Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cureus ; 16(5): e59684, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38836164

RESUMEN

SMARCB1 is a gene known to cause carcinogenesis in many soft tissue tumors, including malignant rhabdoid tumors and epithelioid sarcoma. Since the first report of a subtype of sinonasal carcinoma characterized by a deficiency of the SMARCB1 gene in 2014 to date, fewer than 200 cases have been reported. We report a case of SMARCB1-deficient sinonasal carcinoma with clear cell morphology. In our case, there are no evident basaloid or plasmacytoid/rhabdoid tumor cells, which are typical histopathological features of SMARCB1-deficient sinonasal carcinoma. SMARCB1-deficient sinonasal carcinoma is prone to recurrence and has a very poor prognosis. As the development of molecularly targeted agents progresses, therapeutic efficacy is expected to improve. Simultaneously, the importance of early and accurate diagnosis of SMARCB1-deficient sinonasal carcinoma will increase. With the limited information provided by biopsy specimens, it is necessary to confirm the loss of SMARCB1 expression by immunohistochemistry and investigate the presence of SMARCB1 gene deletion by molecular genetics, considering the possibility of SMARCB1-deficient sinonasal carcinoma even in atypical cases without basaloid or plasmacytoid/rhabdoid cell morphology, as in our case.

2.
Exp Toxicol Pathol ; 67(3): 245-51, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25577727

RESUMEN

Hepatocellular hypertrophy in association with drug-metabolizing enzyme induction is considered to be an adaptive change associated with drug metabolism. To improve our understanding of liver hypertrophy, we determined the effect of a single ip injection of either lipopolysaccharide (LPS) or vehicle in male F344 rats with hepatocellular hypertrophy induced by oral delivery of p,p'-DDT for 2 weeks. The rats were sacrificed 3h or 24h after LPS or vehicle injection. LPS induced a focal hepatocellular necrosis in rats fed the control diet. When rats pre-treated with p,p'-DDT were injected with LPS, necrotic foci surrounded by ballooned hepatocytes were observed in the liver. The change was consistent with reduced LPS-mediated increases in plasma hepatic biomarkers, neutrophil influx, and apoptosis, and also associated with hepatic mRNA levels of TNF-α, CYPs, and NOS2. By contrast, when combined with p,p'-DDT and LPS, faint hepatocellular fatty change was extended, together with a synergistic increase in total blood cholesterol. These results suggest that hepatocytes exposed to p,p'-DDT are protected from the cell-lethal toxic effects of an exogenous stimulus, resulting in cell ballooning rather than necrosis in association with reduced inflammation and apoptosis, but compromised by an adverse effect on lipid metabolism.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/patología , DDT/toxicidad , Lipopolisacáridos/toxicidad , Hígado/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Hígado/patología , Masculino , Ratas , Ratas Endogámicas F344 , Reacción en Cadena en Tiempo Real de la Polimerasa
3.
J Toxicol Sci ; 38(5): 775-82, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24067725

RESUMEN

Emerging evidence suggests that chronic exposure to DDT and its derivatives is associated with a variety of human disorders such as anemia. The present study demonstrated that p,p'-DDT caused microcystic anemia in a dose-dependent manner (0, 5, 50, and 500 ppm) in the long-term study up to 2 years. To elucidate the mechanism(s) by which p,p'-DDT induces anemia, certain hematological parameters were assessed in rats fed specific doses of p,p'-DDT for 2 weeks, and the effect of lipopolysaccharide on anemia of inflammation was also examined in p,p'-DDT-treated rats. The parameters included the content of hemoglobin per reticulocyte, mean corpuscular volume of reticulocytes and mature erythrocytes, corpuscular hemoglobin concentration mean of mature erythrocytes, and saturation levels of transferrin and iron. During the 2-week treatment period, hypochromic microcytic reticulocytes and hypochromic normocytic mature erythrocytes were observed in p,p'-DDT-treated rats, with no evidence of alteration in plasma iron levels. p,p'-DDT enhanced microcytosis of reticulocytes, as well as mature erythrocytes, which occurred due to severe hypoferremia resulting from anemia of inflammation; however, plasma iron levels were attenuated probably through the inhibition of interleukin-6. Our data suggests that long-term treatment with p,p'-DDT induces microcytic anemia, possibly because of the impairment of iron utility in erythrocytes.


Asunto(s)
Anemia Hipocrómica/sangre , Anemia Hipocrómica/inducido químicamente , DDT/toxicidad , Insecticidas/toxicidad , Animales , DDT/administración & dosificación , DDT/efectos adversos , Relación Dosis-Respuesta a Droga , Eritrocitos/metabolismo , Hemoglobinas/metabolismo , Insecticidas/administración & dosificación , Insecticidas/efectos adversos , Interleucina-6/antagonistas & inhibidores , Hierro/sangre , Hierro/metabolismo , Lipopolisacáridos , Masculino , Ratas , Ratas Endogámicas F344 , Reticulocitos/metabolismo , Factores de Tiempo , Transferrina/metabolismo
4.
J Carcinog ; 11: 10, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22919283

RESUMEN

BACKGROUND: Caloric restriction (CR), a lowering of caloric intake without malnutrition, is associated with longevity. CR also decreases incidences of age-related diseases including cancer. The sirtuins (SIRTs) have been implicated as a key mediator for the beneficial effects of CR on longevity. However, the underlying mechanisms by which CR decreases cancer risk have not yet been fully elucidated. MATERIALS AND METHODS: The present study was conducted to determine whether CR would modify the growth of preneoplastic colonic aberrant crypt foci (ACF). We also analyzed the expression of SIRTs to elucidate the molecular mechanisms of cancer-preventive effects of CR. F344 rats were fed a CR diet (60% of ad libitum diet) or a basal diet ad libitum. Then, the animals were given subcutaneous injection of either 1, 2-dimethylhydrazine (DMH) that enhances cell proliferation of colonic mucosa or saline. All animals were sacrificed at 5 weeks after the beginning of the experiment. RESULTS: The number of ACF in colonic mucosa was significantly decreased in DMH-treated rats with CR as compared to in those without CR. No ACF was found in DMH-untreated animals with or without CR. Also, we found that CR decreased the cell proliferation of colonic mucosa in DMH-treated rats. The expressions of anti-apoptotic gene, Survivin, and cell cycle progression-associated gene, Cyclin D1, were increased by DMH-treatment. Both of the genes expressions were declined by CR in those of DMH-treated rats. The expressions of all SIRT1-7 mRNAs were significantly increased by CR in DMH-treated rats. CONCLUSION: As previous studies demonstrated that SIRT1 down-regulates Survivin and Cyclin D1, our findings suggest that at least SIRT1 protect colonic mucosa from formation and development of ACF by increasing apoptosis and reducing excessive cell growth in colon epithelial cells.

5.
Int J Cancer ; 130(10): 2300-9, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20017139

RESUMEN

Human T-cell leukemia virus type 1 (HTLV-1) is the causative agent of adult T-cell leukemia (ATL), which is an aggressive and fatal CD4(+) T cell malignancy. MicroRNA (miRNA), a novel class of RNA that regulates gene expression, is involved in many cellular processes such as growth, development and apoptosis. It has recently been linked to several cancer phenotypes. However, aberrant miRNA expression and its pathologic significance in ATL are not well documented. Here, we investigated the role of miRNAs in HTLV-1-related leukemogenesis. The results showed that miR-146a was upregulated in HTLV-1-infected T-cell lines compared to uninfected T-cell lines. Tax-induced miR-146a expression in a NF-κB-dependent manner and inhibited the expression of gene harboring the target sequence of miR-146a on its 3'UTR. Inhibition of miR-146a function by anti-miRNA inhibitor reduced the proliferation of HTLV-1-infected T-cell lines but not that of uninfected T-cell lines. Moreover, overexpression of miR-146a enhanced the growth of an HTLV-1-infected T-cell line. Our findings suggest that miR-146a is a potentially suitable therapeutic target of ATL.


Asunto(s)
Productos del Gen tax/fisiología , Virus Linfotrópico T Tipo 1 Humano/metabolismo , Leucemia-Linfoma de Células T del Adulto/genética , MicroARNs/genética , Linfocitos T/virología , Apoptosis , Línea Celular Tumoral , Humanos , Leucemia-Linfoma de Células T del Adulto/virología , MicroARNs/metabolismo , MicroARNs/fisiología , FN-kappa B/metabolismo , Linfocitos T/metabolismo , Linfocitos T/patología , Activación Transcripcional
6.
ISRN Microbiol ; 2012: 978607, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23762762

RESUMEN

Human T-cell leukemia virus type 1 (HTLV-1) is the pathogen that causes the aggressive and lethal malignancy of CD4+ T-lymphocytes called adult T-cell leukemia/lymphoma (ATLL). MicroRNAs (miRNAs), a class of short, noncoding RNAs, regulate gene expression by targeting mRNAs for translational repression or cleavage. miRNAs are involved in many aspects of cell biology linked with formation of several cancer phenotypes. However, the relation between miRNAs and pathologic implication in ATLL is not well elucidated. Here, we evaluated the roles of cellular miRNAs in ATLL caused by HTLV-1. We found that the expression of miR-155 was increased in HTLV-1-positive T-cell lines. miR-155 expression was enhanced by Tax and binding of transcription factors, NF- κ B and AP-1, on the transcription binding sites of miR-155 gene promoter region is important to increase the expression of miR-155 by Tax. Transfection of anti-miR-155 inhibitor, which inhibits the function of miR-155, inhibited the growth of HTLV-1-positive T-cell lines. On the other hand, the growth of HTLV-1-negative T-cell lines was not changed by transfection of anti-miR-155. Forced expression of miR-155 enhanced the growth of HTLV-1-positive T-cell lines. These findings indicate that targeting the functions of miRNAs is a novel approach to the prevention or treatment of ATLL.

7.
Inhal Toxicol ; 23(8): 476-85, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21689009

RESUMEN

Didecyldimethylammonium chloride (DDAC), a representative dialkyl-quaternary ammonium compound (QAC), could contaminate working atmospheres when used in disinfectant operation and adversely affect human health. Furthermore, the development of bacteria resistant to DDAC might become public health concern. We postulated that DDAC instillation in the lungs alters pulmonary antioxidant and antimicrobial responses and increases susceptibility to systemic administration of a bacterial component lipopolysaccharide (LPS). Mice were intratracheally instilled with DDAC and sacrificed 1, 3, or 7 days after treatment. Pulmonary cytotoxicity in recovered bronchoalveolar lavage was evident on Days 1 and 7, and inflammatory cell influx and interleukin-6 expression peaked on Day 7, in association with altered antioxidant and antimicrobial responses, as demonstrated by measuring heme oxygenase-1, glutathione peroxidase 2, lactoferrin, and mouse ß-defensin-2 and -3 mRNA in the lung samples. The impaired defense system tended to enhance the inflammatory reaction caused by a systemic administration of LPS; the effect was in association with increased expression of toll-like receptor-4 mRNA. The results suggest that DDAC alters pulmonary defense system, which may contribute to susceptibility to an exogenous infectious agent.


Asunto(s)
Lesión Pulmonar Aguda/inducido químicamente , Contaminantes Ocupacionales del Aire/toxicidad , Inmunidad Innata/efectos de los fármacos , Pulmón/efectos de los fármacos , Compuestos de Amonio Cuaternario/toxicidad , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/metabolismo , Animales , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Expresión Génica/efectos de los fármacos , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/metabolismo , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Inmunidad Innata/genética , Interleucina-6/metabolismo , Intubación Intratraqueal , Lactoferrina/genética , Lactoferrina/metabolismo , Lipopolisacáridos/farmacología , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , beta-Defensinas/genética , beta-Defensinas/metabolismo
9.
Cancer Sci ; 101(5): 1204-11, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20180813

RESUMEN

Aurora A kinase plays an essential role in the proper assembly and function of the mitotic spindle. We have shown previously that Aurora A expression is increased aberrantly in human T-cell leukemia virus type 1 (HTLV-1)-infected T-cell lines and primary adult T-cell leukemia cells, and a pan-Aurora kinase inhibitor, which inhibits both Aurora A and Aurora B kinases, reduces viability and induces apoptosis in these cells. However, the specific effects of Aurora A inhibition on HTLV-1-infected T-cells are poorly understood. In this study, we addressed this question by comparing the effects of MLN8237, a selective inhibitor of Aurora A, on cell viability, cell cycle progression, and induction of apoptosis in HTLV-1-infected and -uninfected T-cell lines. MLN8237 reduced the viability of HTLV-1-infected T-cell lines within 24 h, but its effects on that of HTLV-1-uninfected T-cell lines were moderate. MLN8237 induced early apoptosis of HTLV-1-infected T-cell lines without induction of polyploidy. It induced p53 and p21 expression in HTLV-1-infected but not in -uninfected T-cell lines, suggesting that MLN8237-treated HTLV-1-infected T-cell lines exit from mitosis and activate a p53-dependent postmitotic G(1) checkpoint, leading to G(1) arrest followed by the induction of apoptosis. Our results suggest that specific inhibition of Aurora A kinase is a potentially useful therapeutic strategy in the treatment of adult T-cell leukemia and that further in vivo exploration is warranted.


Asunto(s)
Azepinas/farmacología , Leucemia-Linfoma de Células T del Adulto/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Pirimidinas/farmacología , Linfocitos T/virología , Apoptosis/efectos de los fármacos , Aurora Quinasa B , Aurora Quinasas , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Leucemia-Linfoma de Células T del Adulto/patología
10.
Int J Cancer ; 127(7): 1584-94, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20091867

RESUMEN

Aurora kinases play an essential role in regulating mitosis and cell division. Inhibition of Aurora kinases results in suppression of cell division, phosphorylation of histone H3 and induction of apoptosis in many cell types. These characteristics have prompted the testing of Aurora kinase inhibitors as chemotherapeutic agents. In our study, we report the in vitro activities of AZD1152, a selective inhibitor of Aurora B kinase in human T-cell leukemia virus type 1 (HTLV-1), the causative agent of adult T-cell leukemia (ATL), -infected T-cell lines. Overexpression of Aurora B was noted in HTLV-1-infected T-cell lines compared to HTLV-1-uninfected T-cell lines. AZD1152 reduced the viability of HTLV-1-infected T-cell lines within 24 hr but did not affect that of -uninfected T-cell lines. Although AZD1152 inhibited phosphorylation of histone H3 on Ser10 in both HTLV-1-infected and -uninfected T-cell lines, it induced polyploidy only in HTLV-1-uninfected T-cell lines. AZD1152 induced early apoptosis of HTLV-1-infected T-cells without induction of polyploidy. We have reported previously that a pan-Aurora kinase inhibitor induced apoptosis through inhibition of NF-kappaB signaling activity in HTLV-1-infected T-cell lines. In contrast, AZD1152 did not affect NF-kappaB activity in these cells. It induced p53 and p21 expression in HTLV-1-infected but not in HTLV-1-uninfected T-cell lines, suggesting that activation of p53-dependent postmitotic checkpoint might prevent polyploidy in HTLV-1-infected T-cells. Our results suggest that specific inhibition of Aurora B kinase is a potentially useful therapeutic strategy in the treatment of ATL and that further in vivo exploration is warranted.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Virus Linfotrópico T Tipo 1 Humano/patogenicidad , Organofosfatos/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Quinazolinas/farmacología , Linfocitos T/virología , Aurora Quinasa B , Aurora Quinasas , Linfocitos B/citología , Linfocitos B/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Virus Linfotrópico T Tipo 1 Humano/efectos de los fármacos , Humanos , Células Jurkat/efectos de los fármacos , Células Jurkat/patología , FN-kappa B/efectos de los fármacos , FN-kappa B/fisiología , ARN Interferente Pequeño/genética , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Transfección
11.
Toxicology ; 267(1-3): 118-24, 2010 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-19895865

RESUMEN

Chromated copper arsenate, which is used worldwide as a wood preservative, can adversely affect human health. Accumulating evidence suggests that chromium (Cr) and arsenic (As) can potentially disrupt the redox balance and cause respiratory diseases and cancer in humans. The present study was designed to determine the combined toxic effects of these metals in the lungs and to clarify the specific molecules that are stimulated by combined exposure to both metals. Male C57BL/6J mice were intratracheally instilled with arsenate [As(V)], hexavalent chromium [Cr(VI)], or a combination of both metals. Mice were sacrificed 2 days after treatment to collect bronchoalveolar lavage fluid and lung tissue samples. Inflammation, cytotoxicity, apoptosis, and oxidative stress markers were measured. Our results indicated that administration of Cr(VI) alone or in combination with As(V) induced neutrophil-dominant inflammation as well as phosphorylation of mitogen-activated protein kinases; effects of treatment with As(V) alone were comparatively less potent. By analyzing the production of interleukin-6 and activity of lactate dehydrogenase and caspase, we confirmed that co-treatment intensified pulmonary injury and that it was accompanied by oxidative stress, as confirmed by marked increases in the production of reactive oxygen species, reduced glutathione content, and thioredoxin reductase (TRXRD) activity. Expressed mRNA levels of heme oxygenase-1, glutamylcysteine ligase, glutathione peroxidase 2, thioredoxin (TRX) 1, and TRXRD1 were also enhanced by co-treatment, whereas treatment with As(V) alone reduced the mRNA expression level of TRX2. Our data suggest that co-treatment with As(V) exacerbated Cr(VI)-induced pulmonary injury and that this effect may be exerted through a disruption in the balance among several antioxidant genes.


Asunto(s)
Arseniatos/toxicidad , Cromo/toxicidad , Lesión Pulmonar/inducido químicamente , Pulmón/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Biomarcadores/metabolismo , Caspasas/metabolismo , Glutatión Peroxidasa/metabolismo , Hemo-Oxigenasa 1/metabolismo , Interleucina-6/biosíntesis , L-Lactato Deshidrogenasa/metabolismo , Pulmón/enzimología , Pulmón/metabolismo , Lesión Pulmonar/enzimología , Lesión Pulmonar/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo/efectos de los fármacos , Neumonía/inducido químicamente , Neumonía/metabolismo , Reductasa de Tiorredoxina-Disulfuro/metabolismo
12.
Exp Toxicol Pathol ; 62(6): 643-51, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19762220

RESUMEN

Didecyldimethylammonium chloride (DDAC) is used worldwide as a germicide, in antiseptics, and as a wood preservative, and can cause adverse pulmonary disease in humans. However, the pulmonary toxicity of DDAC has not yet been thoroughly investigated. Mice were intratracheally instilled with DDAC to the lung and the bronchoalveolar lavage (BAL) fluid and lung tissues were collected to assess dose- and time-related pulmonary injury. Exposure to 1500 µg/kg of DDAC caused severe morbidity with pulmonary congestive oedema. When the BAL fluid from survivors was examined on day 3 after treatment, exposure to 150 µg/kg of DDAC caused weakly induced inflammation, and exposure to 15µg/kg did not cause any visible effects. Next, we observed pulmonary changes that occurred up to day 20 after 150 µg/kg of DDAC exposure. Pulmonary inflammation peaked on day 7 and was confirmed by expression of interleukin-6, monocyte chemotactic protein-1, macrophage inflammatory protein (MIP)-1α, MIP-1ß, and regulated upon activation, normal T-cell expressed and secreted in the BAL fluid; these changes were accompanied by altered gene expression of their chemokine (C-C motif) receptor (Ccr) 1, Ccr2, Ccr3, and Ccr5. Cytotoxicity evoked by DDAC was related to the inflammatory changes and was confirmed by an in vitro study using isolated mouse lung fibroblasts. The inflammatory phase was accompanied or followed by pulmonary remodeling, i.e., fibrosis, which was evident in the mRNA expression of type I procollagen. These results suggest that administering DDAC by intratracheal instillation causes pulmonary injury in mice, and occupational exposure to DDAC might be a potential hazard to human health.


Asunto(s)
Neumonía/inducido químicamente , Fibrosis Pulmonar/inducido químicamente , Compuestos de Amonio Cuaternario/toxicidad , Animales , Líquido del Lavado Bronquioalveolar/citología , Células Cultivadas , Colágeno Tipo I/genética , Relación Dosis-Respuesta a Droga , Pulmón/efectos de los fármacos , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Quimiocina/genética
13.
J Toxicol Sci ; 34(5): 527-39, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19797861

RESUMEN

A 4-week repeated dose oral toxicity study of phenobarbital (PB) sodium was conducted in F344 rats of both sexes at PB doses of 0.8, 8, and 80 mg/kg/day to fully elucidate its general toxicity including hematological changes. Both sexes in the 80 mg/kg/day group showed staggering gait, lacrimation, and/or sedation, which were more evident in the early stage of treatment. The body weight gain and food consumption were greater in these animals than in controls. Hematology revealed a significant reduction in the hematocrit (Ht), hemoglobin concentration (Hb), and erythrocyte count (RBC) in both sexes at 80 mg/kg/day, which was accompanied by a decrease in the cell mean Hb (CHCM) in mature erythrocytes with an increase in unsaturated iron binding capacity. Female rats also showed reduction in the CHCM in reticulocytes, content of hemoglobin per reticulocyte, and transferrin saturation. PB prolonged the activated partial thromboplastin time and inversely increased the platelet count with no evidence of platelet activation. Well-known toxic effects of PB on the liver and thyroid were observed in a dose-dependent manner, along with altered lipid, glucose, and electrolyte metabolism. The serum levels of PB increased dose-dependently, when examined in females received 8 and 80 mg/kg/day on day 1 and 28; there were no difference in C(max) and AUC(0-24) values between day 1 and day 28. These results indicated that PB has the potential to elicit multiple organ toxicity including an effect on the hematopoietic system. The hematological analysis provided evidence for hypochromic anemia, plausibly caused by the impairment of iron utility.


Asunto(s)
Anemia Hipocrómica/inducido químicamente , Fenobarbital/toxicidad , Administración Oral , Animales , Peso Corporal/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Femenino , Marcha/efectos de los fármacos , Hierro/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Tamaño de los Órganos/efectos de los fármacos , Fenobarbital/administración & dosificación , Fenobarbital/farmacocinética , Ratas , Ratas Endogámicas F344 , Pruebas de Toxicidad , Urinálisis
14.
Cancer Sci ; 100(5): 807-12, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19309363

RESUMEN

The Epstein-Barr virus-encoded latent membrane protein 1 is considered the Epstein-Barr virus oncogene based on its importance in Epstein-Barr virus-induced B-lymphocyte transformation. Beta-catenin is a potential oncogene, and its accumulation has been implicated in a variety of human cancers. Here, we found that beta-catenin protein was highly expressed in Epstein-Barr virus-immortalized B-cell lines compared with peripheral blood mononuclear cells from healthy donors. Beta-catenin expression in Epstein-Barr virus-immortalized B-cell line decreased following treatment with LY294002, an inhibitor of phosphatidylinositol 3-kinase. Treatment with LY294002 or knockdown of beta-catenin by small interfering RNA reduced the growth of Epstein-Barr virus-immortalized B-cell line. Transient transfection of latent membrane protein 1 expression plasmid increased beta-catenin protein expression and beta-catenin-dependent transcription. Latent membrane protein 1 deletions mutants lacking the carboxyl-terminal activating region 1 domain failed to enhance beta-catenin protein expression and beta-catenin-dependent transcriptional activity. They also failed to increase phosphorylated AKT expression. Dominant-negative AKT suppressed latent membrane protein 1-induced beta-catenin-dependent transcriptional activity. These results suggest that latent membrane protein 1 activates beta-catenin through the phosphatidylinositol 3-kinase/AKT signaling pathway. Activation of the beta-catenin pathway by Epstein-Barr virus may contribute to the lymphoproliferation characteristic of Epstein-Barr virus-infected B-cells.


Asunto(s)
Linfocitos B/metabolismo , Herpesvirus Humano 4/metabolismo , Transducción de Señal , Proteínas de la Matriz Viral/metabolismo , beta Catenina/metabolismo , Linfocitos B/citología , Línea Celular , Proliferación Celular , Herpesvirus Humano 4/genética , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factores de Transcripción TCF/metabolismo , Transcripción Genética/genética , Proteínas de la Matriz Viral/genética , beta Catenina/genética
15.
BMC Microbiol ; 9: 36, 2009 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-19216748

RESUMEN

BACKGROUND: The inflammatory response in Helicobacter pylori-infected gastric tissue is mediated by cag pathogenicity island (PAI)-dependent activation of nuclear factor-kappaB (NF-kappaB). Phosphatidylinositol 3-kinase (PI3K)/Akt signaling is known to play a role in NF-kappaB activation, but little information is available on the relationship between H. pylori and PI3K/Akt signaling in gastric epithelial cells. We examined whether H. pylori activates Akt in gastric epithelial cells, the role of cag PAI in this process and the role of Akt in regulating H. pylori-induced NF-kappaB activation. RESULTS: Phosphorylated Akt was detected in epithelial cells of H. pylori-positive gastric tissues. Although Akt was activated in MKN45 and AGS cells by coculture with cag PAI-positive H. pylori strains, a cag PAI-negative mutant showed no activation of Akt. H. pylori also induced p65 phosphorylation. PI3K inhibitor suppressed H. pylori-induced p65 phosphorylation and NF-kappaB transactivation, as well as interleukin-8 expression. Furthermore, transfection with a dominant-negative Akt inhibited H. pylori-induced NF-kappaB transactivation. Transfection with small interference RNAs for p65 and Akt also inhibited H. pylori-induced interleukin-8 expression. CONCLUSION: The results suggest that cag PAI-positive H. pylori activates Akt in gastric epithelial cells and this may contribute to H. pylori-mediated NF-kappaB activation associated with mucosal inflammation and carcinogenesis.


Asunto(s)
Helicobacter pylori/metabolismo , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor de Transcripción ReIA/metabolismo , Línea Celular , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiología , Regulación de la Expresión Génica , Islas Genómicas , Infecciones por Helicobacter/metabolismo , Helicobacter pylori/genética , Humanos , Interleucina-8/biosíntesis , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Interferencia de ARN
16.
Int J Cancer ; 124(11): 2607-15, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19230025

RESUMEN

Human T-cell leukemia virus type 1 (HTLV-1) is the etiologic agent for adult T-cell leukemia (ATL). Aurora A, a mitotic checkpoint protein, is overexpressed in human cancer cells. The cell cycle-dependent turnover of Aurora A is regulated by E3 ubiquitin ligases such as checkpoint with fork head-associated and ring finger (CHFR). Here, we found overexpression of Aurora A protein in HTLV-1-infected T-cell lines and primary ATL cells. The expression of CHFR mRNA was reduced in these cells by abnormal methylation of CHFR promoter region. Knockdown of Aurora A using small interfering RNA suppressed the growth of HTLV-1-infected T-cell line. Transfection of Aurora A expression plasmid enhanced Tax-induced nuclear factor-kappaB (NF-kappaB) reporter activity. Transfection of CHFR expression plasmid into an HTLV-1-infected T-cell line reduced cell growth, Aurora A protein level and constitutive NF-kappaB reporter activity. Aurora kinase inhibitor suppressed the growth and survival of HTLV-1-infected T-cell lines and primary ATL cells. It also reduced constitutive NF-kappaB activity in an HTLV-1-infected T-cell line by reducing IkappaB kinase beta phosphorylation and the expression of antiapoptotic protein survivin. Our results suggested that loss of CHFR expression resulted to accumulation of Aurora A, which increased NF-kappaB activity. These findings highlight the critical role of Aurora A in HTLV-1-infected T cells, making this molecule a potentially suitable target for future therapies for ATL.


Asunto(s)
Proteínas de Ciclo Celular/genética , Virus Linfotrópico T Tipo 1 Humano/fisiología , Leucemia-Linfoma de Células T del Adulto/patología , FN-kappa B/metabolismo , Proteínas de Neoplasias/genética , Proteínas Serina-Treonina Quinasas/fisiología , Linfocitos T/virología , Apoptosis , Aurora Quinasas , Ciclo Celular , Línea Celular , Proliferación Celular , Supervivencia Celular , Metilación de ADN , Humanos , Leucemia-Linfoma de Células T del Adulto/enzimología , Proteínas de Unión a Poli-ADP-Ribosa , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Transducción de Señal , Linfocitos T/fisiología , Ubiquitina-Proteína Ligasas
17.
Int J Cancer ; 124(3): 622-9, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-18973272

RESUMEN

Epstein-Barr Virus (EBV)-associated immunoblastic lymphoma occurs in immunocompromised patients such as those with AIDS or transplant recipients after primary EBV infection or reactivation of a preexisting latent EBV infection. In the present study, we evaluated the effect of ritonavir, an HIV protease inhibitor, on EBV-positive lymphoblastoid B cells in vitro and in mice model. We found that it induced cell-cycle arrest at G1-phase and apoptosis through down-regulation of cell-cycle gene cyclin D2 and antiapoptotic gene survivin. Furthermore, ritonavir suppressed transcriptional activation of NF-kappaB in these cells. Ritonavir efficiently prevented growth and infiltration of lymphoma cells in various organs of NOD/SCID/gammacnull mice at the same dose used for treatment of patients with AIDS. Our results indicate that ritonavir targets NF-kappaB activated in tumor cells and shows anti-tumor effects. These data also suggest that this compound may have promise for treatment or prevention of EBV-associated lymphoproliferative diseases that occur in immunocompromised patients.


Asunto(s)
Infecciones por Virus de Epstein-Barr/tratamiento farmacológico , Inhibidores de la Proteasa del VIH/farmacología , Linfoma de Células B/tratamiento farmacológico , FN-kappa B/efectos de los fármacos , Ritonavir/farmacología , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Ciclo Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ensayo de Cambio de Movilidad Electroforética , Infecciones por Virus de Epstein-Barr/complicaciones , Herpesvirus Humano 4 , Humanos , Hibridación Fluorescente in Situ , Linfoma de Células B/virología , Ratones , FN-kappa B/metabolismo , Reacción en Cadena de la Polimerasa , Activación Transcripcional/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Cancer Sci ; 99(11): 2286-94, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18771528

RESUMEN

Clinical trials for treatment of adult T-cell leukemia (ATL) caused by human T-cell leukemia virus type I (HTLV-I) using all-trans-retinoic acid (ATRA) have shown satisfactory therapeutic responses, although efficacies were limited. Recently, many synthetic retinoids have been developed and among them, a novel synthetic retinoid, Am80 (Tamibarotene) is an RARalpha- and RARbeta-specific retinoid expected to overcome ATRA resistance. The present study examined the inhibitory effects of Am80 on HTLV-I-infected T-cell lines and ATL cells. Am80 had negligible growth inhibition of peripheral blood mononuclear cells but marked growth inhibition of both HTLV-I-infected T-cell lines and ATL cells. Am80 arrested cells in the G1 phase of the cell cycle and induced apoptosis in HTLV-I-infected T-cell lines. It inhibited also the phosphorylation of IkappaBalpha and NF-kappaB-DNA binding, in conjunction with reduction of expression of proteins involved in the G1/S cell cycle transition and apoptosis. Am80 also inhibited the expression of JunD, resulting in suppression of AP-1-DNA binding. Furthermore, severe combined immunodeficient mice with tumors induced by subcutaneous inoculation of HTLV-I-infected T cells, responded to Am80 treatment with partial regression of tumors and no side-effects. These findings demonstrate that Am80 is a potential inhibitor of NF-kappaB and AP-1, and is a potentially useful therapeutic agent against ATL.


Asunto(s)
Antineoplásicos/farmacología , Benzoatos/farmacología , Leucemia-Linfoma de Células T del Adulto/tratamiento farmacológico , Tetrahidronaftalenos/farmacología , Animales , Antineoplásicos/uso terapéutico , Benzoatos/uso terapéutico , Ciclo Celular , División Celular , Línea Celular Tumoral , Femenino , Virus Linfotrópico T Tipo 1 Humano/patogenicidad , Humanos , Leucemia-Linfoma de Células T del Adulto/metabolismo , Leucemia-Linfoma de Células T del Adulto/patología , Ratones , Ratones SCID , Tetrahidronaftalenos/uso terapéutico
19.
Int J Cancer ; 123(11): 2702-12, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18798263

RESUMEN

Adult T-cell leukemia (ATL) is a fatal malignancy of T lymphocytes caused by human T-cell leukemia virus type 1 (HTLV-1) infection and remains incurable. Carotenoids are a family of natural pigments and have several biological functions. Among carotenoids, fucoxanthin is known to have antitumorigenic activity, but the precise mechanism of action is not elucidated. We evaluated the anti-ATL effects of fucoxanthin and its metabolite, fucoxanthinol. Both carotenoids inhibited cell viability of HTLV-1-infected T-cell lines and ATL cells, and fucoxanthinol was approximately twice more potent than fucoxanthin. In contrast, other carotenoids, beta-carotene and astaxanthin, had mild inhibitory effects on HTLV-1-infected T-cell lines. Importantly, uninfected cell lines and normal peripheral blood mononuclear cells were resistant to fucoxanthin and fucoxanthinol. Both carotenoids induced cell cycle arrest during G(1) phase by reducing the expression of cyclin D1, cyclin D2, CDK4 and CDK6, and inducing the expression of GADD45alpha, and induced apoptosis by reducing the expression of Bcl-2, XIAP, cIAP2 and survivin. The induced apoptosis was associated with activation of caspase-3, -8 and -9. Fucoxanthin and fucoxanthinol also suppressed IkappaBalpha phosphorylation and JunD expression, resulting in inactivation of nuclear factor-kappaB and activator protein-1. Mice with severe combined immunodeficiency harboring tumors induced by inoculation of HTLV-1-infected T cells responded to treatment with fucoxanthinol with suppression of tumor growth, showed extensive tissue distribution of fucoxanthinol, and the presence of therapeutically effective serum concentrations of fucoxanthinol. Our preclinical data suggest that fucoxanthin and fucoxanthinol could be potentially useful therapeutic agents for patients with ATL.


Asunto(s)
Antineoplásicos/uso terapéutico , Leucemia-Linfoma de Células T del Adulto/tratamiento farmacológico , Leucemia-Linfoma de Células T del Adulto/patología , Phaeophyceae/química , Xantófilas/uso terapéutico , beta Caroteno/análogos & derivados , Acetilación , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Caspasas/metabolismo , Supervivencia Celular/efectos de los fármacos , Femenino , Virus Linfotrópico T Tipo 1 Humano/efectos de los fármacos , Humanos , Leucemia-Linfoma de Células T del Adulto/metabolismo , Ratones , FN-kappa B/metabolismo , Factor de Transcripción AP-1/metabolismo , Células Tumorales Cultivadas , Xantófilas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , beta Caroteno/química , beta Caroteno/farmacología , beta Caroteno/uso terapéutico
20.
Retrovirology ; 5: 86, 2008 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-18808681

RESUMEN

BACKGROUND: Human T-cell leukemia virus type I (HTLV-I) is associated with pulmonary diseases, characterized by bronchoalveolar lymphocytosis, which correlates with HTLV-I proviral DNA in carriers. HTLV-I Tax seems to be involved in the development of such pulmonary diseases through the local production of inflammatory cytokines and chemokines in T cells. However, little is known about induction of these genes by HTLV-I infection in lung epithelial cells. RESULTS: We tested infection of lung epithelial cells by HTLV-I by coculture studies in which A549 alveolar and NCI-H292 tracheal epithelial cell lines were cocultured with MT-2, an HTLV-I-infected T-cell line. Changes in the expression of several cellular genes were assessed by reverse transcription-polymerase chain reaction, enzyme-linked immunosorbent assay and flow cytometry. Coculture with MT-2 cells resulted in infection of lung epithelial cells as confirmed by detection of proviral DNA, HTLV-I Tax expression and HTLV-I p19 in the latter cells. Infection was associated with induction of mRNA expression of various cytokines, chemokines and cell adhesion molecule. NF-kappaB and AP-1 were also activated in HTLV-I-infected lung epithelial cells. In vivo studies showed Tax protein in lung epithelial cells of mice bearing Tax and patients with HTLV-I-related pulmonary diseases. CONCLUSION: Our results suggest that HTLV-I infects lung epithelial cells, with subsequent production of cytokines, chemokines and cell adhesion molecules through induction of NF-kappaB and AP-1. These changes can contribute to the clinical features of HTLV-I-related pulmonary diseases.


Asunto(s)
Moléculas de Adhesión Celular/biosíntesis , Citocinas/biosíntesis , Células Epiteliales/virología , Virus Linfotrópico T Tipo 1 Humano/fisiología , Pulmón/virología , Complejo 1 de Proteína Adaptadora/metabolismo , Animales , Línea Celular , Técnicas de Cocultivo , ADN Viral/biosíntesis , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales/inmunología , Citometría de Flujo , Perfilación de la Expresión Génica , Productos del Gen tax/biosíntesis , Virus Linfotrópico T Tipo 1 Humano/inmunología , Humanos , Pulmón/inmunología , Pulmón/patología , Ratones , Ratones Transgénicos , FN-kappa B/metabolismo , Provirus/aislamiento & purificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/virología , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...