ADN , Modelos Animales de Enfermedad , Humanos , Estructura Molecular , Purinas/química , Purinas/farmacología , Pirimidinas/química , Pirimidinas/farmacología , Radiación Ionizante , Radioterapia/efectos adversos , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Biochem Biophys Res Commun ; 366(1): 244-9, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-18062918

RESUMEN

We examined the influence of D177N (D178N in humans) mutation on the conformational stability of the S2 region of moPrP(C) with varying pHs by using the SDSL-ESR technique. The ESR spectrum of D177N at pH 7.5 was narrower than that of Y161R1, referred to as WT( *). The ESR spectrum of D177N did not change when pH in the solution decreased to pH 4.0. Our results suggested that the disappearance of a salt bridge (D177-R163) induced the increase in the instability of S2 region. Moreover, the line shape of the ESR spectrum obtained from H176S neighboring the salt bridge linked to the S2 region was similar to D177N. These results indicate that the protonation of H176 is strongly associated with the stability of S2 region. These findings are important for understanding the mechanism by which the disruption of the salt bridge in the S2 region forms the pathogenic PrP(Sc) structure in hereditary prion disease.


Asunto(s)
Proteínas PrPC/química , Proteínas PrPC/ultraestructura , Mutagénesis Sitio-Dirigida , Proteínas PrPC/genética , Conformación Proteica , Pliegue de Proteína , Relación Estructura-Actividad
6.
Cancer Lett ; 259(1): 71-81, 2008 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-17967504

RESUMEN

To investigate the mechanism of radioresistance of solid tumor cells, we created two expression vectors encoding Survivin mutants, T34A and D53A. When T34A and D53A were overexpressed in NIH3T3, A549 and HeLa cells, radiation-induced apoptosis was significantly enhanced. Furthermore, we examined the binding capability of Survivin with Smac/DIABLO in the cells that overexpressed these mutants. Coimmunoprecipitation analysis revealed that mutant form of Survivin, D53A and T34A could bind to Smac/DIABLO, but with much less affinity compared to the authentic form. These results suggest that radiation-induced apoptosis of tumor cells is increased by inhibition of the interaction between Survivin and Smac/DIABLO through overexpression of T34A and D53A.


Asunto(s)
Apoptosis/efectos de la radiación , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/radioterapia , Tolerancia a Radiación , Transducción de Señal/efectos de la radiación , Adenoviridae/genética , Animales , Proteínas Reguladoras de la Apoptosis , Caspasas/metabolismo , Citocromos c/metabolismo , Vectores Genéticos , Células HeLa , Humanos , Proteínas Inhibidoras de la Apoptosis , Ratones , Proteínas Asociadas a Microtúbulos/genética , Mitocondrias/enzimología , Mitocondrias/efectos de la radiación , Mutación , Células 3T3 NIH , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Fosforilación , Survivin , Factores de Tiempo , Transfección , Rayos X
7.
J Radiat Res ; 49(2): 113-21, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18075225

RESUMEN

The aim of this study is to evaluate the individual differences in radiosensitivity of lineage-committed myeloid hematopoietic progenitors, colony-forming cells (CFC), detected in steady-state human peripheral blood (PB). Mononuclear cells were prepared from the buffy-coat of 30 individuals PB, and were assayed for CFC by semi-solid culture supplemented with cytokines. X irradiation was performed in the range of 0.5-4 Gy at a dose rate of about 80 cGy/min. The mean number of hematopoietic progenitor cells is 5866 alpha 3408 in 1 ml of buffy-coat, suggesting that the erythroid progenitor cells are the major population. The total CFC radiosensitivity parameter D(0) and n value are 1.18 alpha 0.24 and 1.89 alpha 0.98, respectively. Using a linear regression analysis, a statistically significant correlation is observed between the D(0) value and the surviving fraction at 4 Gy (r = 0.611 p < 0.001). Furthermore, we evaluate the relationship between individual radiosensitivity and the level of antioxidants, plasma uric acid, plasma bilirubin, and intracellular glutathione. No statistically significant correlations are observed, however, between the D(0) parameter and the level of antioxidants, plasma uric acid, plasma bilirubin, and intracellular glutathione. The present study demonstrates that there are large individual differences in the radiosensitivity of hematopoietic progenitor cells as detected in steady-state human PB. These differences demonstrate almost no correlation with plasma or intracellular antioxidants. The prediction of individual differences in radiosensitivity of CFC can only be measured by 4 Gy irradiation.


Asunto(s)
Células Madre Hematopoyéticas/efectos de la radiación , Tolerancia a Radiación , Humanos , Técnicas In Vitro , Células Madre/efectos de la radiación
8.
J Radiat Res ; 49(2): 153-61, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18227621

RESUMEN

Our previous study showed that X irradiation induced the expression of death receptor DR5 on the cell surface in tumor cell lines under not only normoxia but also hypoxia. X irradiation combined with TNF alpha-related apoptosis-inducing ligand (TRAIL), which is the ligand of DR5, induced apoptosis in vitro (Takahashi et al., (2007) Journal of Radiation Research, 48: 461-468). In this report, we examined the in vivo antitumor efficacy of X irradiation combined with TRAIL treatment in tumor xenograft models derived from human gastric adenocarcinoma MKN45 and MKN28 cells in SCID mice. X irradiation combined with TRAIL synergistically suppressed the tumor growth rates in the xenograft models derived from MKN45 and MKN28 cells, which have wild type Tp53 and mutated Tp53, respectively, indicating that the antitumor effects occurred in a Tp53-independent manner. Histological analysis showed that the combination of X irradiation and TRAIL induced caspase-3-dependent apoptotic cell death. Moreover, the immunohistochemical detection of hypoxic regions using the hypoxic marker pimonidazole revealed that caspase-3-dependent apoptosis occurred in the hypoxic regions in the tumors. These results indicated that X irradiation combined with TRAIL may be a useful treatment to reduce tumor growth in not only normoxic but also hypoxic regions.


Asunto(s)
Adenocarcinoma/patología , Neoplasias Gástricas/patología , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Rayos X , Animales , Apoptosis , Hipoxia de la Célula , Humanos , Masculino , Ratones , Ratones SCID , Trasplante de Neoplasias , Trasplante Heterólogo , Células Tumorales Cultivadas
9.
J Vet Med Sci ; 70(5): 443-8, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18525164

RESUMEN

Lactoferrin has several biological activities, including antitumor activities in some human and animal tumor cells. Clinical trials have been carried out in human medicine based on these effects. However, the antitumor effects of lactoferrin in veterinary medicine remain unknown. In this in vitro study, we demonstrated that co-incubation of canine mammary gland tumor cells (CIPp and CHMp) and bovine lactoferrin induced growth arrest of tumor cells. This growth arrest was associated with induction of G1 arrest. Furthermore, this effect was stronger in tumor cells than in normal cells. These findings demonstrate that bovine lactoferrin has anti-tumor activity in canine mammary tumors and has the potential for use in tumor-bearing dogs.


Asunto(s)
Adenocarcinoma/veterinaria , Antineoplásicos/farmacología , Lactoferrina/farmacología , Neoplasias Mamarias Animales/tratamiento farmacológico , Adenocarcinoma/tratamiento farmacológico , Animales , Bovinos , Línea Celular Tumoral , Perros , Relación Dosis-Respuesta a Droga , Factores de Tiempo
10.
Jpn J Vet Res ; 56(2): 99-107, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18828447

RESUMEN

This study was performed to examine whether the brain activities induced by noxious algesic chemical substances in anesthetized animals could be detected by blood oxygen-level-dependent functional magnetic resonance imaging (BOLD-fMRI). Multislice gradient echo images of the primary somatosensory cortex were obtained using a 7.05 T superconducting system and a one-turned surface coil centered over the primary somatosensory cortex of the 1.0%-isoflurane-anesthetized rat. The Z-score t-map of BOLD signals and its time-course analysis revealed that subcutaneous injection of formalin into the left forepaw immediately induced an early response in the contralateral primary sensory cortex lasting for a few minutes, followed by a late response until 20 min after stimulation. In contrast, injection of capsaicin into the left forepaw evoked only the early response. Furthermore, pretreatment with morphine completely abolished these responses induced by the chemical algesic substances. Thus BOLD-fMRI is a useful method to analyze the brain activities of painful stimulation in anesthetized animals.


Asunto(s)
Dolor/inducido químicamente , Dolor/metabolismo , Corteza Somatosensorial/fisiología , Animales , Capsaicina/farmacología , Relación Dosis-Respuesta a Droga , Miembro Anterior , Formaldehído/farmacología , Isoflurano/farmacología , Imagen por Resonancia Magnética , Dimensión del Dolor/métodos , Ratas , Ratas Sprague-Dawley , Organismos Libres de Patógenos Específicos , Estimulación Química
11.
J Clin Biochem Nutr ; 43(2): 51-7, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18818753

RESUMEN

Oxidative stress stimulates two opposite signaling pathways leading to cell death and cell survival. Preferential selection of survival signals leads to the protection of cells against damage induced by reactive oxygen species, whereas preferential acceleration of death signals can be used to advantage in tumor therapy with oxidizing agents such as ionizing radiation and anticancer drugs. In vitro and in vivo experiments using cultured mammalian cells and experimental animals showed that ERK was included in survival signals and SAPK and p38 MAPK in death signals in oxidative stress. The activation of SAPK/JNK and subsequent expression of death receptor Fas on the cell surface caused the induction of cell death. The results mean that the acceleration of the activation of SAPK/JNK might lead to the enhancement of cell death by oxidizing agents like ionizing radiation and anticancer drugs. In fact, when cultured mammalian cells were exposed to ionizing radiation with 2-nitroimidazole derivatives having electrophilicity, the lethal effect of ionizing radiation was found to be enhanced together with the activation of SAPK/JNK and the enhancement of Fas expression. The activation of both survival and death signals was suppressed by the antioxidants N-acetylcystein and Trolox, suggesting that both signaling pathways are redox-regulated.

12.
Int J Radiat Oncol Biol Phys ; 68(1): 218-28, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17448876

RESUMEN

PURPOSE: To examine the in vivo antitumor efficacy of X-irradiation combined with administration of a ribonucleoside anticancer drug, 1-(3-C-ethynyl-beta-D-ribo-pentofuranosyl)cytosine (TAS106, ECyd), to tumor cell-transplanted mice. METHODS AND MATERIALS: Colon26 murine rectum adenocarcinoma cells and MKN45 human gastric adenocarcinoma cells were inoculated into the footpad in BALB/c mice and severe combined immunodeficient mice, respectively. They were treated with a relatively low dose of X-irradiation (2 Gy) and low amounts of TAS106 (0.1 mg/kg and 0.5 mg/kg). The tumor growth was monitored by measuring the tumor volume from Day 5 to Day 16 for Colon26 and from Day 7 to Day 20 for MKN45. Histologic analyses for proliferative and apoptotic cells in the tumors were performed using Ki-67 immunohistochemical and terminal deoxynucleotidyl transferase-mediated nick end labeling staining. The expression of survivin, a key molecule related to tumor survival, was assessed by quantitative polymerase chain reaction and immunohistochemical analysis. RESULTS: When X-irradiation and TAS106 treatment were combined, significant inhibition of tumor growth was observed in both types of tumors compared with mice treated with X-irradiation or TAS106 alone. Marked inhibition of tumor growth was observed in half of the mice that received the combined treatment three times at 2-day intervals. Parallel to these phenomena, the suppression of survivin expression and appearance of Ki-67-negative and apoptotic cells were observed. CONCLUSIONS: X-irradiation and TAS106 effectively suppress tumor growth in mice. The inhibition of survivin expression by TAS106 is thought to mainly contribute to the suppression of the tumor growth.


Asunto(s)
Adenocarcinoma , Antineoplásicos/uso terapéutico , Neoplasias del Colon , Citidina/análogos & derivados , Neoplasias Gástricas , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/metabolismo , Adenocarcinoma/radioterapia , Animales , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/metabolismo , Neoplasias del Colon/radioterapia , Terapia Combinada , Citidina/uso terapéutico , Evaluación Preclínica de Medicamentos , Femenino , Proteínas Inhibidoras de la Apoptosis , Antígeno Ki-67/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Trasplante de Neoplasias , Proteínas Represoras , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/radioterapia , Survivin
13.
Radiat Res ; 167(5): 563-71, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17474786

RESUMEN

To clarify the relationship between CDC2 kinase activity and radiation-induced apoptosis, we examined whether the cyclin-dependent kinase (CDK) inhibitor purvalanol A enhanced radiation-induced apoptosis in gastric tumor cells. MKN45 cells exposed to 20 Gy of X rays increased the CDC2 kinase activity and the expression of regulatory proteins (phospho-CDC2 and cyclin B1) of the G2/M phase, followed by activation of the G2/M checkpoint, whereas the treatment of X-irradiated MKN45 cells with 20 microM purvalanol A suppressed the increase in the CDC2 kinase activity and expression of the G2/M-phase regulatory proteins and reduced the fraction of the cells in the G2/M phase in the cell cycle. Furthermore, this treatment resulted in not only a significant increase in radiation-induced apoptosis but also the loss of clonogenicity in both MKN45 (p53-wild) and MKN28 (p53-mutated) cells. The expression of anti-apoptosis proteins, inhibitor of apoptosis protein (IAP) family members (survivin and XIAP) and BCL2 family members (Bcl-X(L) and Bcl-2), in purvalanol A-treated cells with and without X rays was significantly lower than for cells exposed to X rays alone. These results suggest that the inhibition of radiation-induced CDC2 kinase activity by purvalanol A induces apoptosis through the enhancement of active fragments of caspase 3.


Asunto(s)
Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Proteína Quinasa CDC2/metabolismo , Neoplasias/enzimología , Purinas/farmacología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/efectos de la radiación , Proteínas de Ciclo Celular/metabolismo , División Celular/efectos de los fármacos , División Celular/efectos de la radiación , Línea Celular , Fase G2/efectos de los fármacos , Fase G2/efectos de la radiación , Humanos , Neoplasias/patología , Rayos X
14.
J Radiat Res ; 48(6): 461-8, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17895594

RESUMEN

Our previous study showed that ionizing radiation induced the expression of death receptor DR5 on the cell surface in tumor cell lines and that the death receptor of the TNF alpha-related apoptosis-inducing ligand TRAIL enhanced the apoptotic pathway (Hamasu et al., (2005) Journal of Radiation Research, 46:103-110). The present experiments were performed to examine whether treatment with TRAIL enhanced the cell killing in tumor cells exposed to ionizing radiation under hypoxia, since the presence of radioresistant cells in hypoxic regions of solid tumors is a serious problem in radiation therapy for tumors. When human lung carcinoma A549 cells were irradiated under normoxia and hypoxia, respectively, radiation-induced enhancement of expression of DR5 was observed under both conditions. Incubation in the presence of TRAIL enhanced the caspase-dependent and chymotrypsin-like-protease-dependent apoptotic cell death in A549 cells exposed to X rays. Furthermore, it was shown that treatment with TRAIL enhanced apoptotic cell death and loss of clonogenic ability in A549 cells exposed to X rays not only under normoxia but also under hypoxia, suggesting that combination treatment with TRAIL and X irradiation is effective for hypoxic tumor cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/antagonistas & inhibidores , Ligando Inductor de Apoptosis Relacionado con TNF/administración & dosificación , Factor de Necrosis Tumoral alfa/metabolismo , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/efectos de la radiación , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Humanos , Oxígeno/metabolismo , Tolerancia a Radiación/efectos de los fármacos , Rayos X
15.
Biofactors ; 29(2-3): 113-21, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17673828

RESUMEN

Oxidative damage due to ischemia/reperfusion has been implicated as one of the leading causes for delayed neuronal cell death in a number of neurodegenerative diseases, including stroke. The purpose of this research was to investigate whether oral administration of a fermented grain food mixture (AOB(R)) might offer protective effects against ischemia/reperfusion-induced neuronal damage in Mongolian gerbils, a model known for delayed neuronal death in the hippocampal CA1 region. Histological analysis revealed that AOB administration ad libitum for 3 weeks (preoperative administration) and 1 week (postoperative administration) dose-dependently suppressed the induction of transient ischemia/reperfusion-induced neuronal cell death. TUNEL assay also revealed that AOB suppressed it by inhibiting the induction of apoptosis. A significant increase of superoxide dismutase-like (SOD-like) activity was observed in the hippocampal CA1 region of the AOB-treated gerbil. Furthermore, immunoblot analysis showed that AOB administration down-regulated the expression of heat shock proteins HSP27 and HSP70 in the same region. These results indicated that oral administration of AOB protected against ischemia/reperfusion-induced brain injury by minimizing oxidative damage via its SOD-like activity and inhibiting apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Neuronas/patología , Fenoles/uso terapéutico , Extractos Vegetales/uso terapéutico , Daño por Reperfusión/prevención & control , Administración Oral , Animales , Peso Corporal/efectos de los fármacos , Isquemia Encefálica/patología , Ingestión de Alimentos/efectos de los fármacos , Gerbillinae , Proteínas de Choque Térmico/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/patología , Masculino , Fenoles/administración & dosificación , Extractos Vegetales/administración & dosificación , Superóxido Dismutasa/metabolismo
16.
Chem Biodivers ; 4(9): 2253-67, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17886845

RESUMEN

An amphiphilic alpha-phenyl-N-(tert-butyl) nitrone (PBN) derivative, N-{[4-(lactobionamido)methyl]benzylidene}-1,1-dimethyl-2-(octylsulfanyl)ethylamine N-oxide (LPBNSH), newly synthesized from its original form PBN in hopes of clinical use, was intraperitoneally administered to Long-Evans Cinnamon (LEC) rats every 2 days at the concentrations of 0.1, 0.5, 1.0, and 2.0 mg/kg. We found that LPBNSH protected against copper-induced hepatitis with jaundice in LEC rats at concentrations of 0.1 and 0.5 mg/kg, which were extremely low compared with that of PBN. It also effectively prevented the loss of body weight, reduced the death rate, and suppressed the increase in serum aspartate aminotransferase and alanine aminotransferase values arising from fulminant hepatitis with jaundice at the same concentrations. Similar results were observed when PBN was administered at the concentration of 150 mg/kg. Immunohistochemical analysis of 8-hydroxy-2'-deoxyguanosine and measurement of thiobarbituric acid-reactive substances in the liver showed that LPBNSH largely suppressed the formation of these oxidative products at same concentrations. No difference in the abnormal accumulation of copper in the liver between the LPBNSH administered and control groups was observed. From these results, it was concluded that LPBNSH exhibited liver-protective effects against fulminant hepatitis with jaundice at ca. 1/1000, 500 the molar concentration of PBN and, therefore, was clinically promising.


Asunto(s)
Antioxidantes/uso terapéutico , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Cobre/toxicidad , Disacáridos/uso terapéutico , Iminas/uso terapéutico , Fallo Hepático Agudo/prevención & control , 8-Hidroxi-2'-Desoxicoguanosina , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Cobre/análisis , Óxidos N-Cíclicos/uso terapéutico , Desoxiguanosina/análogos & derivados , Desoxiguanosina/análisis , Inmunohistoquímica , Hígado/química , Hígado/patología , Fallo Hepático Agudo/inducido químicamente , Fallo Hepático Agudo/patología , Masculino , Ratas , Ratas Endogámicas LEC , Sustancias Reactivas al Ácido Tiobarbitúrico/análisis
17.
J Vet Med Sci ; 69(10): 993-7, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17984584

RESUMEN

The present study evaluated the costimulatory effects of complement receptor type 3 (CR3) and Fc receptor for IgG (FcgammaR) on superoxide production and intracellular signal transduction in bovine neutrophils. Stimulation with opsonized zymosan (OPZ) and heat-aggregated bovine IgG (Agg-IgG) resulted in much greater superoxide production and chemiluminescent (CL) responses in normal neutrophils compared with those stimulated with OPZ or Agg-IgG only. Superoxide production and CL response were closely associated with the stimulant-induced rise of the intracellular calcium ([Ca2+]i) concentration, amount of tyrosine phosphorylated 100 kDa protein, and activation of p38 mitogen-activated protein kinase (p38 MAPK). No costimulatory effect was found for these receptors on superoxide production in CR3-deficient neutrophils. Costimulation of CR3 and FcgammaR on bovine neutrophils leads to enhancement of superoxide production and their signaling pathways and appears to be associated with enhancement of neutrophil functions.


Asunto(s)
Neutrófilos/metabolismo , Receptores de Complemento/metabolismo , Receptores de IgG/metabolismo , Transducción de Señal , Superóxidos/metabolismo , Animales , Bovinos , Células Cultivadas , Femenino , Regulación de la Expresión Génica , Neutrófilos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Zimosan/farmacología
18.
Leuk Res ; 30(12): 1555-61, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16730061

RESUMEN

To clarify the role of lipid rafts in 2-chloro-2'-deoxyadenosine (2CdA; Cladribine)-induced apoptosis, the effects of disruption of lipid rafts by methyl-beta-cyclodextrin (MbetaCD) and filipin on 2CdA-induced apoptosis were investigated in four human acute lymphoblastic leukemia (ALL) cell lines comprised of T cells (MOLT-4, Jurkat) and B cells (NALM, BALL-1). The disruption of lipid rafts significantly inhibited 2CdA-induced apoptosis, indicating the crucial role of lipid rafts in the induction of apoptosis in leukemia cells. These reagents significantly inhibited 2CdA-induced elevation of the intracellular calcium concentration ([Ca(2+)](i)) in MOLT-4 cells, and 2CdA-induced apoptosis was partly inhibited by the Ca(2+) chelators BAPTA-AM and EGTA, and the L-type Ca(2+) channel blocker nifedipine. On the other hand, they had no effects on the cellular uptake of 2CdA. These results indicated that lipid rafts partly contributed to 2CdA-induced apoptosis by regulating Ca(2+) influx via the plasma membrane.


Asunto(s)
Apoptosis/efectos de los fármacos , Cladribina/farmacología , Microdominios de Membrana/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Calcio/metabolismo , Línea Celular Tumoral , Quelantes/farmacología , Cladribina/antagonistas & inhibidores , Ensayos de Selección de Medicamentos Antitumorales , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Filipina/farmacología , Humanos , Microdominios de Membrana/efectos de los fármacos , Nifedipino/farmacología , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Factores de Tiempo , beta-Ciclodextrinas/farmacología
19.
Radiat Res ; 166(2): 345-51, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16881735

RESUMEN

In the present study, we investigated whether X-irradiated hematopoietic stem cells can be induced to undergo megakaryocytopoiesis and thrombopoiesis in vitro using cytokine combinations that have been demonstrated to be effective for conferring increased survival on irradiated human CD34(+) megakaryocytic progenitor cells (colony-forming unit megakaryocytes; CFU-Meg), such as thrombopoietin (TPO), interleukin 3 (IL3), stem cell factor and FLT3 ligand. Culture of nonirradiated CD34(+) cells in serum-free medium supplemented with multiple cytokine combinations led to an approximately 200- to 600-fold increase in the total cell numbers by day 14 of culture. In contrast, the growth of X-irradiated cells was observed to be one-sixth to one-tenth that of the nonirradiated cultures. Similarly, total megakaryocytes were increased by 50- to 130-fold, while culture of X-irradiated cells yielded one-fourth to one-eighth of the control numbers. At this time, CD41(+) particles, which appeared to be platelets, were produced in the medium harvested from nonirradiated and irradiated cultures. Although radiation suppressed cell growth and megakaryocytopoiesis, there were no significant differences in thrombopoiesis between the two types of culture. These results suggest that X-irradiated CD34(+) cells can be induced to undergo nearly normal terminal maturation through megakaryocytopoiesis and thrombopoiesis by stimulation with appropriate cytokine combinations.


Asunto(s)
Diferenciación Celular , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de la radiación , Megacariocitos/citología , Trombopoyesis , Antígenos CD34/metabolismo , Biomarcadores , Plaquetas/metabolismo , Plaquetas/efectos de la radiación , Células Cultivadas , Células Madre Hematopoyéticas/metabolismo , Humanos , Activación Plaquetaria/efectos de la radiación , Rayos X
20.
FEMS Immunol Med Microbiol ; 46(2): 236-43, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16487305

RESUMEN

Severe acute respiratory syndrome (SARS) is caused by SARS-coronavirus (SARS-CoV). Infection of Vero E6 cells with SARS-CoV inhibits cell proliferation. Our previous study indicated that Akt, which is poorly phosphorylated in confluent cultures of Vero E6 cells, is phosphorylated and then dephosphorylated upon infection by SARS-CoV. In the present study, we showed that a serine residue of Akt was phosphorylated in Vero E6 cells in subconfluent culture and that Akt was dephosphorylated rapidly after SARS-CoV infection without up-regulation of its phosphorylation. Phosphorylation of glycogen synthase kinase-3beta, which is one of the downstream targets of Akt, was prevented in SARS-CoV-infected cells. However, treatment with glycogen synthase kinase-3beta small interfering RNA indicated that the glycogen synthase kinase-3beta signaling pathway was not related to inhibition of cell proliferation. Treatment of Vero E6 cells with the phosphatidylinositol 3'-kinase/Akt inhibitor, LY294002, which induces dephosphorylation of Akt, inhibited cell proliferation. As shown in our previous studies, apoptosis occurred in virus-infected cells within 18 h postinfection. Cellular mRNA transcription, which was reported to be up-regulated in SARS-CoV-infected Caco-2 cells, was not up-regulated in virus-infected Vero E6 cells, partially as a result of apoptosis. These results suggested that inhibition of cell proliferation is regulated by both the phosphatidylinositol 3'-kinase/Akt signaling pathway and by apoptosis in SARS-CoV-infected Vero E6 cells. This is the first study to analyze SARS-CoV-induced cell growth inhibition.


Asunto(s)
Proliferación Celular , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/patogenicidad , Animales , Apoptosis , Chlorocebus aethiops , Regulación de la Expresión Génica , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Células Vero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA