Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Biochem Biophys Res Commun ; 416(1-2): 222-5, 2011 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-22120628

RESUMEN

Gliomas are the most common brain tumors in adults and account for more than half of all brain tumors. Despite intensive clinical investigations, average survival for the patients harboring the malignancy has not been significantly improved. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), shown to have potent and cancer-selective killing activity, has drawn considerable attention as a promising anti-cancer therapy. In an attempt to develop TRAIL as an anti-cancer therapy for gliomas, tumor suppressor activity of TRAIL was assessed using human glioma cell lines such as U373MG, U343MG, U87MG and LN18. U343MG, U87MG and LN18 cells were susceptible to TRAIL; however, U373MG cells were completely refractory to TRAIL. Resistance to the applied therapies is a key issue in cancer treatment; thus, various combination treatments were evaluated using U373MG cells to identify a better regimen. Unlike Doxorubicin, Etoposide, Actinomycin D and Wortmannin, a proteasome inhibitor MG132 significantly enhanced TRAIL-induced apoptosis. Similarly, other proteasome inhibitors, including Lactacystin, Proteasome inhibitor I and Velcade (Bortezomib), also enhanced apoptotic activity of TRAIL. Among these proteasome inhibitors, Velcade, the only approved drug, was as effective as MG132 in enhancing TRAIL-induced apoptosis. Both Velcade and MG132 increased the protein levels of DR5, a TRAIL receptor known to be up-regulated by p53, in U373MG cells where p53 is mutated. Our data indicate that proteasome inhibitors up-regulate DR5 in a p53-independent manner and a combination therapy comprising TRAIL and Velcade become a better treatment regimen for gliomas.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica , Ácidos Borónicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Inhibidores de Cisteína Proteinasa/uso terapéutico , Glioma/tratamiento farmacológico , Pirazinas/uso terapéutico , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/biosíntesis , Ligando Inductor de Apoptosis Relacionado con TNF/uso terapéutico , Apoptosis/efectos de los fármacos , Bortezomib , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Glioma/patología , Humanos , Leupeptinas/uso terapéutico , Inhibidores de Proteasoma , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba
2.
Biochem Biophys Res Commun ; 396(4): 861-5, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20451494

RESUMEN

Gene silencing by RNA interference (RNAi) using short interfering RNAs (siRNAs) or short hairpin RNAs (shRNAs) has become a valuable tool for evaluating the target gene function. Here, we report an approach for silencing multiple target genes simultaneously by expressing one single transcript encoding different target shRNAs. We first constructed the cytomegalovirus (CMV) promoter-driven expression vectors, each of which expresses microRNA mir-30-mimicked shRNA specifically targeting X-chromosome-linked inhibitor of apoptosis protein (XIAP), Akt, or Bcl-2. Adenovirus harbouring each shRNA expression cassette silenced corresponding target gene expression. Using these mono-cistronic shRNA cassettes, we again constructed the CMV promoter-driven expression vector, into which multi-cistronic shRNAs for XIAP, Akt and Bcl-2 in order were cloned. Adenovirus delivering this multi-cistronic expression cassette silenced each of the target genes as effectively as adenovirus containing individual shRNA did. Our data indicate that single promoter-driven multi-cistronic shRNAs effectively silence multiple target genes. Our approach provides a new smart tool for silencing multiple target genes and will potentially serve as an RNAi-based tailored therapy requiring suppression of target gene expression.


Asunto(s)
Técnicas de Silenciamiento del Gen/métodos , Interferencia de ARN , ARN Interferente Pequeño/genética , Apoptosis/genética , Línea Celular , Citomegalovirus/genética , Vectores Genéticos , Humanos , Conformación de Ácido Nucleico , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , ARN Interferente Pequeño/química , Transcripción Genética , Proteína Inhibidora de la Apoptosis Ligada a X/genética
3.
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
4.
BMB Rep ; 53(11): 565-575, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32958121

RESUMEN

Gene therapy is emerging as a treatment option for inherited genetic diseases. The success of this treatment approach greatly depends upon gene delivery vectors. Researchers have attempted to harness the potential of viral vectors for gene therapy applications over many decades. Among the viral vectors available, gutless adenovirus (GLAd) has been recognized as one of the most promising vectors for in vivo gene delivery. GLAd is constructed by deleting all the viral genes from an adenovirus. Owing to this structural feature, the production of GLAd requires a helper that supplies viral proteins in trans. Conventionally, the helper is an adenovirus. Although the helper adenovirus efficiently provides helper functions, it remains as an unavoidable contaminant and also generates replicationcompetent adenovirus (RCA) during the production of GLAd. These two undesirable contaminants have raised safety concerns and hindered the clinical applications of GLAd. Recently, we developed helper virus-free gutless adenovirus (HF-GLAd), a new version of GLAd, which is produced by a helper plasmid instead of a helper adenovirus. Utilization of this helper plasmid eliminated the helper adenovirus and RCA contamination in the production of GLAd. HF-GLAd, devoid of helper adenovirus and RCA contaminants, will facilitate its clinical applications. In this review, we discuss the characteristics of adenoviruses, the evolution and production of adenoviral vectors, and the unique features of HF-GLAd as a new platform for gene therapy. Furthermore, we highlight the potential applications of HF-GLAd as a gene delivery vector for the treatment of various inherited genetic diseases. [BMB Reports 2020; 53(11): 565-575].


Asunto(s)
Adenoviridae/genética , Adenoviridae/metabolismo , Terapia Genética/métodos , Línea Celular , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Virus Helper/genética , Virus Helper/metabolismo , Humanos , Integrasas/genética , Plásmidos/genética , Proteínas Virales/genética
5.
Biochem Biophys Res Commun ; 378(3): 640-4, 2009 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-19056351

RESUMEN

The increasing importance of adenoviral vectors for gene therapy clinical trials necessitates the development of processes suitable for large-scale and commercial production of adenovirus. Here, we evaluated a novel purification process combining an anion-exchange chromatography and an immobilized metal affinity membrane chromatography for the purification of recombinant adenovirus. Adenovirus was initially purified from clarified infectious lysate by anion-exchange chromatography using Q Sepharose XL resin and further polished using a Sartobind IDA membrane unit charged with Zn(2+) ions as affinity ligands. The metal affinity membrane chromatography efficiently removed residual host cell impurities that co-eluted with adenovirus during the previous anion-exchange chromatography step. The metal affinity membrane chromatography also separated defective adenovirus particles from the infectious adenovirus fraction. Furthermore, the metal affinity membrane chromatography showed an improved yield, when compared with a conventional bead-based metal affinity chromatography. The purity and specific activity of the adenovirus prepared using this two-step chromatography was comparable to those of adenovirus produced by the conventional CsCl density centrifugation. Therefore, our data provide an improved method for the purification of adenoviral vectors for clinical applications.


Asunto(s)
Adenoviridae/aislamiento & purificación , Cromatografía de Afinidad/métodos , Cromatografía por Intercambio Iónico/métodos , Vectores Genéticos/aislamiento & purificación , Zinc/química , Adenoviridae/genética , Línea Celular , Glicina/química , Humanos , Membranas/química , Recombinación Genética
6.
Biochem Biophys Res Commun ; 381(4): 513-7, 2009 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-19233125

RESUMEN

The present study investigates the effect of low oxygen concentrations on thapsigargin-induced apoptosis and reactive oxygen species (ROS)-related signaling in articular chondrocytes. Chondrocytes were obtained from normal canine knee cartilage and were treated with different concentrations of thapsigargin for 24h under normoxic (21% oxygen tension) or hypoxic (1% oxygen tension) conditions. The cells treated with thapsigargin under normoxic conditions showed a dose-dependent induction of apoptosis. However, the cellular changes and apoptotic events that occurred following thapsigargin treatment, were completely inhibited by hypoxia, including loss of mitochondrial transmembrane potential (MTP), ROS generation and JNK phosphorylation. Moreover, the cells exposed to hypoxic conditions showed increased expression of the anti-apoptotic proteins xIAP-2 and Bcl-2. We demonstrate that hypoxia inhibited thapsigargin-induced apoptosis in chondrocytes by regulating ROS-related signaling and the expression of anti-apoptotic proteins. We propose that maintaining hypoxic conditions in articular cartilage may be required for the prevention of chondrocyte and cartilage diseases such as arthritis.


Asunto(s)
Apoptosis , Cartílago Articular/metabolismo , Condrocitos/metabolismo , Inhibidores Enzimáticos/farmacología , Oxígeno/metabolismo , Tapsigargina/farmacología , Animales , Cartílago Articular/citología , Cartílago Articular/efectos de los fármacos , Hipoxia de la Célula , Células Cultivadas , Condrocitos/efectos de los fármacos , Perros , MAP Quinasa Quinasa 4/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Fosforilación , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo
7.
Hum Gene Ther ; 30(12): 1531-1546, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31547718

RESUMEN

Preclinical studies showed that tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) therapy is safe and effective to combat cancers, but clinical outcomes have been less than optimal due to short half-life of TRAIL protein, insufficient induction of apoptosis, and TRAIL resistance displayed in many tumors. In this study, we explored co-delivery of a TRAIL expressing plasmid (pTRAIL) and complementary small interfering RNAs (siRNAs) (silencing Bcl2-like 12 [BCL2L12] and superoxide dismutase 1 [SOD1]) to improve the response of breast cancer cells against TRAIL therapy. It is desirable to co-deliver the pDNA along with siRNA using a single delivery agent, but this is challenging given different structures of long/flexible pDNA and short/rigid siRNA. Toward this goal, we identified an aliphatic lipid-grafted low-molecular weight polyethylenimine (PEI) that accommodated both pDNA and siRNA in a single complex. The co-delivery of pTRAIL with BCL2L12- or SOD1-specific siRNAs resulted more significant cell death in different breast cancer cells compared with separate delivery without affecting nonmalignant cells viability. Ternary complexes of lipopolymer with pTRAIL and BCL2L12 siRNA significantly retarded the growth of breast cancer xenografts in mice. The enhanced anticancer activity was attributed to increased in situ secretion of TRAIL and sensitization of breast cancer cells against TRAIL by the co-delivered siRNAs. The lipid-grafted PEIs capable of co-delivering multiple types of nucleic acids can serve as powerful carriers for more effective complementary therapeutics. Graphical Abstract [Figure: see text].


Asunto(s)
Neoplasias de la Mama/genética , Terapia Genética , Proteínas Musculares/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Superóxido Dismutasa-1/genética , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Animales , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Silenciador del Gen/efectos de los fármacos , Técnicas de Transferencia de Gen , Xenoinjertos , Humanos , Ratones , Proteínas Musculares/antagonistas & inhibidores , Plásmidos/genética , Plásmidos/farmacología , Polietileneimina/química , Polietileneimina/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Superóxido Dismutasa-1/antagonistas & inhibidores , Ligando Inductor de Apoptosis Relacionado con TNF/antagonistas & inhibidores
8.
Exp Mol Med ; 51(10): 1-18, 2019 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-31659156

RESUMEN

Gene therapy is emerging as an effective treatment option for various inherited genetic diseases. Gutless adenovirus (GLAd), also known as helper-dependent adenovirus (HDAd), has many notable characteristics as a gene delivery vector for this particular type of gene therapy, including broad tropism, high infectivity, a large transgene cargo capacity, and an absence of integration into the host genome. Additionally, GLAd ensures long-term transgene expression in host organisms owing to its minimal immunogenicity, since it was constructed following the deletion of all the genes from an adenovirus. However, the clinical use of GLAd for the treatment of inherited genetic diseases has been hampered by unavoidable contamination of the highly immunogenic adenovirus used as a helper for GLAd production. Here, we report the production of GLAd in the absence of a helper adenovirus, which was achieved with a helper plasmid instead. Utilizing this helper plasmid, we successfully produced large quantities of recombinant GLAd. Importantly, our helper plasmid-based system exclusively produced recombinant GLAd with no generation of helper plasmid-originating adenovirus and replication-competent adenovirus (RCA). The recombinant GLAd that was produced efficiently delivered transgenes regardless of their size and exhibited therapeutic potential for Huntington's disease (HD) and Duchenne muscular dystrophy (DMD). Our data indicate that our helper plasmid-based GLAd production system could become a new platform for GLAd-based gene therapy.


Asunto(s)
Adenoviridae/genética , Terapia Genética , Vectores Genéticos/genética , Transgenes/genética , Línea Celular , Vectores Genéticos/uso terapéutico , Genoma Humano/genética , Humanos , Enfermedad de Huntington/genética , Enfermedad de Huntington/terapia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Plásmidos/genética
9.
Biochem Biophys Res Commun ; 377(2): 508-511, 2008 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-18929542

RESUMEN

Under normal cell physiology, a balance between cell survival and apoptosis is crucial for homeostasis. Many studies have demonstrated that apoptosis is modulated by cell survival stimuli. Active Akt, a common mediator of cell survival signals, has been shown to inhibit apoptosis by attenuating activity of pro-apoptotic factors Bad and caspase-9. However, the anti-apoptotic mechanisms mediated by various cell survival signals are poorly understood. Human prostate cancer LNCaP cells, known to contain constitutively activated Akt as a result of a frame-shift mutation in PTEN, an inhibitor of PI-3K/Akt pathway, were observed to be completely resistant to TRAIL-induced apoptosis. In agreement with the known action of Akt, blockade of the PI-3K/Akt pathway rendered LNCaP cells highly susceptible to TRAIL. Importantly, active PI-3K/Akt prevented processing/activation of caspase-3, a phenomenon associated with the function of inhibitor of apoptosis proteins (IAPs). In fact, inhibition of PI-3K activity using Wortmannin significantly decreased the protein levels of IAPs, concomitantly promoting processing/activation of caspase-3 and TRAIL-induced apoptosis. My data indicate that in addition to blocking Bad and caspase-9 through Akt, PI-3K also inhibits caspase-3 through up-regulating IAPs, thereby attenuates apoptosis.


Asunto(s)
Apoptosis , Proteínas Inhibidoras de la Apoptosis/biosíntesis , Fosfatidilinositol 3-Quinasas/metabolismo , Androstadienos/farmacología , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Regulación hacia Arriba , Wortmanina , Proteína Letal Asociada a bcl/metabolismo
10.
Biochem Biophys Res Commun ; 375(4): 684-8, 2008 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-18761322

RESUMEN

Tumor hypoxia, which is caused by the rapid proliferation of tumor cells and aberrant vasculature in tumors, results in inadequate supplies of oxygen and nutrients to tumor cells. Paradoxically, these unfavorable growth conditions benefit tumor cell survival, although the mechanism is poorly understood. We have demonstrated for the first time that hypoxia inhibits TRAIL-induced apoptosis by blocking translocation of Bax from cytosol to the mitochondria in tumor cells. However, it is largely unknown how hypoxia-inhibited Bax translocation attenuates TRAIL-induced apoptosis. Here, we demonstrate that despite its inhibitory activity in TRAIL-induced apoptosis, hypoxia does not affect TRAIL-triggered proximal apoptotic signaling events, including caspase-8 activation and Bid cleavage. Instead, hypoxia inhibited processing of caspase-3, leading to incomplete activation of the caspase. Importantly, hypoxia-blocked translocation of Bax to the mitochondria significantly inhibited releasing the mitochondrial factors, such as cytochrome c and Smac/DIABLO, to the cytosol in response to TRAIL. It is well-known that complete processing/activation of caspase-3 requires Smac/DIABLO released from mitochondria. Therefore, our data indicate that an engagement of the apoptotic mitochondrial events leading to caspase-3 activation is blocked by hypoxia. Our data shed new light on understanding of the apoptotic signal transduction and targets regulated by tumor hypoxia.


Asunto(s)
Apoptosis , Caspasa 3/metabolismo , Neoplasias/enzimología , Oxígeno/metabolismo , Proteínas Reguladoras de la Apoptosis , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Caspasa 8/metabolismo , Hipoxia de la Célula , Activación Enzimática , Células HCT116 , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Neoplasias/patología , Transporte de Proteínas , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Proteína X Asociada a bcl-2/metabolismo
11.
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
12.
Mol Cell Biol ; 22(3): 724-36, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11784850

RESUMEN

Among the tumor necrosis factor (TNF) family of cytokines, FasL and TNF-related apoptosis-inducing ligand (TRAIL) are known to induce cell death via caspase activation. Recently, other biological functions of these death ligands have been postulated in vitro and in vivo. It was previously shown that Fas ligation induces chemokine expression in human glioma cells. In this study, we investigated whether the TRAIL-DR5 system transduces signals similar to those induced by other TNF family ligands and receptors. To address this issue, two human glioma cell lines, CRT-MG and U87-MG, were used, and an agonistic antibody against DR5 (TRA-8) and human recombinant TRAIL were used to ligate DR5. We demonstrate that DR5 ligation by either TRAIL or TRA-8 induces two functional outcomes, apoptosis and expression of the chemokine interleukin-8 (IL-8); the nonspecific caspase inhibitor Boc-D-Fmk blocks both TRAIL-mediated cell death and IL-8 production; the caspase 3-specific inhibitor z-DEVD-Fmk suppresses TRAIL-mediated apoptosis but not IL-8 induction; caspase 1- and 8-specific inhibitors block both TRAIL-mediated cell death and IL-8 production; and DR5 ligation by TRAIL mediates AP-1 and NF-kappaB activation, which can be inhibited by caspase 1- and 8-specific inhibitors. These findings collectively indicate that DR5 ligation on human glioma cells leads to apoptosis and that the activation of AP-1 and NF-kappaB leads to the induction of IL-8 expression; these responses are dependent on caspase activation. Therefore, the TRAIL-DR5 system has a role not only as an inducer of apoptotic cell death but also as a transducer for proinflammatory and angiogenic signals in human brain tumors.


Asunto(s)
Astrocitoma/patología , Astrocitoma/fisiopatología , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/fisiopatología , Interleucina-8/biosíntesis , Glicoproteínas de Membrana/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Anticuerpos Monoclonales/farmacología , Apoptosis , Proteínas Reguladoras de la Apoptosis , Astrocitoma/genética , Neoplasias Encefálicas/genética , Caspasa 3 , Caspasas/metabolismo , Activación Enzimática , Humanos , Interleucina-8/genética , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/genética , Modelos Biológicos , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF , Receptores del Factor de Necrosis Tumoral/antagonistas & inhibidores , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/fisiología , Transducción de Señal , Ligando Inductor de Apoptosis Relacionado con TNF , Transfección , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/genética
13.
Oncol Rep ; 18(3): 523-9, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17671697

RESUMEN

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF family and a potent inducer of apoptosis. Gemcitabine, chemotherapeutic agent is well known to be involved in synergistic cytotoxic effect with TRAIL. But the mechanism of this synergistic effect is still unclear. To examine the effects and synergistic mechanism of gemcitabine on TRAIL-induced apoptosis, A549, HCT116 and SNU638 cells were pretreated with gemcitabine and treated with TRAIL protein. Gemcitabine significantly enhanced A549 cell death induced by TRAIL, but it was inhibited by the pan-caspase inhibitor, Z-VAD-fmk. The combined treatment of both induced the activation of caspase-8 and reduced the mitochondrial transmembrane potential (MTP) and also translocated Bax protein and released the cytochrome-c in A549 cells when compared with that of negative control. In addition, the gemcitabine pretreatment up-regulated DR-5 and p53 protein expression in a time-dependent manner, which suggests the possible involvement of the p53 protein as a transcriptional factor for DR-5 up-regulation. Thus, we report our findings that gemcitabine enhanced the TRAIL-induced apoptosis and the apoptotic signals are mediated by DR-5-dependent pathway and mitochondrial pathway. Taken together, gemcitabine enhanced TRAIL-induced apoptosis via DR-5 up-regulation and lowering MTP, and suggest that gemcitabine may be used as a successful chemotherapeutic agent for ligand type tumor therapy combined with TRAIL.


Asunto(s)
Apoptosis/fisiología , Desoxicitidina/análogos & derivados , Membranas Mitocondriales/fisiología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Antimetabolitos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Neoplasias del Colon , Fragmentación del ADN , Desoxicitidina/farmacología , Neoplasias Gastrointestinales , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pulmonares , Potenciales de la Membrana/efectos de los fármacos , Membranas Mitocondriales/efectos de los fármacos , Gemcitabina
14.
Protein J ; 26(2): 107-16, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17203394

RESUMEN

We have developed a novel reporter system involving a yeast two-hybrid assay, which utilizes the reconstitution of the split EGFP reporter in order to characterize the relevant protein-protein interactions. To our knowledge, this study represents the first application of the split EGFP system as a read-out in a yeast two-hybrid assay. In comparison with the existing two-hybrid system, the bait and prey vectors were improved with regard to the reporter and the replication control element. As a result, the reconstituted EGFP has been observed to evidence a restored fluorescence upon protein-protein interactions in yeast, thereby allowing for the characterization of its interactor. The use of a split EGFP reporter has some salient advantages. Firstly, no substrates are required for the production of fluorescence. Secondly, low copy number plasmids may help to solve the protein toxicity problem, via the reduction of expression. Thirdly, this technique may prove useful in overcoming the autoactivation problem, due to the fact that the read-out of the yeast two-hybrid system is transcription-independent. Collectively, our results showed that the split EGFP reporter system might potentially be applied in yeast two-hybrid assays for the high-throughput screening of protein-protein interactions, with a simple and direct fluorescent read-out.


Asunto(s)
Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Técnicas del Sistema de Dos Híbridos , Escherichia coli/genética , Proteínas Recombinantes de Fusión/genética
15.
J Microbiol Biotechnol ; 17(4): 638-43, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18051276

RESUMEN

The full encoding sequence for human type II hexokinase (HXK II) was cloned into the E. coli expression vector pET 21b and expressed as a C-terminally hexahistidine-tagged protein in the BL21 (DE3) strain. The IPTG-induced HXK II approximately accounted for 17% of the total E. coli proteins, and 81% of HXK II(6xHis) existed in inclusion bodies. To improve the production of soluble recombinant HXK II protein, in the functionally active form, we used low temperature, and the osmotic stress expression method. When expressed at 18 degrees C, about 83% of HXK II(6xHis) existed in the soluble fraction, which amounted to a 4.1-fold yield over that expressed at 37 degrees C. The soluble form of HXK II(6xHis) was also highly produced in the presence of 1 M sorbitol under the standard condition (37 degrees C), which indicated that temperature downshift and low water potentials were required to improve the yield of active recombinant HXK II protein. The expressed protein was purified by metal chelate affinity chromatography performed in an IDA Excellose column charged with Ni2+ ions, resulting in about 40 mg recombinant HXK II protein obtained with purity over 89% from 5 l of E. coli culture. The identity of HXK II(6xHis) was confirmed by Western blotting analysis. Taken together, using the stress-governed expression described in this study, human active HXK II can be purified in sufficient amounts for biochemical and biomedical studies.


Asunto(s)
Escherichia coli/genética , Hexoquinasa/genética , Proteínas Recombinantes/biosíntesis , Vectores Genéticos , Hexoquinasa/aislamiento & purificación , Humanos , Presión Osmótica , Proteínas Recombinantes/aislamiento & purificación , Solubilidad , Temperatura
16.
Oncotarget ; 8(11): 18095-18105, 2017 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-28178647

RESUMEN

Lung cancer, especially lung adenocarcinoma, is one of the main causes of death worldwide. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a primary anticancer agent and a member of the tumor necrosis factor family that selectively induces apoptosis in various tumor cells, but not in normal cells. Combination chemotherapy can be used for treating specific cancer types even at progressive stages. In the present study, we observed that 5-fluorouracil, which exerts anticancer effects by inhibiting tumor cell proliferation, enhanced TRAIL-induced apoptosis of TRAIL-resistant human adenocarcinoma A549 cells. Interestingly, 5-fluorouracil treatment markedly increased Bax and p53 levels and 5-fluorouracil and TRAIL cotreatment increased Ac-cas3 and Ac-cas8 levels compared with those in control cells. Taken together, the present study demonstrated that 5-fluorouracil enhances TRAIL-induced apoptosis in TRAIL-resistant lung adenocarcinoma cells by activating Bax and p53, and also suggest that TRAIL and 5-fluorouracil cotreatment can be used as an adequate therapeutic strategy for TRAIL-resistant human cancers.


Asunto(s)
Adenocarcinoma/patología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Fluorouracilo/farmacología , Neoplasias Pulmonares/patología , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Adenocarcinoma/metabolismo , Adenocarcinoma del Pulmón , Apoptosis/efectos de los fármacos , Western Blotting , Supervivencia Celular/efectos de los fármacos , Humanos , Inmunohistoquímica , Neoplasias Pulmonares/metabolismo , Transducción de Señal/efectos de los fármacos , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Proteína p53 Supresora de Tumor/biosíntesis , Proteína X Asociada a bcl-2/metabolismo
17.
Cancer Res ; 64(12): 4078-81, 2004 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15205314

RESUMEN

The hypoxic environment in solid tumors results from oxygen consumption by rapid proliferation of tumor cells. Hypoxia has been shown to facilitate the survival of tumor cells and to be a cause of malignant transformation. Hypoxia also is well known to attenuate the therapeutic activity of various therapies in cancer management. These observations indicate that hypoxia plays a critical role in tumor biology. However, little is known about the effects of hypoxia on apoptosis, especially on apoptosis induced by tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), a potent apoptosis inducer that has been shown to specifically limit tumor growth without damaging normal cells and tissues in vivo. To address the effects of hypoxia on TRAIL-induced apoptosis, HCT116 human colon carcinoma cells were exposed to hypoxic or normoxic conditions and treated with soluble TRAIL protein. Hypoxia dramatically inhibited TRAIL-induced apoptosis in HCT116 cells, which are highly susceptible to TRAIL in normoxia. Hypoxia increased antiapoptotic Bcl-2 family member proteins and inhibitors of apoptosis proteins. Interestingly, these hypoxia-increased antiapoptotic molecules were decreased by TRAIL treatment to the levels lower than those of the untreated conditions, suggesting that hypoxia inhibits TRAIL-induced apoptosis via other mechanisms rather than up-regulation of these antiapoptotic molecules. Additional characterization revealed that hypoxia significantly inhibits TRAIL-induced translocation of Bax from the cytosol to the mitochondria in HCT116 and A549 cells, with the concomitant inhibition of cytochrome c release from the mitochondria. Bax-deficient HCT116 cells were completely resistant to TRAIL regardless of oxygen content, demonstrating a pivotal role of Bax in TRAIL-induced apoptotic signaling. Thus, our data indicate that hypoxia inhibits TRAIL-induced apoptosis by blocking Bax translocation to the mitochondria, thereby converting cells to a Bax-deficient state.


Asunto(s)
Apoptosis/efectos de los fármacos , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/farmacología , Proteínas Proto-Oncogénicas c-bcl-2 , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/farmacología , Antineoplásicos/antagonistas & inhibidores , Antineoplásicos/farmacología , Apoptosis/fisiología , Proteínas Reguladoras de la Apoptosis , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Células HCT116 , Humanos , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Recombinantes/farmacología , Fracciones Subcelulares/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF , Proteína X Asociada a bcl-2
18.
Cancer Sci ; 94(12): 1066-73, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14662022

RESUMEN

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a potent inducer of apoptosis in some, but not all cancer cells. To assess the regulation of TRAIL-resistance in the human gastric cancer cells, we examined TRAIL sensitivity, TRAIL receptor expression, and intracellular signaling events induced by TRAIL. All the gastric cancer cell lines tested were susceptible to TRAIL to some extent, except for SNU-216 cell line, which was completely resistant. TRAIL receptor expression was not related to the TRAIL-sensitivity. Of the cell lines tested, SNU-216 showed the highest level of constitutively active Akt and the short form of FLICE inhibitory protein (FLIP(S)). Treatment with the phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002 or with the protein synthesis inhibitor cycloheximide induced a suppression of constitutive Akt activation in SNU-216 cells and a concomitant decrease in the expression of FLIP(S). The reduction of Akt activity by LY294002 affected the transcriptional level of FLIP(S), but not the mRNA stability. As a result, LY294002 or cycloheximide significantly enhanced TRAIL-induced apoptosis. Moreover, the overexpression of constitutively active Akt in the TRAIL-sensitive cell line, SNU-668, rendered the cell line resistant to TRAIL. In addition, infection of the same cell line with retrovirus expressing FLIP(S) completely inhibited TRAIL-induced apoptosis by blocking the activation of caspase-8. Therefore, our results suggest that Akt activity promotes human gastric cancer cell survival against TRAIL-induced apoptosis via upregulation of FLIP(S), and that the cytotoxic effect of TRAIL can be enhanced by modulating the Akt/FLIP(S) pathway in human gastric cancers.


Asunto(s)
Adenocarcinoma/metabolismo , Apoptosis/fisiología , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Neoplasias Gástricas/metabolismo , Adenocarcinoma/patología , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Línea Celular Tumoral , Cromonas , Cicloheximida/farmacología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Inhibidores Enzimáticos/farmacología , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Expresión Génica/efectos de los fármacos , Humanos , Immunoblotting , Morfolinas , Inhibidores de la Síntesis de la Proteína/farmacología , ARN Mensajero/análisis , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Receptores del Factor de Necrosis Tumoral/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Gástricas/patología , Regulación hacia Arriba
19.
Mol Cells ; 15(3): 283-93, 2003 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-12872982

RESUMEN

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a membrane-bound cytokine molecule that belongs to the family of tumor necrosis factor (TNF). TRAIL has been shown to be a potent apoptosis inducer in a wide variety of cancer cells in vitro and to limit tumor growth efficiently in vivo without damaging normal tissues. These features have focused considerable attention on TRAIL as a potential therapeutic agent to treat human cancers. Recent data also suggest the implication of TRAIL in a natural defense mechanism since its abrogation results in certain autoimmune disorders. This review will summarize recent progress in TRAIL research, including understanding of apoptotic signaling, regulation of TRAIL action, and possible therapeutic applications.


Asunto(s)
Apoptosis , Péptidos y Proteínas de Señalización Intracelular , Glicoproteínas de Membrana/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Antineoplásicos/uso terapéutico , Proteínas Reguladoras de la Apoptosis , Enfermedades Autoinmunes/tratamiento farmacológico , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD , Proteínas Portadoras/metabolismo , Caspasas/metabolismo , Sinergismo Farmacológico , Proteínas Ligadas a GPI , Humanos , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/uso terapéutico , Ratones , Proteínas/metabolismo , Proteínas de Unión al ARN , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF , Receptores del Factor de Necrosis Tumoral/fisiología , Miembro 10c de Receptores del Factor de Necrosis Tumoral , Proteínas Ribosómicas , Transducción de Señal , Ligando Inductor de Apoptosis Relacionado con TNF , Receptores Señuelo del Factor de Necrosis Tumoral , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/uso terapéutico
20.
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
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda