Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
1.
Adv Protein Chem Struct Biol ; 107: 77-115, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28215229

RESUMEN

In addition to regulating immune responses, the NF-κB family of transcription factors also promotes cellular proliferation and survival. NF-κB and its activating kinase, IKK, have become appealing therapeutic targets because of their critical roles in the progression of many diseases including chronic inflammation and cancer. Here, we discuss the conditions that lead to pathway activation, the effects of constitutive activation, and some of the strategies used to inhibit NF-κB signaling.


Asunto(s)
Quinasa I-kappa B/antagonistas & inhibidores , FN-kappa B/antagonistas & inhibidores , Proliferación Celular , Supervivencia Celular , Ensayos Clínicos como Asunto , Resistencia a Antineoplásicos , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Transducción de Señal
2.
Oncogene ; 33(10): 1297-305, 2014 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-23474754

RESUMEN

Tumor-initiating cells (TICs) are a sub-population of cells that exhibit a robust ability to self-renew and contribute to the formation of primary tumors, the relapse of previously treated tumors and the development of metastases. TICs have been identified in various tumors including those of the breast, and are particularly enriched in the basal-like and claudin-low subtypes of breast cancer. The signaling pathways that contribute to the function and maintenance of TICs are under intense study. We explored the potential involvement of the nuclear factor-κB (NF-κB) family of transcription factors in TICs in cell lines that are representative of basal-like and claudin-low breast cancer. NF-κB was found to be activated in breast cancer cells that form tumorspheres efficiently. Moreover, both canonical and non-canonical NF-κB signaling is required for these cells to self-renew in vitro and to form xenograft tumors efficiently in vivo using limiting dilutions of cells. Consistent with this fact, canonical and non-canonical NF-κB signaling is activated in TICs isolated from breast cancer cell lines. Experimental results indicate that NF-κB promotes the function of TICs by stimulating epithelial-to-mesenchymal transition and by upregulating the expression of the inflammatory cytokines interleukin-1ß and interleukin-6. The results suggest the use of NF-κB inhibitors for clinical therapy of certain breast cancers.


Asunto(s)
Neoplasias de la Mama/metabolismo , Células Madre Neoplásicas/metabolismo , Transducción de Señal , Factor de Transcripción ReIA/metabolismo , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular , Transición Epitelial-Mesenquimal , Femenino , Humanos , Quinasa I-kappa B/metabolismo , Interleucina-1beta/fisiología , Interleucina-6/fisiología , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Fosforilación , Procesamiento Proteico-Postraduccional , Esferoides Celulares/metabolismo , Factor de Crecimiento Transformador beta/fisiología
3.
Oncogene ; 30(45): 4557-66, 2011 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-21625221

RESUMEN

Elevated levels of reactive oxygen species (ROS) are found in most oncogenically transformed cells and are proposed to promote cellular transformation through mechanisms such as inhibition of phosphatases. BCR-ABL, the oncoprotein associated with the majority of chronic myeloid leukemias (CMLs), induces accumulation of intracellular ROS, causing enhanced signaling downstream of PI3K. Previously we have shown that the transcription factor nuclear factor-kappa B (NF-κB) is activated by BCR-ABL expression and is required for BCR-ABL-mediated cellular transformation. Inhibition of IκB kinase (IKKß) and NF-κB leads to cell death through an unknown mechanism. Here, we analyze the potential involvement of NF-κB in moderating BCR-ABL-induced ROS levels to protect from death. The data confirm that BCR-ABL promotes ROS and demonstrate that NF-κB prevents excessive levels. Inhibition of NF-κB leads to an increase in ROS levels and to cell death, which is at least partially controlled through ROS-induced c-Jun N-terminal kinase activity. The data demonstrate that one function for NF-κB in oncogenesis is the suppression of oncoprotein-induced ROS levels and that inhibition of NF-κB in some cancers, including CML, will increase ROS levels and promote cell death.


Asunto(s)
Apoptosis , Genes abl/fisiología , MAP Quinasa Quinasa 4/metabolismo , Subunidad p50 de NF-kappa B/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Células Sanguíneas/efectos de los fármacos , Células Sanguíneas/metabolismo , Muerte Celular , Línea Celular , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Quinasa I-kappa B/antagonistas & inhibidores , Quinasa I-kappa B/metabolismo , Ratones , Subunidad p50 de NF-kappa B/antagonistas & inhibidores
4.
Oncogene ; 30(14): 1727-32, 2011 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-21151171

RESUMEN

The induction of mammalian autophagy, a cellular catabolic bulk-degradation process conserved from humans to yeast, was recently shown to require IκB kinase (IKK), the upstream regulator of the nuclear factor (NF)-κB pathway. Interestingly, it was shown that this response did not involve NF-κB. Thus, the mechanism by which IKK promotes stimulus-induced autophagy is largely unknown. Here, we investigate the role of IKK/NF-κB in response to nutrient deprivation, the well-understood autophagy-inducing stimulus. IKK and both the classic and non-canonical pathways of NF-κB are robustly induced in response to cellular starvation. Notably, cells lacking either catalytic subunit of IKK (IKK-α or IKK-ß) fail to induce autophagy in response to cellular starvation. Importantly, we show that IKK activity but not NF-κB controls basal expression of the proautophagic gene LC3. We further demonstrate that starvation induces the expression of LC3 and two other essential autophagic genes ATG5 and Beclin-1 in an IKK-dependent manner. These results indicate that the IKK complex is a central mediator of starvation-induced autophagy in mammalian cells, and suggest that this requirement occurs at least in part through the regulation of autophagic gene expression. Interestingly, NF-κB subunits are dispensable for both basal and starvation-induced expression of proautophagic genes. However, starvation-induced activation of NF-κB is not inconsequential, as increases in expression of antiapoptotic NF-κB target genes such as Birc3 are observed in response to cellular starvation. Thus, IKK likely has multiple roles in response to starvation by regulating NF-κB-dependent antiapoptotic gene expression as well as controlling expression of autophagic genes through a yet undetermined mechanism.


Asunto(s)
Autofagia , Fibroblastos/metabolismo , Expresión Génica , Quinasa I-kappa B/metabolismo , FN-kappa B/metabolismo , Animales , Ratones , Inanición/metabolismo
5.
Oncogene ; 29(8): 1238-48, 2010 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-19946332

RESUMEN

The membrane bound receptor tyrosine kinase Her2 is overexpressed in approximately 30% of human breast cancers, which correlates with poor prognosis. Her2-induced signaling pathways include MAPK and PI3K/Akt, of which the latter has been shown to be critical for Her2(+) breast cancer cell growth and survival. In addition, the NF-kappaB pathway has been shown to be activated downstream of Her2 overexpression; however, the mechanisms leading to this activation are not currently clear. Using Her2(+)/ER(-) breast cancer cells, we show that Her2 activates NF-kappaB through the canonical pathway which, surprisingly, involves IKKalpha. Knockdown of IKKalpha led to a significant decrease in transcription levels of multiple NF-kappaB-regulated cytokine and chemokine genes. siRNA-mediated knockdown of IKKalpha resulted in a decrease in cancer cell invasion, but had no effect on cell proliferation. Inhibition of the PI3K/Akt pathway had no effect on NF-kappaB activation, but significantly inhibited cell proliferation. Our study suggests different roles for the NF-kappaB and PI3K pathways downstream of Her2, leading to changes in invasion and proliferation of breast cancer cells. In addition this work indicates the importance of IKKalpha as a mediator of Her2-induced tumor progression.


Asunto(s)
Neoplasias de la Mama/metabolismo , Quinasa I-kappa B/fisiología , FN-kappa B/metabolismo , Receptor ErbB-2/metabolismo , Proteínas Supresoras de Tumor/farmacología , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Quinasa I-kappa B/metabolismo , FN-kappa B/fisiología , Invasividad Neoplásica/patología , Invasividad Neoplásica/fisiopatología , Receptor ErbB-2/genética , Receptor ErbB-2/fisiología , Transducción de Señal/fisiología
6.
Am J Transplant ; 9(3): 452-62, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19260829

RESUMEN

The ex vivo induction of alloantigen-specific hyporesponsiveness by costimulatory pathway blockade or exposure to immunoregulatory cytokines has been shown to inhibit proliferation, IL-2 production, and the graft-versus-host disease (GVHD) capacity of adoptively transferred T-cells. We hypothesized that inhibition of the intracellular NF-kappaB pathway in alloreactive T-cells, which is critical for T-cell activation events including IL-2 transcription, could lead to alloantigen hyporesponsiveness and loss of GVHD capacity. We demonstrate that treatment of mixed lymphocyte reaction (MLR) cultures with PS1145, a potent inhibitor of NF-kappaB activation, can induce T-cell hyporesponsiveness to alloantigen in primary and secondary responses while preserving in vitro responses to potent mitogenic stimulation. GVHD lethality in recipients of ex vivo PS1145-treated cells was profoundly inhibited. Parking of control or PS1145-treated MLR cells in syngeneic Rag(-/-) recipients resulted in intact contact hypersensitivity (CHS) responses. However, GVHD lethality capacity also was restored, suggesting that lymphopenic expansion uncoupled alloantigen hyporesponsiveness. These results indicate that the NF-kappaB pathway is a critical regulator of alloresponses and provide a novel small molecule inhibitor based approach that is effective in preventing early posttransplant GVHD lethality but that also permits donor T-cell responses to recover after a period of lymphopenic expansion.


Asunto(s)
Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/metabolismo , Isoantígenos/inmunología , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Transducción de Señal/efectos de los fármacos , Linfocitos T/inmunología , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Femenino , Compuestos Heterocíclicos con 3 Anillos/farmacología , Prueba de Cultivo Mixto de Linfocitos , Masculino , Ratones , Modelos Inmunológicos , Piridinas/farmacología , Linfocitos T/citología , Linfocitos T/efectos de los fármacos
7.
Oncogene ; 25(51): 6817-30, 2006 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-17072330

RESUMEN

Abundant data support a key role for the transcription factor nuclear factor-kappaB (NF-kappaB) signaling pathway in controlling the initiation and progression of human cancer. NF-kappaB and associated regulatory proteins such as IkappaB kinase (IKK) are activated downstream of many oncoproteins and there is much evidence for the activation of NF-kappaB-dependent target genes in a variety of solid tumors and hematologic malignancies. This review focuses on the mechanisms by which the NF-kappaB pathway is activated in cancer and on the oncogenic functions controlled by activated NF-kappaB. Additionally, the effects of NF-kappaB activation in tumors relative to cancer therapy are also discussed.


Asunto(s)
Transformación Celular Neoplásica , Quinasa I-kappa B/antagonistas & inhibidores , FN-kappa B/fisiología , Animales , Genes Supresores de Tumor , Humanos , FN-kappa B/antagonistas & inhibidores
8.
Cell Death Differ ; 13(5): 738-47, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16485028

RESUMEN

The transcription factor NF-kappaB and associated regulatory factors (including IkappaB kinase subunits and the IkappaB family member Bcl-3) are strongly implicated in a variety of hematologic and solid tumor malignancies. A role for NF-kappaB in cancer cells appears to involve regulation of cell proliferation, control of apoptosis, promotion of angiogenesis, and stimulation of invasion/metastasis. Consistent with a role for NF-kappaB in oncogenesis are observations that inhibition of NF-kappaB alone or in combination with cancer therapies leads to tumor cell death or growth inhibition. However, other experimental data indicate that NF-kappaB can play a tumor suppressor role in certain settings and that it can be important in promoting an apoptotic signal downstream of certain cancer therapy regimens. In order to appropriately move NF-kappaB inhibitors in the clinic, thorough approaches must be initiated to determine the molecular mechanisms that dictate the complexity of oncologic and therapeutic outcomes that are controlled by NF-kappaB.


Asunto(s)
Quinasa I-kappa B/fisiología , Modelos Biológicos , FN-kappa B/metabolismo , Neoplasias/etiología , Neoplasias/terapia , Apoptosis , Proliferación Celular , Progresión de la Enfermedad , Humanos , Quinasa I-kappa B/metabolismo , Inflamación/metabolismo , FN-kappa B/antagonistas & inhibidores , Metástasis de la Neoplasia/patología , Neoplasias/inmunología , Neoplasias/metabolismo , Sensibilidad y Especificidad , Transducción de Señal
9.
Mol Genet Metab ; 75(4): 360-8, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12051968

RESUMEN

Nitric oxide (NO) has been shown to mediate multiple physiological and toxicological functions. The inducible nitric oxide synthase (iNOS) is responsible for the high output generation of NO by macrophages following their stimulation by cytokines or bacterial antigens. The inhibition of TNF alpha-stimulated HIV expression and the anti-inflammatory property of PD144795, a new benzothiophene derivative, have been recently described. We have now analyzed whether some of these properties could be mediated by an effect of PD144795 on NO-dependent inflammatory events. We show that PD144795 suppresses the lipopolysaccharide-elicited production of nitrite (NO(-)(2)) by primary peritoneal mouse macrophages and by a macrophage-derived cell line, RAW 264.7. This effect was dependent on the dose and timing of addition of PD144795 to the cells. Suppression of NO(-)(2) production was associated with a decrease in the amount of iNOS protein, iNOS enzyme activity and mRNA expression. The effect of PD144795 was partially abolished by coincubation of the cells with LPS and IFN gamma. However, the inhibitory effect of PD144795 was not abrogated by the simultaneous addition of LPS and TNF alpha, which indirectly suggests that the effect of PD144795 was not due to the inhibition of TNF alpha synthesis. Additionally, PD144795 did not block NF-kappa B nuclear translocation induced by LPS. Inhibition of iNOS gene expression represents a novel mechanism of PD144795 action that underlines the anti-inflammatory effects of this immunosuppressive drug.


Asunto(s)
Macrófagos Peritoneales/efectos de los fármacos , Óxido Nítrico Sintasa/genética , Tiofenos/farmacología , Animales , Fármacos Anti-VIH/farmacología , Línea Celular , Células Cultivadas , Expresión Génica/efectos de los fármacos , Inmunosupresores/farmacología , Lipopolisacáridos/farmacología , Macrófagos Peritoneales/enzimología , Macrófagos Peritoneales/inmunología , Ratones , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Tiofenos/química
10.
Mol Cell Biol ; 21(24): 8428-36, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11713278

RESUMEN

Bcl-3 is a distinctive member of the IkappaB family of NF-kappaB inhibitors because it can function to coactivate transcription. A potential involvement of Bcl-3 in oncogenesis is highlighted by the fact that it was cloned due to its location at a breakpoint junction in some cases of human B-cell chronic lymphocytic leukemia and that it is highly expressed in human breast tumor tissue. To analyze the effects of Bcl-3 dysregulation in breast epithelial cells, we created stable immortalized human breast epithelial cell lines either expressing Bcl-3 or carrying the corresponding vector control plasmid. Analysis of the Bcl-3-expressing cells suggests that these cells have a shortened G(1) phase of the cell cycle as well as a significant increase in hyperphosphorylation of the retinoblastoma protein. Additionally, the cyclin D1 gene was found to be highly expressed in these cells. Upon further analysis, Bcl-3, acting as a coactivator with NF-kappaB p52 homodimers, was demonstrated to directly activate the cyclin D1 promoter through an NF-kappaB binding site. Therefore, our results demonstrate that dysregulated expression of Bcl-3 potentiates the G(1) transition of the cell cycle by stimulating the transcription of the cyclin D1 gene in human breast epithelial cells.


Asunto(s)
Ciclina D1/metabolismo , Fase G1 , Proteínas Proto-Oncogénicas/metabolismo , Células 3T3 , Animales , Proteínas del Linfoma 3 de Células B , Sitios de Unión , Northern Blotting , Western Blotting , Mama/metabolismo , Células COS , Ciclo Celular , División Celular , Línea Celular , Núcleo Celular/metabolismo , Separación Celular , Clonación Molecular , Células Epiteliales/metabolismo , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica , Humanos , Leucemia de Células B/metabolismo , Luciferasas/metabolismo , Ratones , FN-kappa B/antagonistas & inhibidores , Fosforilación , Plásmidos/metabolismo , Regiones Promotoras Genéticas , Proteína de Retinoblastoma/metabolismo , Factores de Transcripción , Transfección
11.
Mol Cell Biol ; 21(20): 7065-77, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11564889

RESUMEN

Regulation of NF-kappaB transactivation function is controlled at several levels, including interactions with coactivator proteins. Here we show that the transactivation function of NF-kappaB is also regulated through interaction of the p65 (RelA) subunit with histone deacetylase (HDAC) corepressor proteins. Our results show that inhibition of HDAC activity with trichostatin A (TSA) results in an increase in both basal and induced expression of an integrated NF-kappaB-dependent reporter gene. Chromatin immunoprecipitation (ChIP) assays show that TSA treatment causes hyperacetylation of the wild-type integrated NF-kappaB-dependent reporter but not of a mutant version in which the NF-kappaB binding sites were mutated. Expression of HDAC1 and HDAC2 repressed tumor necrosis factor (TNF)-induced NF-kappaB-dependent gene expression. Consistent with this, we show that HDAC1 and HDAC2 target NF-kappaB through a direct association of HDAC1 with the Rel homology domain of p65. HDAC2 does not interact with NF-kappaB directly but can regulate NF-kappaB activity through its association with HDAC1. Finally, we show that inhibition of HDAC activity with TSA causes an increase in both basal and TNF-induced expression of the NF-kappaB-regulated interleukin-8 (IL-8) gene. Similar to the wild-type integrated NF-kappaB-dependent reporter, ChIP assays showed that TSA treatment resulted in hyperacetylation of the IL-8 promoter. These data indicate that the transactivation function of NF-kappaB is regulated in part through its association with HDAC corepressor proteins. Moreover, it suggests that the association of NF-kappaB with the HDAC1 and HDAC2 corepressor proteins functions to repress expression of NF-kappaB-regulated genes as well as to control the induced level of expression of these genes.


Asunto(s)
Regulación hacia Abajo , Regulación Enzimológica de la Expresión Génica , Histona Desacetilasas/metabolismo , FN-kappa B/metabolismo , FN-kappa B/fisiología , Proteínas Represoras , Células 3T3 , Acetilación , Animales , Sitios de Unión , Northern Blotting , Western Blotting , Células COS , Línea Celular , Cromatina/metabolismo , Inhibidores Enzimáticos/farmacología , Células HeLa , Histona Desacetilasa 1 , Histona Desacetilasa 2 , Humanos , Ácidos Hidroxámicos/farmacología , Luciferasas/metabolismo , Ratones , Plásmidos/metabolismo , Pruebas de Precipitina , Unión Proteica , Factor de Transcripción ReIA , Activación Transcripcional , Transfección
12.
J Biol Chem ; 276(44): 41197-204, 2001 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-11483591

RESUMEN

We have previously shown that fetal rat brain cells, preneuronal (PC12), and hepatocyte (CWSV-1) cells undergo apoptosis during choline deficiency (CD). The PC12 and epithelial cell culture models were used to determine the molecular mechanism by which CD induces apoptosis. Our data indicate that CD leads to both growth arrest and apoptosis in a subpopulation of cells, which correlate with the up-regulation of the tumor suppressor protein p53 and concurrent up-regulation of the cyclin-dependent kinase-inhibitor p21(WAF1/CIP1). Additionally, CD induced both a G1/S and a G2/M arrest. Transient transfection of a dominant negative p53 (p53DN) construct into PC12 cells, which inhibited endogenous p53 activation, significantly reduced the induction of apoptosis associated with CD. Interestingly, CD also induced the persistent activation of the transcription factor NF-kappaB. Activation of NF-kappaB has been shown to promote cell survival and proposed to antagonize p53. Consistent with this, expression of a super-repressor form of IkappaBalpha (SR-IkappaBalpha) that functions to strongly inhibit NF-kappaB activation, profoundly enhanced cell death during CD. In summary, these results suggest that the effects of CD on apoptosis and subsequent cell survival are mediated through two different signaling pathways, p53 and NF-kappaB, respectively. Taken together, our data demonstrates the induction of opposing mechanisms associated with nutrient deficiency that may provide a molecular mechanism by which CD promotes carcinogenesis.


Asunto(s)
Apoptosis/fisiología , Deficiencia de Colina/complicaciones , FN-kappa B/fisiología , Proteína p53 Supresora de Tumor/fisiología , Animales , Ciclo Celular , Línea Celular , Supervivencia Celular , Ensayo de Cambio de Movilidad Electroforética , Etiquetado Corte-Fin in Situ , Neoplasias Hepáticas Experimentales/etiología , Ratas
13.
J Biol Chem ; 276(42): 38658-64, 2001 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-11509575

RESUMEN

The prostaglandin H synthases (PGHS) catalyze the conversion of arachidonic acid to prostaglandin H(2), the committed step in prostanoid synthesis. Two forms of PGHS exist, PGHS-1 (COX-1) and PGHS-2 (COX-2). The gene encoding the latter form is known to be inducible by a number of stimuli including several inflammatory mediators. Recent evidence indicates that the inducible cyclooxygenase may have both pro- and anti-inflammatory properties through the generation of different prostaglandins. Previous reports indicate that the transcription factor NF-kappaB can function upstream of COX-2 to control transcription of this gene and that the cyclopentenone prostaglandins can inhibit NF-kappaB activation via the inhibition of the IkappaB kinase. Thus, it is suggested that cyclopentenones feed back to inhibit continued nuclear accumulation of NF-kappaB. In this report we demonstrate COX-2 expression inhibits nuclear translocation of NF-kappaB, and we confirm that the cyclopentenone prostaglandins inhibit NF-kappaB. In addition, we show that prostaglandin E(2) and its analogs promote the inherent transcriptional activity of the p65/RelA subunit of NF-kappaB in a manner independent of induced nuclear accumulation. Consistent with this evidence, prostaglandin E(2) strongly synergizes with the inflammatory cytokine tumor necrosis factor-alpha to promote NF-kappaB-dependent transcription and gene expression. The data provide a molecular rationale to explain both the pro- and anti-inflammatory nature of COX-2.


Asunto(s)
Regulación de la Expresión Génica , Isoenzimas/metabolismo , FN-kappa B/metabolismo , Prostaglandina-Endoperóxido Sintasas/metabolismo , Prostaglandinas/fisiología , Transporte Activo de Núcleo Celular , Sitios de Unión , Northern Blotting , Línea Celular , Núcleo Celular/metabolismo , Ciclooxigenasa 2 , Citoplasma/metabolismo , Dinoprostona/fisiología , Regulación hacia Abajo , Activación Enzimática , Vectores Genéticos , Humanos , Quinasa I-kappa B , Inflamación , Interleucina-8/metabolismo , Luciferasas/metabolismo , Proteínas de la Membrana , Modelos Biológicos , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína , Factores de Tiempo , Transcripción Genética , Transfección , Células Tumorales Cultivadas , Regulación hacia Arriba
14.
J Immunol ; 167(3): 1461-8, 2001 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-11466366

RESUMEN

Insulin-dependent diabetes mellitus (IDDM) is characterized by the T cell-mediated destruction of insulin-producing beta cells. Accordingly, APCs, such as macrophage, have also been shown to be important in the disease process. However, the role(s) of dendritic cells (DCs) that exhibit potent APC function remains undefined in IDDM. Here we demonstrate that DCs derived from nonobese diabetic (NOD) mice, a model for IDDM, are more sensitive to various forms of stimulation compared with those from C57BL/6 and BALB/c mice, resulting in increased IL-12 secretion. This property is a consequence of hyperactivation of NF-kappaB, a transcription factor known to regulate IL-12 gene expression. Specifically, NOD DCs exhibit persistent hyperactivation of both IkappaB kinase and NF-kappaB in response to stimuli, in addition to selective degradation of IkappaBepsilon. Transfection of NOD DCs with a modified form of IkappaBalpha significantly reduced IL-12 secretion, suggesting that hyperactivation of NF-kappaB was in part responsible for increased IL-12 production. An enhanced capacity of NOD DCs to secrete IL-12 would be expected to contribute to the development of pathogenic Th1 (Tc1) cells during the diabetogenic response.


Asunto(s)
Células Dendríticas/enzimología , Células Dendríticas/inmunología , Proteínas I-kappa B , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transporte Activo de Núcleo Celular/inmunología , Animales , Núcleo Celular/metabolismo , Células Cultivadas , Células Dendríticas/metabolismo , Activación Enzimática/inmunología , Femenino , Quinasa I-kappa B , Interleucina-12/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , FN-kappa B/fisiología , Proteínas Serina-Treonina Quinasas/aislamiento & purificación , Proteínas Proto-Oncogénicas/metabolismo , Activación Transcripcional/inmunología
16.
Int J Radiat Oncol Biol Phys ; 50(1): 183-93, 2001 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-11316563

RESUMEN

PURPOSE: NF-kappaB is activated by tumor necrosis factor, certain chemotherapeutic agents, and ionizing radiation, leading to inhibition of apoptosis. NF-kappaB activation is regulated by phosphorylation of IkappaB inhibitor molecules that are subsequently targeted for degradation by the ubiquitin-proteasome pathway. PS-341 is a specific and selective inhibitor of the proteasome that inhibits NF-kappaB activation and enhances cytotoxic effects of chemotherapy in vitro and in vivo. The objective of this study was to determine if proteasome inhibition leads to enhanced radiation sensitivity. METHODS AND MATERIALS: Inhibition of NF-kappaB activation in colorectal cancer cells was performed by treatment of LOVO cells with PS-341 or infection with an adenovirus encoding IkappaB super-repressor, a selective NF-kappaB inhibitor. Cells were irradiated at 0, 2, 4, 6, 8, and 10 Gy with or without inhibition of NF-kappaB. NF-kappaB activation was determined by electrophoretic mobility gel shift assay, and apoptosis was evaluated using the TUNEL assay. Growth and clonogenic survival data were obtained to assess effects of treatment on radiosensitization. In vitro results were tested in vivo using a LOVO xenograft model. RESULTS: NF-kappaB activation was induced by radiation and inhibited by pretreatment with either PS-341 or IkappaBalpha super-repressor in all cell lines. Inhibition of radiation-induced NF-kappaB activation resulted in increased apoptosis and decreased cell growth and clonogenic survival. A 7-41% increase in radiosensitivity was observed for cells treated with PS-341 or IkappaBalpha. An 84% reduction in initial tumor volume was obtained in LOVO xenografts receiving radiation and PS-341. CONCLUSIONS: Inhibition of NF-kappaB activation increases radiation-induced apoptosis and enhances radiosensitivity in colorectal cancer cells in vitro and in vivo. Results are encouraging for the use of PS-341 as a radiosensitizing agent in the treatment of colorectal cancer.


Asunto(s)
Ácidos Borónicos/farmacología , Proteínas I-kappa B , Complejos Multienzimáticos/antagonistas & inhibidores , FN-kappa B/fisiología , Inhibidores de Proteasas/farmacología , Pirazinas/farmacología , Tolerancia a Radiación/fisiología , Fármacos Sensibilizantes a Radiaciones/farmacología , Adenoviridae/genética , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Bortezomib , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/radioterapia , Cisteína Endopeptidasas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Femenino , Humanos , Ratones , Ratones Desnudos , Inhibidor NF-kappaB alfa , FN-kappa B/antagonistas & inhibidores , Complejo de la Endopetidasa Proteasomal , Tolerancia a Radiación/efectos de los fármacos , Proteínas Represoras/genética , Proteínas Represoras/fisiología , Transducción Genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
17.
J Biol Chem ; 276(25): 22382-7, 2001 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-11297551

RESUMEN

The sedative and anti-nausea drug thalidomide, which causes birth defects in humans, has been shown to have both anti-inflammatory and anti-oncogenic properties. The anti-inflammatory effect of thalidomide is associated with suppression of cytokine expression and the anti-oncogenic effect with inhibition of angiogenesis. It is presently unclear whether the teratogenic properties of thalidomide are connected in any way to the beneficial, anti-disease characteristics of this drug. The transcription factor NF-kappaB has been shown to be a key regulator of inflammatory genes such as tumor necrosis factor-alpha and interleukin-8. Inhibition of NF-kappaB is associated with reduced inflammation in animal models, such as those for rheumatoid arthritis. We show here that thalidomide can block NF-kappaB activation through a mechanism that involves the inhibition of activity of the IkappaB kinase. Consistent with the observed inhibition of NF-kappaB, thalidomide blocked the cytokine-induced expression of NF-kappaB-regulated genes such as those encoding interleukin-8, TRAF1, and c-IAP2. These data indicate that the therapeutic potential for thalidomide may be based on its ability to block NF-kappaB activation through suppression of IkappaB kinase activity.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , FN-kappa B/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Talidomida/farmacología , Secuencia de Bases , Sondas de ADN , Humanos , Quinasa I-kappa B , Interleucina-1/antagonistas & inhibidores , Interleucina-1/fisiología , Interleucina-8/genética , Células Jurkat , FN-kappa B/biosíntesis , Transcripción Genética/efectos de los fármacos , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/fisiología
18.
Cancer Res ; 61(9): 3535-40, 2001 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-11325813

RESUMEN

Inducible activation of nuclear factor-kappaB (NF-kappaB) inhibits the apoptotic response to chemotherapy and irradiation. Activation of NF-kappaB via phosphorylation of an inhibitor protein IkappaB leads to degradation of IkappaB through the ubiquitin-proteasome pathway. We hypothesized that inactivation of proteasome function will inhibit inducible NF-kappaB activation, thereby increasing levels of apoptosis in response to chemotherapy and enhancing antitumor effects. To assess the effects of proteasome inhibition on chemotherapy response, human colorectal cancer cells were pretreated with the dipeptide boronic acid analogue PS-341 (1 microM) prior to exposure to SN-38, the active metabolite of the topoisomerase I inhibitor, CPT-11. Inducible activation of NF-kappaB and growth response were evaluated in vitro and in vivo. Effects on p53, p21, p27 and apoptosis were determined. Pretreatment with PS-341 inhibited activation of NF-kappaB induced by SN-38 and resulted in a significantly higher level of growth inhibition (64-75%) compared with treatment with PS-341 alone (20-30%) or SN-38 alone (24-47%; P < 0.002). Combination therapy resulted in a 94% decrease in tumor size compared with the control group and significantly improved tumoricidal response to treatment compared with all treatment groups (P = 0.02). The level of apoptosis was 80-90% in the treatment group that received combination treatment compared with treatment with single agent alone (10%). Proteasome inhibition blocks chemotherapy-induced NF-kappaB activation, leading to a dramatic augmentation of chemosensitivity and enhanced apoptosis. Combining proteasome inhibition with chemotherapy has significant potential to overcome the high incidence of chemotherapy resistance. Clinical studies are currently in development to evaluate the role of proteasome inhibition as an important adjuvant to systemic chemotherapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Ácidos Borónicos/farmacología , Camptotecina/análogos & derivados , Camptotecina/farmacología , Inhibidores Enzimáticos/farmacología , FN-kappa B/antagonistas & inhibidores , Pirazinas/farmacología , Animales , Apoptosis/efectos de los fármacos , Ácidos Borónicos/administración & dosificación , Bortezomib , Camptotecina/administración & dosificación , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Cisteína Endopeptidasas , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Inhibidores Enzimáticos/administración & dosificación , Femenino , Humanos , Irinotecán , Ratones , Ratones Desnudos , Complejos Multienzimáticos/antagonistas & inhibidores , Complejo de la Endopetidasa Proteasomal , Pirazinas/administración & dosificación , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
19.
J Biol Chem ; 276(22): 18934-40, 2001 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-11259436

RESUMEN

The serine/threonine kinase Akt/PKB is a potent regulator of cell survival and has oncogenic transformation potential. Previously, it has been shown that Akt can activate the transcription factor NF-kappaB and that this functions to block apoptosis induced by certain stimuli. The mechanism whereby Akt activates NF-kappaB has been controversial, with evidence supporting induction of nuclear translocation of NF-kappaB via activation of IkappaB kinase activity and/or the stimulation of the transcription function of NF-kappaB. Here we demonstrate that Akt targets the transactivation function of NF-kappaB by stimulating the transactivation domain of RelA/p65 in a manner that is dependent on IkappaB kinase beta activity and on the mitogen-activated protein kinase p38 (p38). Activation of RelA/p65 transactivation function requires serines 529 and 536, sites shown previously to be inducibly phosphorylated. Consistent with the requirement of p38 in the activation of NF-kappaB transcriptional function, expression of activated Akt induces p38 activity. Furthermore, the ability of IL-1beta to activate NF-kappaB is known to involve Akt, and we show here that IL-1beta induces p38 activity in manner dependent on Akt and IkappaB kinase activation. Interestingly, activated Akt and the transcriptional co-activators CBP/p300 synergize in the activation of the RelA/p65 transactivation domain, and this synergy is blocked by p38 inhibitors. These studies demonstrate that Akt, functioning through IkappaB kinase and p38, induces the transcription function of NF-kappaB by stimulating the RelA/p65 transactivation subunit of NF-kappaB.


Asunto(s)
Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/fisiología , Activación Transcripcional , Células 3T3 , Animales , Apoptosis , Sitios de Unión , Western Blotting , Núcleo Celular/metabolismo , Supervivencia Celular , Proteína p300 Asociada a E1A , Genes Dominantes , Genes Reporteros , Humanos , Quinasa I-kappa B , Interleucina-1/metabolismo , Luciferasas , Ratones , Proteínas Quinasas Activadas por Mitógenos/genética , Modelos Biológicos , Mutagénesis Sitio-Dirigida , Mutación , Proteínas Nucleares/metabolismo , Fosforilación , Plásmidos/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Transactivadores/metabolismo , Factor de Transcripción ReIA , Transcripción Genética , Transfección , Proteínas Quinasas p38 Activadas por Mitógenos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA