Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Medicine (Baltimore) ; 98(10): e14489, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30855435

RESUMEN

We hypothesized that circulating osteoprotegerin (OPG) and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) levels could be associated with vascular calcification, which is predominant in diabetes.The study included 71 Korean participants (36 with diabetes and 35 without diabetes), who were sub-grouped according to the results of the ankle-brachial index (ABI) and/or X-ray computed tomography scan (CT scan). Serum OPG and TRAIL levels were assayed using the respective enzyme-linked immunosorbent assay kits. Statistical significance was analyzed using Student's t test between the 2 groups or analysis of variance (ANOVA) among the 4 groups.Serum OPG was up-regulated in the participants with diabetes, with peripheral arterial disease (PAD), and/or with vascular calcification. TRAIL down-regulation was more strictly controlled than OPG up-regulation; it was significantly downregulated in the participants with PAD and vascular calcification, but not in the participants with diabetes. Serum OPG and TRAIL were regulated in the participants with femoral, popliteal, and peroneal artery calcification but not in the participants with aortic calcification.OPG up-regulation and TRAIL down-regulation were found to be associated with leg lesional vascular calcification; therefore, the average OPG/TRAIL ratio was significantly increased by 3.2-fold in the leg lesional vascular calcification group.


Asunto(s)
Osteoprotegerina/sangre , Enfermedad Arterial Periférica/sangre , Ligando Inductor de Apoptosis Relacionado con TNF/sangre , Calcificación Vascular/sangre , Enfermedades de la Aorta/sangre , Enfermedades de la Aorta/complicaciones , Enfermedades de la Aorta/diagnóstico por imagen , Biomarcadores/sangre , Complicaciones de la Diabetes/sangre , Complicaciones de la Diabetes/diagnóstico por imagen , Humanos , Pierna , Enfermedad Arterial Periférica/complicaciones , Enfermedad Arterial Periférica/diagnóstico por imagen , Calcificación Vascular/complicaciones , Calcificación Vascular/diagnóstico por imagen
3.
BMC Cancer ; 18(1): 432, 2018 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-29661248

RESUMEN

BACKGROUND: Multiple trials have attempted to demonstrate the effective induction of cell death in TRAIL-resistant cancer cells, including using a combined treatment of recombinant TRAIL and various proteasome inhibitors. These studies have yielded limited success, as the mechanism of cell death is currently unidentified. Understanding this mechanism's driving forces may facilitate the induction of cell death in TRAIL-resistant cancer cells. METHODS: Three kinds of recombinant soluble TRAIL proteins were treated into TRAIL-resistant cells and TRAIL-susceptible cells, with or without bortezomib, to compare their respective abilities to induce cell death. Recombinant TRAIL was treated with bortezomib to investigate whether this combination treatment could induce tumor regression in a mouse syngeneic tumor model. To understand the mechanism of combined treatment-induced cell death, cells were analyzed by flow cytometry and the effects of various cell death inhibitors on cell death rates were examined. RESULTS: ILz:rhTRAIL, a recombinant human TRAIL containing isoleucine zipper hexamerization domain, showed the highest cell death inducing ability both in single treatment and in combination treatment with bortezomib. In both TRAIL-resistant and TRAIL-susceptible cells treated with the combination treatment, an increase in cell death rates was dependent upon both the dose of TRAIL and its intrinsic properties. When a syngeneic mouse tumor model was treated with the combination of ILz:rhTRAIL and bortezomib, significant tumor regression was seen as a result of the effective induction of cancer cell death. The combination treatment-induced cell death was both inhibited by TRAIL blocking antibody and caspase-dependent. However, it was not inhibited by various ER stress inhibitors and autophagy inhibitors. CONCLUSIONS: The combination treatment with ILz:rhTRAIL and bortezomib was able to induce cell death in both TRAIL-susceptible and TRAIL-resistant cancer cells through the intracellular TRAIL signaling pathway. The efficiency of cell death was dependent on the properties of TRAIL under the environment provided by bortezomib. The combination treatment-induced cell death was not regulated by bortezomib-induced ER stress response or by autophagy.


Asunto(s)
Bortezomib/administración & dosificación , Proliferación Celular/efectos de los fármacos , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Animales , Apoptosis/efectos de los fármacos , Caspasas/genética , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Sinergismo Farmacológico , Humanos , Ratones , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
4.
BMB Rep ; 50(12): 647, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29279065

RESUMEN

The BMB Reports would like to correct in the ACKNOWLEDGEMENTS of BMB Rep. 49(5), 282-287 titled "Potentiation of TRAIL killing activity by multimerization through isoleucine zipper hexamerization motif."

5.
Chonnam Med J ; 53(3): 173-177, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29026704

RESUMEN

Melanoma is one of the most aggressive cancers in the world and is responsible for the majority of skin cancer deaths. Recent advances in the field of immunotherapy using active, adoptive, and antigen-specific therapeutic approaches, have generated the expectation that these technologies have the potential to improve the treatment of advanced malignancies, including melanoma. Treatment options for metastatic melanoma patients have been dramatically improved by the FDA approval of new therapeutic agents including vemurafenib, dabrafenib, and sorafenib. These kinase inhibitors have the potential to work in tandem with MEK, PI3K/AKT, and mTOR to inhibit the activity of melanoma inducing BRAF mutations. This review summarizes the effects of the new therapeutic agents against melanoma and the underlying biology of these BRAF inhibitors.

6.
Oncotarget ; 7(22): 32449-61, 2016 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-27083053

RESUMEN

The therapeutic efficacy of most anti-cancer drugs depends on their apoptosis-inducing abilities. Previously, we showed that a peptide containing the mitochondrial targeting domain (MTD) found in Noxa, a BH-3 only protein of Bcl-2 family, induces necrosis. Here, a fusion peptide of neuropilin-1 (NRP-1) targeting peptide and MTD peptide, designated tumor homing motif 17:MTD (TU17:MTD), was found to induce necrosis in cancer cells in vitro and to cause the regression of tumors when intravenously injected into mice bearing subcutaneous CT26 colorectal carcinoma tumors. The necrosis within tumor tissues was evident upon administering TU17:MTD. TU17:MTD penetrated into tumor cells by targeting to Neuropilin-1, which could be blocked by anti-NRP-1 antibody. The efficacy of TU17:MTD on tumor regression was higher than that of TU17:D(KLAKLAK)2, a fusion peptide of NRP-1 targeting peptide and a pro-apoptotic peptide. The necrotic cell death within tumor tissues was evident at day 1 after administering TU17:MTD systemically. Transplanted subcutaneous substantially reduced in size within two weeks and 5 days, respectively, with no apparent side effects. Together, these results propose that the pro-necrotic peptide MTD may present an alternative approach for development of targeted anti-cancer agents.


Asunto(s)
Neoplasias Colorrectales/tratamiento farmacológico , Neuropilina-1/metabolismo , Fragmentos de Péptidos/farmacología , Proteínas Recombinantes de Fusión/farmacología , Animales , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Terapia Molecular Dirigida , Necrosis , Neuropilina-1/genética , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Dominios Proteicos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/farmacología , Proteínas Recombinantes de Fusión/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Blood Coagul Fibrinolysis ; 27(1): 70-6, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26340462

RESUMEN

This study was designed to establish the relationship of high-sensitivity C-reactive protein (hsCRP) and mean platelet volume (MPV) with the development of adverse outcomes after percutaneous coronary intervention (PCI). hsCRP levels and MPV were analysed in 372 patients who underwent PCI, with the primary endpoint as major adverse cardiac and cerebrovascular events (MACCE): a composite of cardiac death, myocardial infarction (MI), target vessel revascularization (TVR), ischemic stroke and stent thrombosis. During the follow-up period (mean, 25.8 months), there were 21 cardiac deaths, 10 MIs including four stent thrombosis events, seven ischemic strokes and 29 TVRs. The hsCRP cut-off level was set at 0.31 mg/dl using the receiver operating characteristic curve to differentiate between the groups with and without MACCE. The MPV cut-off level was set at 8.00 fl by the receiver operating characteristic curve to differentiate between the groups with and without MACCE. A Kaplan-Meier analysis revealed that the high hsCRP group (≥0.31 mg/dl) had a significantly higher cardiac death and MACCE rate than the low hsCRP group (<0.31 mg/dl), and the high MPV group (>8.00 fl) had a significantly higher cardiac death and MACCE rate than the low MPV group (≤8.00 fl). Furthermore, the high hsCRP and MPV groups were significantly associated with an increased risk of MACCE. These results show that hsCRP and MPV are predictive markers after PCI for MACCE; they are also additively associated with a higher risk of MACCE.


Asunto(s)
Proteína C-Reactiva/metabolismo , Volúmen Plaquetario Medio/métodos , Intervención Coronaria Percutánea/métodos , Anciano , Protocolos Clínicos , Supervivencia sin Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Factores de Riesgo
8.
BMB Rep ; 49(5): 282-7, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26674343

RESUMEN

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a homo-trimeric cytotoxic ligand. Several studies have demonstrated that incorporation of artificial trimerization motifs into the TRAIL protein leads to the enhancement of biological activity. Here, we show that linkage of the isoleucine zipper hexamerization motif to the N-terminus of TRAIL, referred as ILz(6):TRAIL, leads to multimerization of its trimeric form, which has higher cytotoxic activity compared to its native state. Size exclusion chromatography of ILz(6):TRAIL revealed possible existence of various forms such as trimeric, hexameric, and multimeric (possibly containing one-, two-, and multi-units of trimeric TRAIL, respectively). Increased number of multimerized ILz(6):TRAIL units corresponded with enhanced cytotoxic activity. Further, a high degree of ILz(6):TRAIL multimerization triggered rapid signaling events such as activation of caspases, tBid generation, and chromatin condensation. Taken together, these results indicate that multimerization of TRAIL significantly enhances its cytotoxic activity. [BMB Reports 2016; 49(5): 282-287].


Asunto(s)
Isoleucina/química , Multimerización de Proteína , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Secuencias de Aminoácidos , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células HeLa , Humanos , Células Jurkat , Proteínas Recombinantes/farmacología
9.
J Pept Sci ; 19(8): 485-90, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23794461

RESUMEN

Noxa is a key player in p53-induced cell death via mitochondrial dysfunction, and the mitochondrial-targeting domain (MTD) of Noxa is responsible for the translocation of Noxa to mitochondria and for the induction of necrotic cell death. The purpose of this study was to define the minimal killing unit of MTD in vitro and in vivo. It was found that the peptides R8:MTD(10), R8:MTD(9), and R8:MTD(8) can kill various human tumor cells (HCT116, HeLa, MCF-7, BJAB), but that R8:MTD(7) abolishes the killing activity of MTD mainly because of the loss of mitochondrial targeting activity. We find it interesting that R8:MTD(8) was found to kill tumor cells but showed a limited killing activity on normal peritoneal macrophages. Furthermore, R8:MTD(10), R8:MTD(9), and R8:MTD(8) limitedly suppressed tumor growth when injected i.v. into BalB/C mice bearing CT26 cell-derived tumors. These results indicate that MTD(8) is the minimal killing unit of MTD.


Asunto(s)
Antineoplásicos/farmacología , Fragmentos de Péptidos/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/farmacología , Secuencia de Aminoácidos , Animales , Antineoplásicos/química , Apoptosis , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/fisiología , Masculino , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Trasplante de Neoplasias , Fragmentos de Péptidos/química , Señales de Clasificación de Proteína , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-bcl-2/química
10.
Biochem Biophys Res Commun ; 435(2): 234-8, 2013 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-23665015

RESUMEN

MUDENG, also known as AP5M1, was originally identified as an adaptin domain-containing gene that induced cell death in lymphoma cell lines. However, little is known of the mechanism responsible for MUDENG-mediated cell death. In this study, we investigated MUDENG changes during TRAIL-induced cell death. We found that MUDENG is rapidly processed in response to TRAIL in Jurkat and BJAB cells with time line similar to that of caspase activation. Caspase-3-mediated MUDENG cleavage was confirmed by an in vitro cleavage assay using recombinant active caspase proteins. Caspase cleavage sites (D276 and D290) were located in the adaptin domain of MUDENG, and cleaved MUDENG showed the reduced killing activity. These results suggest that the adaptin domain plays a key role in MUDENG-mediated cell death.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Activación Enzimática , Células HeLa , Humanos , Células Jurkat , Neoplasias Experimentales , Unión Proteica
11.
FEBS Lett ; 586(23): 4233-40, 2012 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-23108049

RESUMEN

p53 regulates various cellular responses through transcriptional regulation of distinct sets of target genes. Dual specificity phosphatase 6 (DUSP6) is a cytosolic phosphatase that inactivates the extracellular-signal-regulated kinase 1/2 (ERK1/2). This study demonstrates that p53 transactivates DUSP6 in human colorectal HCT116 cells to regulate ERK1/2 in p53-mediated cell death. DUSP6 is transactivated by p53 overexpression and genotoxic agents, and chromatin immunoprecipitation revealed two p53-binding sites in the DUSP6 promoter responsible for DUSP6 induction. Expression of shDUSP6 inhibited 5'-FU-induced cell death, whereas overexpression of DUSP6 increased susceptibility to 5'-FU. 5'-FU treatment dephosphorylated ERK in a DUSP6-dependent manner, resulting in destabilization of Bcl-2 and stabilization of Bad. These results provide insights on the modulatory role of p53 in the survival pathway by up-regulating DUSP6.


Asunto(s)
Fosfatasa 6 de Especificidad Dual/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/genética , Fosfatasa 6 de Especificidad Dual/genética , Fluorouracilo/farmacología , Células HCT116 , Humanos , Proteínas Proto-Oncogénicas c-bcl-2/genética , Transducción de Señal , Proteína p53 Supresora de Tumor/genética
12.
Mol Cancer Res ; 9(10): 1356-65, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21816905

RESUMEN

IFN-γ plays a critical role in tumor immunosurveillance by affecting either immune cells or tumor cells; however, IFN-mediated effects on tumor elimination are largely unknown. In this study, we showed that IFN regulatory factors (IRF) modulated by IFNs up- and downregulated Noxa expression, a prodeath BH3 protein, in various cancer cells. Inhibition of Noxa expression using short hairpin RNA in tumor cells leads to resistance against lipopolysaccharide (LPS)-induced tumor elimination, in which IFN-γ is known as a critical effecter in mice. Chromatin immunoprecipitation analysis in both CT26 cells and SP2/0 cells, sensitive and resistant to LPS-induced tumor elimination, respectively, revealed that the responsiveness of IRF1, 3, 4, and 7 in the Noxa promoter region in response to IFN-γ might be crucial in LPS-induced tumor elimination. IRF1, 3, and 7 were upregulated by IFN-γ and activated Noxa expression, leading to the death of Noxa wild-type baby mouse kidney (BMK) cells but not of Noxa-deficient BMK cells. In contrast, IRF4 acts as a repressor for Noxa expression and inhibits cell death induced by IRF1, 3, or 7. Therefore, although IFN-γ alone are not able to induce cell death in tumor cells in vitro, Noxa induction by IFN-γ, which is regulated by the balance between its activators (IRF1, 3, and 7) and its repressor (IRF4), is crucial to increasing the susceptibility of tumor cells to immune cell-mediated cytotoxicity.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/inmunología , Factores Reguladores del Interferón/antagonistas & inhibidores , Interferón gamma/genética , Proteínas Proto-Oncogénicas c-bcl-2/inmunología , Animales , Apoptosis/inmunología , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Proteínas Reguladoras de la Apoptosis/metabolismo , Muerte Celular/inmunología , Modelos Animales de Enfermedad , Células HCT116 , Humanos , Factor 1 Regulador del Interferón/inmunología , Factor 3 Regulador del Interferón/inmunología , Factor 7 Regulador del Interferón/inmunología , Factores Reguladores del Interferón/inmunología , Células Jurkat , Masculino , Ratones , Ratones Endogámicos BALB C , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis
13.
Cancer Res ; 69(21): 8356-65, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19826054

RESUMEN

DNA damage stabilizes the p53 tumor suppressor protein that determines the cell fate by either cell cycle arrest or cell death induction. Noxa, the BH3-only Bcl-2 family protein, was shown to be a key player in p53-induced cell death through the mitochondrial dysfunction; however, the molecular mechanism by which Noxa induces the mitochondrial dysfunction to cause cell death in response to genotoxic agents is largely unknown. Here, we show that the mitochondrial-targeting domain (MTD) of Noxa is a prodeath domain. Peptide containing MTD causes massive necrosis in vitro through cytosolic calcium increase; it is released from the mitochondria by opening the mitochondrial permeability transition pore. MTD peptide-induced cell death can be inhibited by calcium chelator BAPTA-AM. Moreover, MTD peptide shows the potent tumor-killing activities in mice by joining with tumor-homing motifs.


Asunto(s)
Apoptosis , Calcio/metabolismo , Mitocondrias/metabolismo , Neoplasias Experimentales/prevención & control , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Animales , Células HeLa , Humanos , Ratones , Ratones Endogámicos BALB C , Proteínas de Transporte de Membrana Mitocondrial/efectos de los fármacos , Poro de Transición de la Permeabilidad Mitocondrial , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-bcl-2/genética , Tasa de Supervivencia , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
14.
BMB Rep ; 42(8): 511-5, 2009 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-19712588

RESUMEN

Downstream of Bid (DOBI) known as Pus10, has been identified as a modulator of TRAIL-induced cell death using RNAi library screening. The crystal structure of DOBI has revealed that it is a crescent-shaped protein containing the pseudouridine synthase catalytic domain and a THUMP-containing domain. Here, we demonstrated that DOBI is expressed in various tissues such as heart and lung, and is also expressed in various tumor cells such as HeLa and A549. Although ectopic expression of DOBI does not promote TRAIL death signaling in HeLa cells, knock-down of DOBI expression using shRNA inhibited TRAIL death signaling. DOBI is cleaved into a 54 kD cleaved DOBI during cell death, and the recombinant DOBI protein can be directly cleaved by caspases-3, or -8 in vitro. Together, these data suggest that the cleaved DOBI may acquire a new function, possibly by cooperating with tBid in the mitochondrial event of cell death caused by TRAIL.


Asunto(s)
Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Hidroliasas/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Animales , Células HeLa , Humanos , Ratones , Mitocondrias Hepáticas/efectos de los fármacos , Mitocondrias Hepáticas/metabolismo , Modelos Biológicos , Proteínas Recombinantes/farmacología , Transfección
15.
FEBS Lett ; 583(14): 2349-54, 2009 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-19540835

RESUMEN

Mitochondria form reticular networks comprised of filamentous tubules and continuously move and change shape. Bcl-2 family proteins actively participate in the regulation of mitochondria fragmentation. Here, we show that human Noxa, which belongs to the BH3-only pro-apoptotic Bcl-2 family, causes mitochondrial fragmentation. We found that while the Bcl-2 homology 3 (BH3) domain of Noxa is not associated with mitochondrial fragmentation, the mitochondrial targeting domain (MTD) of Noxa is the region responsible for inducing fragmentation. Two leucine residues in MTD play a key role in the process. Furthermore, the lack of Noxa causes a significant reduction of Velcade-induced mitochondrial fragmentation. Together, these results provide novel insight into the role of Noxa in mitochondrial dynamics and cell death.


Asunto(s)
Mitocondrias/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Secuencia de Aminoácidos , Animales , Ácidos Borónicos/metabolismo , Bortezomib , Muerte Celular/fisiología , Células HeLa , Humanos , Leucina/metabolismo , Mitocondrias/ultraestructura , Datos de Secuencia Molecular , Inhibidores de Proteasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Pirazinas/metabolismo , Transducción de Señal/fisiología , Proteína X Asociada a bcl-2/metabolismo
16.
Exp Cell Res ; 315(13): 2249-55, 2009 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-19376111

RESUMEN

TRAIL is an apoptotic cell death-inducing ligand that belongs to a TNF superfamily. To identify the regulators that govern the susceptibility to TRAIL, TRAIL-resistant HeLa (TR) cells were established by repeatedly treating HeLa cells with TRAIL. Here we showed that scaffolding protein Homer1 plays a decisive role in regulating the apoptotic susceptibility to TRAIL. TR cells showing the normal susceptibility to FasL and chemotherapeutic agent etoposide expressed the lower protein levels of Homer1 than parental HeLa cells. They showed the delayed activation of caspases-8, Bid cleavage and Bax translocation to mitochondria in response to TRAIL. Reconstitution of Homer1 expression in TR cells significantly restored the susceptibility to TRAIL. In addition, knock-down of Homer1 using interfering shRNA in parental HeLa cells lost the susceptibility to TRAIL. Together, our data indicate that Homer1 plays a critical role in determining the apoptotic susceptibility to TRAIL.


Asunto(s)
Apoptosis/fisiología , Proteínas Portadoras/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Proteínas Portadoras/genética , Perfilación de la Expresión Génica , Células HeLa , Proteínas de Andamiaje Homer , Humanos , Mitocondrias/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Interferencia de ARN , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Proteína X Asociada a bcl-2/metabolismo
17.
Biochem Biophys Res Commun ; 370(3): 504-8, 2008 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-18395520

RESUMEN

A screening system comprised of a randomized hybrid-ribozyme library has previously been used to identify pro-death genes in Fas-mediated apoptosis, and short sequence information of candidate genes from this system was previously reported by Kawasaki and Taira [H. Kawasaki, K. Taira, A functional gene discovery in the Fas-mediated pathway to apoptosis by analysis of transiently expressed randomized hybrid-ribozyme libraries, Nucleic Acids Res. 30 (2002) 3609-3614]. In this study, we have cloned the full-length of the candidate's open reading frames and found that one of the candidates, referred to as MUDENG (Mu-2 related death-inducing gene), which is composed of 490 amino acids that contain the adaptin domain found in the mu2 subunit of APs related to clathrin-mediated endocytosis, is able to induce cell death by itself. Ectopic expression of MUDENG induced cell death in Jurkat T cells and HeLa cells. In addition, when MUDENG expression was evaluated by immnuohistochemical staining, it was found in most tissues, including the intestine and testis. Furthermore, MUDENG appears to be evolutionary conserved from mammals to amphibians, suggesting that it may have a common role in cell death. Taken together, these results suggest that MUDENG is likely to play an important role in cell death in various tissues.


Asunto(s)
Apoptosis , Linfocitos T Citotóxicos/inmunología , Secuencia de Aminoácidos , Animales , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/análisis , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Bovinos , Clonación Molecular , Perros , Evolución Molecular , Células HeLa , Humanos , Células Jurkat , Ratones , Datos de Secuencia Molecular , Ratas
18.
J Biol Chem ; 279(11): 9857-66, 2004 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-14688259

RESUMEN

A human 8-oxoguanine-DNA glycosylase (hOGG1) is the main enzyme that repairs 8-oxoG, which is a critical mutagenic lesion. There is a great deal of interest in the up- or down-regulation of OGG1 expression after DNA damage. In this study, we investigated the effect of a DNA-alkylating agent, methylmethane sulfonate (MMS), on hOGG1 expression level and found that MMS treatment resulted in an increase in the functional hOGG1 expression in HCT116 cells. A region between -121 and -61 of the hOGG1 promoter was found to be crucial for this induction by MMS. Site-directed mutations of the two inverted CCAAT motifs substantially abrogated the induction of the hOGG1 promoter as a result of MMS treatment. In addition, the NF-YA protein (binding to the inverted CCAAT box) was induced after exposing cells to MMS. Moreover, gel shift and supershift analyses with the nuclear extracts prepared from HCT116 cells identified NF-YA as the transcription factor interacting with the inverted CCAAT box. Mutations of the inverted CCAAT box either prevented the binding of this factor or abolished its activation as a result of MMS treatment. Finally, this study showed that hOGG1-expressing HCT116 cells exhibited increased hOGG1 repair activity and resistance to MMS. Overall, these results demonstrate that MMS can up-regulate hOGG1 expression through the induction of the transcription factor, NF-YA, and increased transcription level of the hOGG1 gene correlates with an increase in enzyme activity providing functional protection from MMS.


Asunto(s)
Alquilantes/farmacología , Factor de Unión a CCAAT/fisiología , ADN Glicosilasas/metabolismo , ADN/química , Guanosina/análogos & derivados , Metilmetanosulfonato/farmacología , Factores de Transcripción/fisiología , Adyuvantes Inmunológicos/farmacología , Secuencias de Aminoácidos , Antineoplásicos Alquilantes/farmacología , Western Blotting , Factor de Unión a CCAAT/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Supervivencia Celular , Reparación del ADN , Relación Dosis-Respuesta a Droga , Regulación Neoplásica de la Expresión Génica , Guanina/análogos & derivados , Guanina/química , Guanosina/metabolismo , Humanos , Peróxido de Hidrógeno/farmacología , Luciferasas/metabolismo , Mutagénesis Sitio-Dirigida , Mutación , Plásmidos/metabolismo , Unión Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Factores de Transcripción/metabolismo , Transfección , Regulación hacia Arriba
19.
Biochim Biophys Acta ; 1573(1): 33-8, 2002 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-12383939

RESUMEN

Metallothionein (MT)-III is a member of a brain-specific MT family, in contrast to MT-I and MT-II that are found in most tissues and are implicated in metal ion homeostasis and as an antioxidant. To investigate the defensive role of MT-III in terms of hydroxyl radical-induced DNA damage, we used purified human MT-III. DNA damage was detected by single-strand breaks of plasmid DNA and deoxyribose degradation. In this study, we show that MT-III is able to protect against the DNA damage induced by ferric ion-nitrilotriacetic acid and H(2)O(2), and that this protective effect is inhibited by the alkylation of the sulfhydryl groups of MT-III by treatment with EDTA and N-ethylmaleimide. MT-III was also able to efficiently remove the superoxide anion, which was generated from the xanthine/xanthine oxidase system. These results strongly suggest that MT-III is involved in the protection of reactive oxygen species-induced DNA damage, probably via direct interaction with reactive oxygen species, and that MT-III acts as a neuroprotective agent.


Asunto(s)
Daño del ADN , Etidio/análogos & derivados , Radical Hidroxilo/antagonistas & inhibidores , Proteínas del Tejido Nervioso/farmacología , Fármacos Neuroprotectores/farmacología , Ácido Edético , Etilmaleimida , Fluorescencia , Depuradores de Radicales Libres/farmacología , Humanos , Peróxido de Hidrógeno , Metalotioneína/farmacología , Metalotioneína 3 , Ácido Nitrilotriacético , Plásmidos , Proteínas Recombinantes/farmacología
20.
Pharmacol Res ; 46(3): 221-7, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12220964

RESUMEN

The protective effects of 18beta-glycyrrhetinic acid (GA), the aglycone of glycyrrhizin (GL) derived from licorice, on carbon tetrachloride-induced hepatotoxicity and the possible mechanisms involved in this protection were investigated in mice. Pretreatment with GA prior to the administration of carbon tetrachloride significantly prevented an increase in serum alanine, aspartate aminotransferase activity and hepatic lipid peroxidation in a dose-dependent manner. In addition, pretreatment with GA also significantly prevented the depletion of glutathione (GSH) content in the livers of carbon tetrachloride-intoxicated mice. However, reduced hepatic GSH levels and glutathione-S-transferase activities were unaffected by treatment with GA alone. Carbon tetrachloride-induced hepatotoxicity was also prevented, as indicated by a liver histopathologic study. The effects of GA on the cytochrome P450 (P450) 2E1, the major isozyme involved in carbon tetrachloride bioactivation, were also investigated. Treatment of mice with GA resulted in a significant decrease of the P450 2E1-dependent hydroxylation of p-nitrophenol and aniline in a dose-dependent manner. Consistent with these observations, the P450 2E1 expressions were also decreased, as determined by immunoblot analysis. GA also showed antioxidant effects upon FeCl(2)-ascorbate-induced lipid peroxidation in mice liver homogenate and upon superoxide radical scavenging activity. These results show that protective effects of GA against the carbon tetrachloride-induced hepatotoxicity may be due to its ability to block the bioactivation of carbon tetrachloride, primarily by inhibiting the expression and activity of P450 2E1, and its free radical scavenging effects.


Asunto(s)
Antiinflamatorios/farmacología , Inhibidores del Citocromo P-450 CYP2E1 , Ácido Glicirretínico/farmacología , Fallo Hepático Agudo/prevención & control , Administración Tópica , Alanina/sangre , Alanina/efectos de los fármacos , Animales , Ácido Ascórbico/farmacología , Aspartato Aminotransferasas/sangre , Aspartato Aminotransferasas/efectos de los fármacos , Tetracloruro de Carbono/administración & dosificación , Cloruros , Citocromo P-450 CYP2E1/biosíntesis , Citocromo P-450 CYP2E1/efectos de los fármacos , Compuestos Férricos/farmacología , Depuradores de Radicales Libres/metabolismo , Glutatión/metabolismo , Ácido Glicirretínico/análogos & derivados , Glycyrrhiza , Ácido Glicirrínico/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Fallo Hepático Agudo/inducido químicamente , Masculino , Ratones , Ratones Endogámicos ICR , Fitoterapia , Extractos Vegetales/farmacología , Superóxidos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...