Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Interferon Cytokine Res ; 21(11): 951-9, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11747627

RESUMEN

We have previously identified a nuclear localization sequence (NLS) in interferon-gamma (IFN-gamma). This NLS functions intracellularly by forming a complex with its transcription factor Stat1alpha and the nuclear importer of Stat1alpha, the importin-alpha analog NPI-1. The stability of this complex and the subsequent nuclear translocation of the complexed Stat1alpha are dependent on the integrity of this NLS, showing that Stat1alpha nuclear import is mediated by the IFN-gamma NLS. In this study, to directly evaluate the intrinsic requirement of nuclear IFN-gamma toward its biologic activities, we engineered a chimeric in which the IFN-gamma NLS has been substituted by a heterologous NLS, namely, the prototypical NLS of the SV40 large T antigen, which would drive nuclear translocation of IFN-gamma in a sequence-nonspecific manner. The chimeric, IFN-gamma-SV, was equally active in antiviral and antiproliferative assays as the wild-type IFN-gamma. Interestingly, IFN-gamma-SV was also translocated to the nucleus and was also recovered intracellularly as a complex with the Stat1alpha importer NPI-1, like wild-type IFN-gamma. Comparison with an NLS deletion mutant showed that deletion or changes within the NLS motif of IFN-gamma were inconsequential to the high-affinity extracellular binding to the IFN-gamma receptor complex, yet the presence of an NLS was critical to the expression of the biologic activities of IFN-gamma and its NPI-1 complexation ability. Our data conclusively demonstrate that nuclear translocation of IFN-gamma is an intrinsic requirement for the full expression of the biologic activities of IFN-gamma and strengthen the conclusion that nuclear chaperoning of Stat1alpha is the primary role of IFN-gamma nuclear translocation. This type of ligand imprinting by sequestering of activated Stat may contribute to the specificity of Stat nuclear transcription.


Asunto(s)
Antivirales/metabolismo , Núcleo Celular/metabolismo , Interferón gamma/metabolismo , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Animales , Antígenos Transformadores de Poliomavirus/genética , Antivirales/química , Unión Competitiva , División Celular , Línea Celular , Relación Dosis-Respuesta a Droga , Factor 3 de Genes Estimulados por el Interferón , Interferón gamma/química , Interferón gamma/genética , Microscopía Fluorescente , Datos de Secuencia Molecular , Señales de Localización Nuclear , Receptores de Interferón/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Transducción de Señal , Factores de Transcripción/metabolismo , Receptor de Interferón gamma
2.
J Neurooncol ; 48(3): 225-32, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11100820

RESUMEN

The antiproliferative effect of IFNalpha was tested on the human glioblastoma cell lines, U-373MG and T98G. IFNalpha significantly inhibited the growth of both cell lines, but was more effective in retarding the growth of U-373MG cells. Flow cytometry analysis indicated that synchronized IFNalpha-treated U-373MG cells showed a strong block in the progression of cells out of the S phase of the cell cycle. T98G cells, on the other hand, showed a moderate delay in the transition of cells from G1 to S phase and only a slight delay in the S phase, consistent with the decreased antiproliferative effect of IFNalpha on this cell line. IFNalpha-treated cells were then tested for the induction of the tumor suppressor gene product, p21(WAF1/CIP1). Higher levels of p21(WAF1/CIP1) were detected in lysates from IFNalpha-treated U-373MG cells as compared to media controls for as long as 18 h. In IFNalpha-treated T98G cells, p21(WAF1/CIP1) levels were slightly elevated at 4 and 6 h, but decreased to levels similar to controls thereafter, correlating with the antiproliferative effects of IFNalpha on each cell line. Immunoprecipitation studies on lysates from IFNalpha-treated U-373MG and T98G cells indicated that increased amounts of p21(WAF1/CIP1) were complexed to both cyclin D1 and cyclin E. Further, reduced cyclin-dependent kinase 2 (cdk2) activity was found in both IFNalpha-treated U-373MG and T98G cells, suggesting a mechanism by which p21(WAF1/CIP1) exerted its antiproliferative effects. Lastly, we analyzed the time-dependent production of the cyclins D1, E, and A. No differences in cyclin D1 levels were found between IFNalpha-treated and media-treated U-373MG and T98G cells. However, both IFNalpha-treated U-373MG and T98G cells showed a prolonged elevation in cyclin E, correlating with the G1 to S phase delays observed in these cell lines. Further, the duration of cyclin E production corresponded with the magnitude of the cell cycle delays seen in IFNalpha-treated U-373MG and T98G cells. Prolonged elevation of cyclin A was also seen in both IFNalpha-treated U-373MG and T98G cells, the magnitude of which correlated with the S phase delay observed in these cell lines. Thus, the data indicate that IFNalpha has significant antiproliferative activity against glioblastoma cells that is mediated, at least in part, by the tumor suppressor gene product, p21(WAF1/CIP1).


Asunto(s)
Neoplasias Encefálicas/patología , Quinasas CDC2-CDC28 , Ciclo Celular/efectos de los fármacos , Ciclinas/genética , Glioblastoma/patología , Interferón-alfa/farmacología , División Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/metabolismo , Fase G1/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Cinética , Proteínas Serina-Treonina Quinasas/metabolismo , Fase S/efectos de los fármacos , Células Tumorales Cultivadas
3.
J Interferon Cytokine Res ; 20(5): 463-9, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10841074

RESUMEN

Glioblastoma multiforme is one of the most aggressive and frequently occurring forms of brain cancer. It originates from astrocytes and is characterized by a loss of cell cycle control frequently involving mutations in tumor suppressor genes, such as p53 and p16. Nucleoside analogs, such as acyclovir (ACV), are currently being used in the treatment of viral diseases, such as those caused by members of the herpes family. Further, ACV in combination with type I interferons (IFN) has been shown to be more effective at lower doses in treatment of viral diseases. We show here that ACV at high concentrations (up to 500 microg/ml) inhibited growth in tissue culture of the human glioblastoma cell lines T98G, SNB-19, and U-373 by as much as 68.3% while inhibiting normal human astrocytes by only 38.3%. Related to this, the tumor cells were more than sevenfold more efficient in phosphorylation of ACV to the active phosphate form than normal human astrocytes. Analogous to treatment of virus-infected cells, suboptimal concentrations of ACV were as effective as high concentrations when used in conjunction with low concentrations of IFN-gamma in inhibition of tumor cell growth. At the cellular level, ACV and IFN-gamma inhibited the cell cycle in both the G1 and S phases. The cooperative effect of ACV and IFN-gamma against the glioblastomas appears to be due to direct inhibition of DNA synthesis by ACV in the S phase of the cell cycle and induction by IFN-gamma of the tumor suppressor gene p21wAF1/CIP1, which in turn acts at the level of proliferating cell nuclear antigen (PCNA) and cyclin E/cyclin-dependent kinase 2 (Cdk2) binding and inhibition of function. These studies show that the combination of IFN-gamma and ACV at suboptimal concentrations elicits significant antiproliferative effects on the glioblastoma cell lines T98G, SNB-19, and U-373 while having very little effect on normal human astrocyte cell proliferation.


Asunto(s)
Aciclovir/administración & dosificación , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Quinasas CDC2-CDC28 , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Interferón gamma/administración & dosificación , Antivirales/administración & dosificación , Astrocitos/citología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Neoplasias Encefálicas/metabolismo , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular , Quinasa 2 Dependiente de la Ciclina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , Sinergismo Farmacológico , Glioblastoma/metabolismo , Humanos , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Recombinantes , Células Tumorales Cultivadas
4.
J Interferon Cytokine Res ; 19(6): 631-6, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10433364

RESUMEN

Interferon-tau (IFN-tau) is a novel type I IFN that was originally identified as a pregnancy recognition hormone. IFN-tau shares all of the biological properties of other type I IFNs including antiviral activity and antiproliferative activity through induction of the cell cycle inhibitor gene product p21WAF1. It is a promising therapy for cancers, viral infections, and for autoimmune disorders such as multiple sclerosis, without the adverse side effects associated with IFN-alpha and IFN-beta. Here, we describe novel growth and induction conditions for the expression of functionally active and uniformly 15N-labeled IFN-tau from Pichia pastoris in a minimal media for use in initial 2D- and 3D-NMR studies in solution. Purified 15N-IFN-tau was homogenous, as determined by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and MALDI-TOF mass spectrometer (MS), and retained full biological activity. MS analysis confirmed uniform isotopic labeling of IFN-tau with 15N incorporation exceeding 99%. Circular dichroism (CD) as well as 1D-NMR and 15N-1H heteronuclear single quantum coherence (HSQC) spectra confirmed that purified 15N-labeled IFN-tau has a stable secondary structure. Besides providing a route for isotope labeling of IFN-tau, our procedure may be useful for the expression and purification of other proteins that are difficult to obtain in Pichia pastoris grown in minimal media.


Asunto(s)
Antivirales/metabolismo , Interferón Tipo I/biosíntesis , Pichia/efectos de los fármacos , Proteínas Gestacionales/biosíntesis , Antivirales/aislamiento & purificación , División Celular/efectos de los fármacos , Medios de Cultivo , Femenino , Humanos , Interferón Tipo I/aislamiento & purificación , Espectroscopía de Resonancia Magnética/métodos , Pichia/crecimiento & desarrollo , Embarazo , Proteínas Gestacionales/aislamiento & purificación , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/aislamiento & purificación , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
5.
Blood ; 93(1): 208-16, 1999 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-9864163

RESUMEN

We have analyzed the effects of interleukin-10 (IL-10) on the entry of quiescent CD4(+) T cells into the cell cycle upon stimulation with the superantigen staphylococcal enterotoxin B (SEB). IL-10 arrested cells at G0/G1. IL-10 treatment prevented the downregulation of p27(Kip1), an inhibitory protein that controls progression out of the G0 phase of the cell cycle. IL-10 also prevented the upregulation of the G1 cyclins D2 and D3, proteins necessary for entry and progression through the G1 phase of the cell cycle. Associated with the inhibition of the cell cycle, IL-10 suppressed SEB induction of interleukin-2 (IL-2). Addition of exogenous IL-2 to IL-10-treated cells significantly reversed the antiproliferative effects of IL-10. Moreover, IL-10 effects on the early G1 proteins p27(Kip1) and cyclin D2 were similarly reversed by exogenous IL-2. Although this reversal by IL-2 was pronounced, it was not complete, suggesting that IL-10 may have some effects not directly related to the suppression of IL-2 production. Cell separation experiments suggest that IL-10 can effect purified CD4(+) T cells directly, providing functional evidence for the presence of IL-10 receptors on CD4(+) T cells. IL-10 also inhibited expression of IL-2 transcriptional regulators c-fos and c-jun, which also inhibit other cell functions. Our studies show that the mechanism of IL-10 regulation of quiescent CD4(+) T-cell activation is mainly by blocking induction of IL-2 that is critical to downregulation of p27(Kip1) and upregulation of D cyclins in T-cell activation and entry into the cell cycle.


Asunto(s)
Proteínas de Ciclo Celular , Ciclo Celular/inmunología , Enterotoxinas/farmacología , Inmunosupresores/farmacología , Interleucina-10/fisiología , Activación de Linfocitos , Superantígenos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Proteínas Supresoras de Tumor , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , División Celular/inmunología , Ciclina D2 , Ciclina D3 , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Ciclinas/metabolismo , Regulación de la Expresión Génica/inmunología , Inhibidores de Crecimiento/farmacología , Humanos , Interleucina-10/antagonistas & inhibidores , Interleucina-2/antagonistas & inhibidores , Interleucina-2/farmacología , Leucocitos Mononucleares/citología , Activación de Linfocitos/inmunología , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/enzimología , Regulación hacia Arriba/inmunología
6.
J Biol Chem ; 274(1): 403-7, 1999 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-9867857

RESUMEN

Cytokines such as interferon-gamma (IFN-gamma), which utilize the well studied JAK/STAT pathway for nuclear signal transduction, are themselves translocated to the nucleus. The exact mechanism for the nuclear import of IFN-gamma or the functional role of the nuclear translocation of ligand in signal transduction is unknown. We show in this study that nuclear localization of IFN-gamma is driven by a simple polybasic nuclear localization sequence (NLS) in its COOH terminus, as verified by its ability to specify nuclear import of a heterologous protein allophycocyanin (APC) in standard import assays in digitonin-permeabilized cells. Similar to other nuclear import signals, we show that a peptide representing amino acids 95-132 of IFN-gamma (IFN-gamma(95-132)) containing the polybasic sequence 126RKRKRSR132 was capable of specifying nuclear uptake of the autofluorescent protein, APC, in an energy-dependent fashion that required both ATP and GTP. Nuclear import was abolished when the above polybasic sequence was deleted. Moreover, deletions immediately NH2-terminal of this sequence did not affect the nuclear import. Thus, the sequence 126RKRKRSR132 is necessary and sufficient for nuclear localization. Furthermore, nuclear import was strongly blocked by competition with the cognate peptide IFN-gamma(95-132) but not the peptide IFN-gamma(95-125), which is deleted in the polybasic sequence, further confirming that the NLS properties were contained in this sequence. A peptide containing the prototypical polybasic NLS sequence of the SV40 large T-antigen was also able to inhibit the nuclear import mediated by IFN-gamma(95-132). This observation suggests that the NLS in IFN-gamma may function through the components of the Ran/importin pathway utilized by the SV40 T-NLS. Finally, we show that intact IFN-gamma, when coupled to APC, was also able to mediate its nuclear import. Again, nuclear import was blocked by the peptide IFN-gamma(95-132) and the SV40 T-NLS peptide, suggesting that intact IFN-gamma was also transported into the nucleus through the Ran/importin pathway. Previous studies have suggested a direct intracellular role for IFN-gamma in the induction of its biological activities. Based on our data in this study, we suggest that a key intracellular site of interaction of IFN-gamma is the one with the nuclear transport mechanism that occurs via the NLS in the COOH terminus of IFN-gamma.


Asunto(s)
Interferón gamma/metabolismo , Señales de Localización Nuclear , Células 3T3 , Secuencia de Aminoácidos , Animales , Antígenos Transformadores de Poliomavirus/química , Interferón gamma/química , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular
7.
J Interferon Cytokine Res ; 19(12): 1351-61, 1999 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-10638704

RESUMEN

Interferons (IFN) inhibit the growth of tumor cells by blocking the progression of their cell cycle. Recently, we showed that this cell cycle inhibition correlates with the ability of IFN to upregulate the cyclin-dependent kinase inhibitor p21(WAF1). This, however, is not proof of a causal relationship. Using p21(WAF1)-deficient cells derived from the HCT116 colon adenocarcinoma cell line, we now show that p21(WAF1) is indeed responsible for the antiproliferative effects of the type II IFN, IFN-gamma. IFN-gamma upregulated p21(WAF1) expression in a p53-independent manner, decreased cyclin-dependent kinase 2 activity, and inhibited entry into the S phase of the cell cycle in p21+/+ but not in p21-/- HCT116 cells. We additionally found that the lack of p21(WAF1) expression resulted in an increase in the ability of IFN-gamma to induce apoptosis, as reflected by an earlier induction of DNA fragmentation and caspase 3 activity in p21-/- cell. Our results indicate that p21(WAF1) expression is necessary for IFN-gamma-mediated cell cycle inhibition and suppression of IFN-gamma-induced apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Quinasas CDC2-CDC28 , Ciclo Celular/efectos de los fármacos , Ciclinas/biosíntesis , Interferón gamma/farmacología , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Quinasa 2 Dependiente de la Ciclina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/deficiencia , Ciclinas/genética , Fragmentación del ADN/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Marcación de Gen , Genes p53 , Humanos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Recombinantes , Células Tumorales Cultivadas
8.
Alcohol Clin Exp Res ; 22(5 Suppl): 188S-192S, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9727632

RESUMEN

Initial infection with human immunodeficiency virus (HIV) results in a burst of viremia and an ensuing spread of virus to secondary lymphoid organs, after which a "latency" period occurs with little or no virus detectable in the circulation. The term latent period has been shown to be a misnomer, because substantial viral replication occurs during this time in lymph nodes, although clinically there appears to be few symptoms of disease. However, a telling indicator of active infection during this period is the initiation of decline in CD4+ T-cell numbers. A number of hypotheses have been postulated for the mechanism(s), as of yet not fully elucidated, by which T cells are depleted. Although quiescent cells can be infected, it has been shown that replication of HIV in CD4+ T cells requires cellular activation. The levels of viremia early in infection indicate that a large number of cells are actively infected, further suggesting that a mechanism must exist by which HIV activates a large pool of cells and ultimately causes their depletion. One possible mechanism for activation would be the presence of an HIV-encoded superantigen. Superantigens are proteins that are polyclonal stimulators of CD4+ T lymphocytes. This occurs as a result of their ability to form a trimolecular complex with MHC class II molecules on antigen-presenting cells and the Vbeta-specific region on the T-cell receptor. Thus, superantigen activation of T cells is antigen-nonspecific. The prototype superantigens are the staphylococcal enterotoxins. Putative viral superantigens include a protein from mouse mammary tumor virus and related retroviruses, rabies nucleocapsid, and the Nef protein of HIV. Nef is required for optimal HIV pathogenesis, and this may be due to its superantigen properties, where CD4 cells are transformed to the activated state for virus replication.


Asunto(s)
Infecciones por VIH/inmunología , VIH/patogenicidad , Superantígenos/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Productos del Gen nef/fisiología , VIH/inmunología , Humanos , Ratones , Virulencia , Productos del Gen nef del Virus de la Inmunodeficiencia Humana
9.
Int J Cancer ; 77(1): 138-45, 1998 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-9639405

RESUMEN

Type I and type II interferons (IFNs) are known to exert antitumor effects on a variety of tissues and cell types. We have previously shown that the type I IFN IFN alpha induces the expression of the cyclin-dependent kinase inhibitor p21WAF1 and inhibits the cell cycle of the human prostate adenocarcinoma cell line, DU145, that carries mutations in the tumor suppressor gene products p53 and pRB. We now show that the type II IFN IFN gamma similarly induces the expression of p21WAF1 and inhibits the cell cycle of DU145 cells. In addition, we show that while both IFNs exert antiproliferative activity, only IFN gamma induced phenotypic changes in these cells that accompanied the antiproliferative effect. For example, IFN gamma, but not IFN alpha, caused a significant reduction in epidermal growth factor receptor expression as well as an increase in the adhesion molecules intercellular adhesion molecule-1 and integrin alpha3. These phenotypic changes in DU145 cells are suggestive of the acquisition of a non-tumorigenic state. Consistent with these findings, IFN gamma showed a significantly lower invasive ability in in vitro assays using invasion chambers. Thus, IFN gamma inhibits both the cell cycle and the metastatic potential of DU145 cells independent of the p53 and RB status, and our data describe a mechanism for mediating the antitumor capabilities of IFN gamma that bypasses tumor suppressor genes like p53.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Ciclinas/biosíntesis , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Interferón gamma/farmacología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Inducción Enzimática/efectos de los fármacos , Humanos , Interferón-alfa/farmacología , Masculino , Invasividad Neoplásica , Neoplasias de la Próstata/metabolismo , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas
10.
Oncogene ; 16(14): 1885-90, 1998 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-9583686

RESUMEN

We show, in this study, that type I IFN induction of the cyclin-dependent kinase (cdk) inhibitor p21WAF1 in the human Burkitt lymphoma B cell-line Daudi and ensuing cell cycle arrest correlate with the terminal differentiation of these cells, and is ultimately followed by apoptosis and cell death. The expression of p21WAF1 paralleled the onset of G1 arrest and the reduction of surface IgM expression which was used as a marker of the differentiation response, and the IFN treated cells acquired a typical plasma cell-like morphology. The type II IFN IFNgamma, which does not inhibit the growth of Daudi cells, did not induce the expression of p21WAF1, nor affect the expression of surface IgM. The induction of p21WAF1 which paralleled the inhibition of the phosphorylation of the retinoblastoma protein, pRB, was preceded by the strong reduction in c-myc levels. We propose that the coupled down-regulation of c-myc and induction of p21WAF1 may be crucial to the induction of differentiation and G1 arrest in Daudi cells by type I IFN. Growth arrest and differentiation was followed by apoptosis and cell death, and was accompanied by the induction of the activity of the apoptotic ICE-family protease CPP32. G1 arrest and differentiation followed by apoptotic cell death are characteristics of terminal differentiation. Thus, our data suggest that the induction of p21WAF1 and G1 arrest mediated by type I IFN in Daudi cells is part of terminal differentiation response in these cells, highlighting a role for type I IFN as B cell terminal differentiation factors.


Asunto(s)
Apoptosis/efectos de los fármacos , Linfocitos B/patología , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Ciclinas/biosíntesis , Fase G1/efectos de los fármacos , Interferón Tipo I/farmacología , Apoptosis/genética , Linfocitos B/efectos de los fármacos , Linfocitos B/enzimología , Linfoma de Burkitt , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/fisiología , Fase G1/genética , Humanos , Células Tumorales Cultivadas
11.
Oncogene ; 17(23): 2973-9, 1998 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-9881699

RESUMEN

Glioblastoma is a highly aggressive form of brain cancer characterized by uncontrolled cell growth resulting from a loss of cell cycle regulation. In this study we determined the antiproliferative effects of interferon gamma (IFNgamma) on the glioblastoma cell lines T98G, SNB-19 and U-373, focusing on the ability of IFNgamma to increase levels of p21WAF1/CIP1, an important negative regulator of cell cycle events. IFNgamma was found to inhibit the growth of all cell lines, with inhibition ranging from 82.2% to 45.4%. Flow cytometry analysis showed that IFNgamma treatment caused a cell cycle delay in the G1 or S phases. The strength of this delay varied, correlating with the degree by which IFNgamma inhibited proliferation of each cell line. IFNgamma treatment increased the production of the cyclin dependent kinase inhibitor (CKI) p21WAF1/ CIP1 in all cell lines, the level and kinetics of production of which correlated with the degree and stage of inhibition of cellular proliferation. Further, immunoprecipitation of p21WAF1/CIP1 in complexes of p21WAF1/CIP1/cyclin-dependent kinase 2 (cdk2)/cyclin showed that the amount of p21WAF1/CIP1 in the complexes and the inhibition of cdk2-cyclin kinase activity correlated with the level of p21WAF1/CIP1 produced in the cells by IFNgamma. These results show that IFNgamma has significant antiproliferative effects on the glioblastoma cell lines and suggest that p21WAF1/CIP1 plays a role in mediating these effects.


Asunto(s)
Quinasas CDC2-CDC28 , Ciclinas/metabolismo , Inhibidores de Crecimiento/metabolismo , Interferón gamma/metabolismo , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/metabolismo , Glioblastoma , Inhibidores de Crecimiento/farmacología , Humanos , Interferón gamma/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Células Tumorales Cultivadas
12.
Oncogene ; 14(10): 1165-70, 1997 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-9121765

RESUMEN

Prostate cancer, like other types of cancer, is associated with the loss of cell cycle control, resulting in unregulated growth of cells. We report here on the inhibitory effects of interferon alpha (IFN alpha) on the cell cycle of prostate cancer cells, using the human prostate carcinoma cell line DU145 that has mutations in the tumor suppressor genes pRB, p53 and KAI1. IFN alpha inhibited growth and colony formation of DU145 cells and analysis by flow cytometry suggests that IFN alpha inhibited the progression of these cancer cells from the G1 through S phase of the cell cycle. IFN alpha treatment of DU145 cells reduced cyclin dependent kinase 2 (cdk2) activity. In particular, cyclin E dependent cdk2 activity was inhibited by IFN alpha treatment. IFN alpha treatment, however, did not affect the amount of cdk2 bound to cyclin E. Consistent with this data, IFN alpha was able to induce expression of the kinase inhibitor p21 in DU145 cells. Furthermore, IFN treatment increased the amounts of p21 complexed with cdk2 in these cells. These data support a role for p21 in mediating the antiproliferative action of IFN alpha. The induction of p21 and its growth inhibitory effects in DU145 cells appears independent of p53, pRB and KAI1 status.


Asunto(s)
Antineoplásicos/farmacología , Quinasas CDC2-CDC28 , Ciclinas/biosíntesis , Interferón-alfa/farmacología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/terapia , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/efectos de los fármacos , Quinasas Ciclina-Dependientes/metabolismo , Citometría de Flujo , Genes Supresores de Tumor , Humanos , Masculino , Mutación , Neoplasias de la Próstata/patología , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo
13.
Biochem Biophys Res Commun ; 230(3): 509-13, 1997 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-9015351

RESUMEN

We previously reported that Nef protein from human immunodeficiency virus (HIV) has superantigen properties. However, we were unable to consistently demonstrate V beta-specific expansion by Nef using flow cytometry, possibly due to its lower mitogenic activity compared to prototypic superantigens. Therefore, we developed a simple amplification detection method using immobilized anti-V beta antibodies. Human peripheral blood mononuclear cells from adult donors were treated for 24 h with Nef and restimulated with immobilized anti-V beta antibodies for an additional 72 h. Significant expansion of V beta 5.3 and V beta 18 T-cells were detected in Nef-treated cultures, with expansion of V beta 18 occurring with all donors tested, and V beta 5.3 expansion occurring in 50% of the donors. Variable responses were obtained with V beta 2, V beta 3, and V beta 9. These results were confirmed using the more time-consuming method of reverse transcriptase-polymerase chain reaction (RT-PCR). Thus, this novel, reproducible, and relatively simple method can detect V beta-specific expansion by weak superantigens.


Asunto(s)
Productos del Gen nef/fisiología , VIH/fisiología , Reacción en Cadena de la Polimerasa/métodos , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Adulto , Productos del Gen nef/inmunología , VIH/inmunología , Humanos , Activación de Linfocitos/efectos de los fármacos , Reproducibilidad de los Resultados , Superantígenos/inmunología , Superantígenos/fisiología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana
14.
J Interferon Cytokine Res ; 17(1): 11-5, 1997 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9041466

RESUMEN

Type I interferons (IFN), such as IFN-alpha, are potent antiproliferative and antitumor agents. IFN-tau, originally identified as a pregnancy recognition hormone, is a type I IFN that is related to IFN-alpha. We examine here the mechanism of the antiproliferative effects of IFN-alpha and IFN-tau in terms of their effects on intracellular events that regulate the cell cycle. Both IFN inhibited proliferation of the human Burkitt lymphoma cell line, Daudi, causing accumulation of cells in the G1 phase of the cell cycle. IFN-alpha was more effective than IFN-tau in this regard. Both IFN were found to inhibit the kinase activity of the cyclin-dependent kinase cdk2 in a manner that correlated with their relative abilities to cause cells to accumulate in the G1 phase of the cell cycle. Further, IFN treatment did not affect the expression of cdk2 protein, suggesting that the IFN modulated cdk2 activity through a cdk inhibitor. Consistent with this conclusion, both IFN induced the expression of the cyclin-dependent kinase inhibitor protein p21. The levels of p21 induced also correlated with the relative abilities of the IFN to inhibit cdk2 activity and to arrest cell growth in the G1 phase of the cell cycle. Moreover, following IFN treatment, increased levels of p21 were found complexed with cdk2, consistent with its role in the inhibition of cdk2 activity. These data suggest that p21-mediated inhibition of cdk2 activity plays an important role in the antiproliferative activity of type I IFN. The findings highlight interesting similarities between these cytokines and the products of tumor suppressor genes, such as p53, and may indicate a mechanism for the antitumor effects of the type I IFN.


Asunto(s)
Antineoplásicos/farmacología , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Ciclinas/fisiología , Inhibidores Enzimáticos/metabolismo , Interferón Tipo I , Interferón-alfa/farmacología , Interferón gamma/farmacología , Proteínas Gestacionales/farmacología , Linfoma de Burkitt/tratamiento farmacológico , Linfoma de Burkitt/patología , Proteína Quinasa CDC2/antagonistas & inhibidores , División Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Fase G1/efectos de los fármacos , Humanos , Células Tumorales Cultivadas
15.
J Immunol ; 155(12): 5617-22, 1995 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-7499845

RESUMEN

The tyrosine kinase JAK2 is an integral part of the signal transduction pathways of a number of cytokines and growth factors, including IFN-gamma. Previously, we identified a species-nonspecific binding site for the C terminus of IFN-gamma, encompassed by IFN-gamma peptide IFN-gamma(95-133), on the membrane proximal region of the cytoplasmic domain of the IFN-gamma R alpha-chain. Using both a radioligand binding assay and coimmunoprecipitation with antireceptor antiserum, we were able to demonstrate specific interaction of JAK2 with the murine IFN-gamma R(MIR) alpha-chain. Furthermore, this interaction is increased by the addition of murine IFN-gamma or its C-terminal peptide, muIFN-gamma(95-133). We also identified two regions of the cytoplasmic domain of the receptor that interact with JAK2 using synthetic peptides of the MIR alpha-chain in receptor competition studies. These regions are encompassed by receptor peptide MIR(283-309), which is adjacent to the membrane proximal region at which the C terminus of IFN-gamma binds, and receptor peptide MIR(404-432), which lies near the C terminus of the receptor, encompassing a potentially important phosphorylation site. These data show site-specific interaction between JAK2 and IFN-gamma with the IFN-gamma R and have broader implications for the role of the IFN-gamma ligand in the IFN-gamma signal transduction pathway. Furthermore, the data support previous studies that demonstrated that intracellular IFN-gamma plays a role in cell activation.


Asunto(s)
Interferón gamma/farmacología , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas , Receptores de Interferón/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Células Cultivadas , Janus Quinasa 2 , Ratones , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Fragmentos de Péptidos/farmacología , Unión Proteica/efectos de los fármacos , Proteínas Recombinantes/metabolismo , Regulación hacia Arriba/efectos de los fármacos
16.
Proc Natl Acad Sci U S A ; 92(26): 12270-4, 1995 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-8618883

RESUMEN

Interferon tau (IFN tau), originally identified as a pregnancy recognition hormone, is a type I interferon that is related to the various IFN alpha species (IFN alpha s). Ovine IFN tau has antiviral activity similar to that of human IFN alpha A on the Madin-Darby bovine kidney (MDBK) cell line and is equally effective in inhibiting cell proliferation. In this study, IFN tau was found to differ from IFN alpha A in that is was > 30-fold less toxic to MDBK cells at high concentrations. Excess IFN tau did not block the cytotoxicity of IFN alpha A on MDBK cells, suggesting that these two type I IFNs recognize the type I IFN receptor differently on these cells. In direct binding studies, 125I-IFN tau had a Kd of 3.90 x 10(-10) M for receptor on MDBK cells, whereas that of 125I-IFN alpha A was 4.45 x 10(-11) M. Consistent with the higher binding affinity, IFN alpha A was severalfold more effective than IFN tau in competitive binding against 125I-IFN tau to receptor on MDBK cells. Paradoxically, the two IFNs had similar specific antiviral activities on MDBK cells. However, maximal IFN antiviral activity required only fractional occupancy of receptors, whereas toxicity was associated with maximal receptor occupancy. Hence, IFN alpha A, with the higher binding affinity, was more toxic than IFN tau. The IFNs were similar in inducing the specific phosphorylation of the type I receptor-associated tyrosine kinase Tyk2, and the transcription factors Stat1 alpha and Stat2, suggesting that phosphorylation of these signal transduction proteins is not involved in the cellular toxicity associated with type I IFNs. Experiments using synthetic peptides suggest that differences in the interaction at the N terminal of IFN tau and IFN alpha with the type I receptor complex contribute significantly to differences in high-affinity equilibrium binding of these molecules. It is postulated that such a differential recognition of the receptor is responsible for the similar antiviral but different cytotoxic effects of these IFNs. Moreover, these data imply that receptors are "spare'' with respect to certain biological properties, and we speculate that IFNs may induce a concentration-dependent selective association of receptor subunits.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Interferón Tipo I/metabolismo , Interferón Tipo I/toxicidad , Proteínas Gestacionales/metabolismo , Proteínas Gestacionales/toxicidad , Proteínas Tirosina Quinasas , Receptores de Interferón/metabolismo , Animales , Unión Competitiva , Linfoma de Burkitt , Bovinos , Línea Celular , Proteínas de Unión al ADN/aislamiento & purificación , Proteínas de Unión al ADN/metabolismo , Humanos , Riñón , Cinética , Proteínas de la Membrana , Fosforilación , Proteínas/metabolismo , Receptor de Interferón alfa y beta , Proteínas Recombinantes , Factor de Transcripción STAT1 , Factor de Transcripción STAT2 , TYK2 Quinasa , Transactivadores/aislamiento & purificación , Transactivadores/metabolismo , Células Tumorales Cultivadas
17.
J Immunol ; 155(5): 2747-53, 1995 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-7544384

RESUMEN

Multiple sclerosis (MS) is an inflammatory demyelinating autoimmune disease if the central nervous system (CNS). Recently, the type I IFN, IFN-beta-1b was demonstrated to be a useful immunotherapy for MS. During treatment with IFN-beta-1b, toxicity at higher doses has been observed. IFN-tau, discovered for its role in the reproductive cycle, possesses all of the functions normally ascribed to the type I IFNs but lacks the toxicity normally associate with IFN treatment in vitro. We have examined the effects of IFN-tau treatment on experimental allergic encephalomyelitis (EAE), an animal model useful for the study of MS. EAE is a model of Ag-induced autoimmunity that can be modulated by bacterial superantigen to resemble the relapsing-remitting pattern of autoimmune disease observed in MS. IFN-tau was able to prevent development of EAE as effectively as IFN-beta but without associated toxicity such as lymphocyte suppression and weight loss. In addition, IFN-tau was able to prevent superantigen reactivation of EAE akin to the reduction in disease exacerbations observed in IFN-beta-1b treated MS patients. Mechanisms by which IFN-tau may prevent EAE include reduced proliferation in response to the autoantigen myelin basic protein and reduced TNF-alpha production. Thus, IFN-tau may prove to be a promising new IFN therapy for MS in light of its ability to prevent EAE and the lack of toxicity exhibited by this novel IFN.


Asunto(s)
Encefalomielitis Autoinmune Experimental/prevención & control , Interferón Tipo I/farmacología , Interferón Tipo I/toxicidad , Proteínas Gestacionales/farmacología , Proteínas Gestacionales/toxicidad , Superantígenos/efectos de los fármacos , Animales , Encefalomielitis Autoinmune Experimental/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos , Proteína Básica de Mielina/efectos de los fármacos , Proteína Básica de Mielina/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Ovinos , Superantígenos/inmunología , Subgrupos de Linfocitos T/inmunología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA