Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
FEBS Open Bio ; 13(8): 1495-1506, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37151134

RESUMEN

We have recently shown that IFNγ, produced during cancer therapy, induces expression of the Bcl3 proto-oncogene in ovarian cancer (OC) cells, resulting in their increased proliferation, migration, and invasion, but the mechanisms are unknown. Here, we demonstrate that the IFNγ-induced Bcl3 expression is dependent on JAK1 and STAT1 signaling, and on p65 NFκB. Furthermore, the IFNγ-induced Bcl3 expression is associated with an increased occupancy of Ser-727 phosphorylated STAT1 and acetylated histone H3 at the Bcl3 promoter. Our data indicate that Bcl3 promotes expression of the pro-inflammatory chemokine interleukin-8 (IL-8) in OC cells. These findings identify Bcl3 as a novel target of IFNγ/JAK1/STAT1 signaling and suggest that targeting the JAK1/STAT1 pathway may suppress IFNγ-induced Bcl3 expression in OC.


Asunto(s)
Interleucina-8 , Neoplasias Ováricas , Humanos , Femenino , Interleucina-8/metabolismo , Transducción de Señal , FN-kappa B/metabolismo , Interferón gamma/farmacología , Interferón gamma/metabolismo , Neoplasias Ováricas/genética , Factor de Transcripción STAT1/metabolismo , Janus Quinasa 1/genética , Janus Quinasa 1/metabolismo
2.
Cell Signal ; 97: 110400, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35820543

RESUMEN

Expression of the immune checkpoint programmed death ligand-1 (PD-L1) is increased in ovarian cancer (OC) and correlates with poor prognosis. Interferon-γ (IFNγ) induces PD-L1 expression in OC cells, resulting in their increased proliferation and tumor growth, but the mechanisms that regulate the PD-L1 expression in OC remain unclear. Here, we show that the IFNγ-induced PD-L1 expression in OC cells is associated with increased levels of STAT1, Tyr-701 pSTAT1 and Ser-727 pSTAT1. Suppression of JAK1 and STAT1 significantly decreases the IFNγ-induced PD-L1 expression in OC cells, and STAT1 overexpression increases the IFNγ-induced PD-L1 expression. In addition, IFNγ induces expression of the transcription factor interferon regulatory factor 1 (IRF1) and IRF1 suppression attenuates the IFNγ-induced gene and protein levels of PD-L1. Chromatin immunoprecipitation results show that IFNγ induces PD-L1 promoter acetylation and recruitment of STAT1, Ser-727 pSTAT1 and IRF1 in OC cells. Together, these findings demonstrate that the IFNγ-induced PD-L1 expression in OC cells is regulated by JAK1, STAT1, and IRF1 signaling, and suggest that targeting the JAK1/ STAT1/IRF1 pathway may provide a leverage to regulate the PD-L1 levels in ovarian cancer.


Asunto(s)
Antígeno B7-H1 , Neoplasias Ováricas , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Carcinoma Epitelial de Ovario , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Factor 1 Regulador del Interferón/genética , Factor 1 Regulador del Interferón/metabolismo , Interferón gamma/metabolismo , Interferón gamma/farmacología , Janus Quinasa 1/metabolismo , Neoplasias Ováricas/genética , Factor de Transcripción STAT1/metabolismo
3.
Int J Biochem Cell Biol ; 141: 106093, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34626802

RESUMEN

Interferon-γ (IFNγ) is a pleiotropic cytokine that has a crucial role in immune response and tumor immunity. Because of its anti-tumor effects, IFNγ has been used in cancer treatment. However, IFNγ also has tumor-promoting functions that are less well understood. Here, we show that IFNγ induces expression of the pro-inflammatory and pro-angiogenic chemokine interleukin-8 (IL-8, CXCL8) in ovarian cancer (OC) cells. The IFNγ-induced IL-8 expression is dependent on JAK1, STAT1, and p65 NFκB, and is associated with an increased occupancy of K314/315 acetylated p65 NFκB and Ser-727 phosphorylated STAT1 at the IL-8 promoter. Neutralization of IL-8 using anti-IL-8 antibody reduces IFNγ-induced migration of OC cells, and their invasion ability in 3D spheroids. Together, these findings identify IL-8 as a novel target induced by IFNγ/JAK1/STAT1/p65 NFκB signaling, and indicate that the IFNγ-induced IL-8 contributes to IFNγ pro-tumorigenic effects in ovarian cancer cells.


Asunto(s)
Neoplasias Ováricas , Femenino , Humanos , Interferón gamma , Interleucina-8 , Factor de Transcripción STAT1
4.
Methods Mol Biol ; 2108: 211-220, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31939183

RESUMEN

Expression of the immune checkpoint programmed death ligand 1 (PD-L1, CD274) is increased in many types of cancer, including ovarian cancer (OC), but the mechanisms that regulate the PD-L1 expression are not fully understood. In addition to binding to PD-1 on T cells, thus inhibiting T cell-mediated antitumor responses, PD-L1 has also tumor-intrinsic effects that include increased cancer cell survival and proliferation, and that might be in part mediated by the intracellular PD-L1. In this chapter, we describe a protocol for the analysis of the intracellular PD-L1 protein levels in OC cells by immunoblotting. Our results show that interferon-γ (IFNγ) induces the intracellular levels of PD-L1 and the proto-oncogene Bcl3 in OC cells. However, the PD-L1 expression is significantly decreased in OC cells stably transfected with Bcl3 shRNA, demonstrating that the IFNγ-induced PD-L1 expression in OC cells is mediated by Bcl3. These data identify the IFNγ-Bcl3-PD-L1 axis as a novel therapeutic target in OC, and suggest that targeting Bcl3 may provide a novel strategy to regulate the PD-L1 expression, and especially the tumor-intrinsic PD-L1 effects mediated by the intracellular PD-L1 in OC cells.


Asunto(s)
Proteínas del Linfoma 3 de Células B/genética , Antígeno B7-H1/metabolismo , Immunoblotting , Interferón gamma/metabolismo , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , ARN Interferente Pequeño/genética , Proteínas del Linfoma 3 de Células B/metabolismo , Antígeno B7-H1/genética , Línea Celular Tumoral , Femenino , Expresión Génica , Humanos , Immunoblotting/métodos , Interferón gamma/farmacología , Neoplasias Ováricas/patología , Proto-Oncogenes Mas , Transfección
5.
Methods Mol Biol ; 2108: 221-228, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31939184

RESUMEN

Expression of programmed death ligand-1 (PD-L1, CD274) on cancer cells is regulated by interferon-γ (IFNγ) signaling as well as by epigenetic mechanisms. By binding to PD-1 on cytotoxic T cells, PD-L1 inhibits T cell-mediated antitumor responses, resulting in immune escape. This chapter describes analysis of the surface PD-L1 expression in ovarian cancer (OC) cells using flow cytometry (FC). Our data demonstrate that the surface PD-L1 expression in OC cells is induced by IFNγ as well as by the class I histone deacetylase (HDAC) inhibition by romidepsin, suggesting that class I HDAC inhibition might provide a useful strategy to modulate the PD-L1 levels on OC cells.


Asunto(s)
Antígeno B7-H1/metabolismo , Membrana Celular/metabolismo , Depsipéptidos/farmacología , Citometría de Flujo , Interferón gamma/metabolismo , Neoplasias Ováricas/metabolismo , Antígeno B7-H1/genética , Línea Celular Tumoral , Separación Celular/métodos , Análisis de Datos , Femenino , Citometría de Flujo/métodos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Interferón gamma/farmacología , Neoplasias Ováricas/genética
6.
Methods Mol Biol ; 2108: 229-239, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31939185

RESUMEN

The immune checkpoint molecule, programmed death ligand 1 (PD-L1; B7-H1, CD274), induces T cell apoptosis and tolerance, thus inhibiting the antitumor immunity. PD-L1 expression is increased in many types of cancer, including ovarian cancer (OC), and correlates with poor prognosis. However, the mechanisms that regulate the PD-L1 expression in cancer cells are incompletely understood. The transcriptional regulation of PD-L1 expression is orchestrated by several transcription factors, including NFκB. The human PD-L1 promoter contains five NFκB-binding sites. Interferon-γ (IFNγ) stimulation of OC cells induces p65, and particularly K314/315 acetylated p65 recruitment to all five NFκB-binding sites in PD-L1 promoter, resulting in increased PD-L1 expression. In this chapter, we describe a protocol that uses chromatin immunoprecipitation (ChIP) to analyze the transcriptional regulation of PD-L1 by measuring recruitment of NFκB p65 and K314/315 acetylated p65 to PD-L1 promoter in human OC cells.


Asunto(s)
Antígeno B7-H1/genética , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Transcripción Genética , Antígeno B7-H1/metabolismo , Sitios de Unión , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Inmunoprecipitación de Cromatina/métodos , Femenino , Humanos , Interferón gamma/metabolismo , FN-kappa B/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Reacción en Cadena en Tiempo Real de la Polimerasa
7.
J Biol Chem ; 293(40): 15483-15496, 2018 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-30135206

RESUMEN

The proto-oncogene Bcl3 induces survival and proliferation in cancer cells; however, its function and regulation in ovarian cancer (OC) remain unknown. Here, we show that Bcl3 expression is increased in human OC tissues. Surprisingly, however, we found that in addition to promoting survival, proliferation, and migration of OC cells, Bcl3 promotes both constitutive and interferon-γ (IFN)-induced expression of the immune checkpoint molecule PD-L1. The Bcl3 expression in OC cells is further increased by IFN, resulting in increased PD-L1 transcription. The mechanism consists of an IFN-induced, Bcl3- and p300-dependent PD-L1 promoter occupancy by Lys-314/315 acetylated p65 NF-κB. Blocking PD-L1 by neutralizing antibody reduces proliferation of OC cells overexpressing Bcl3, suggesting that the pro-proliferative effect of Bcl3 in OC cells is partly mediated by PD-L1. Together, this work identifies PD-L1 as a novel target of Bcl3, and links Bcl3 to IFNγ signaling and PD-L1-mediated immune escape.


Asunto(s)
Antígeno B7-H1/genética , Puntos de Control del Ciclo Celular/inmunología , Células Epiteliales/inmunología , Regulación Neoplásica de la Expresión Génica , Proteínas Proto-Oncogénicas/genética , Factores de Transcripción/genética , Escape del Tumor/genética , Anticuerpos Neutralizantes/farmacología , Apoptosis/efectos de los fármacos , Proteínas del Linfoma 3 de Células B , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/inmunología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proteína p300 Asociada a E1A , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Femenino , Humanos , Interferón gamma/farmacología , Ovario/inmunología , Ovario/patología , Regiones Promotoras Genéticas , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/inmunología , Transducción de Señal , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/inmunología , Factores de Transcripción/inmunología , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...