Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
J Biol Chem ; 289(10): 6581-6591, 2014 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-24469448

RESUMEN

We provide evidence that type I IFN-induced STAT activation is diminished in cells with targeted disruption of the Rictor gene, whose protein product is a key element of mTOR complex 2. Our studies show that transient or stable knockdown of Rictor or Sin1 results in defects in activation of elements of the STAT pathway and reduced STAT-DNA binding complexes. This leads to decreased expression of several IFN-inducible genes that mediate important biological functions. Our studies also demonstrate that Rictor and Sin1 play essential roles in the generation of the suppressive effects of IFNα on malignant erythroid precursors from patients with myeloproliferative neoplasms. Altogether, these findings provide evidence for critical functions for Rictor/Sin1 complexes in type I IFN signaling and the generation of type I IFN antineoplastic responses.


Asunto(s)
Antineoplásicos/farmacología , Proteínas Portadoras/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Interferón Tipo I/farmacología , Transcripción Genética/efectos de los fármacos , Animales , Proteínas Portadoras/genética , Células Cultivadas , Fibroblastos/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/patología , Humanos , Ratones , Fosforilación , Policitemia Vera/metabolismo , Policitemia Vera/patología , Proteína Asociada al mTOR Insensible a la Rapamicina , Transducción de Señal
2.
Blood ; 121(18): 3675-81, 2013 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-23509154

RESUMEN

Mnk kinases regulate the phosphorylation and activation of the eukaryotic initiation factor 4E (eIF4E), a protein that plays key roles in the initiation of messenger RNA translation and whose activity is critical for various cellular functions. eIF4E is deregulated in acute myeloid leukemia (AML), and its aberrant activity contributes to leukemogenesis. We determined whether cercosporamide, an antifungal agent that was recently shown to act as a unique Mnk inhibitor, exhibits antileukemic properties. Treatment of AML cells with cercosporamide resulted in a dose-dependent suppression of eIF4E phosphorylation. Such suppression of Mnk kinase activity and eIF4E phosphorylation by cercosporamide resulted in dose-dependent suppressive effects on primitive leukemic progenitors (CFU-L) from AML patients and enhanced the antileukemic properties of cytarabine (Ara-C) or mammalian target of rapamycin (mTOR) complex 1 inhibition. Similarly, the combination of cercosporamide with cytarabine resulted in enhanced antileukemic responses in a xenograft mouse model in vivo. Altogether, this work demonstrates that the unique Mnk inhibitor cercosporamide suppresses phosphorylation of eIF4E and exhibits antileukemic effects, in support of future clinical-translational efforts involving combinations of Mnk inhibitors with cytarabine and/or mTOR inhibitors for the treatment of AML.


Asunto(s)
Adenosina Trifosfatasas/antagonistas & inhibidores , Antineoplásicos/uso terapéutico , Benzofuranos/uso terapéutico , Proteínas de Transporte de Catión/antagonistas & inhibidores , Leucemia Mieloide Aguda/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , ATPasas Transportadoras de Cobre , Regulación hacia Abajo/efectos de los fármacos , Humanos , Células K562 , Ratones , Células Madre Neoplásicas/efectos de los fármacos , Células U937 , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Proc Natl Acad Sci U S A ; 109(20): 7723-8, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22550181

RESUMEN

IFNs transduce signals by binding to cell surface receptors and activating cellular pathways and regulatory networks that control transcription of IFN-stimulated genes (ISGs) and mRNA translation, leading to generation of protein products that mediate biological responses. Previous studies have shown that type I IFN receptor-engaged pathways downstream of AKT and mammalian target of rapamycin complex (mTORC) 1 play important roles in mRNA translation of ISGs and the generation of IFN responses, but the roles of mTORC2 complexes in IFN signaling are unknown. We provide evidence that mTORC2 complexes control IFN-induced phosphorylation of AKT on serine 473 and their function is ultimately required for IFN-dependent gene transcription via interferon-stimulated response elements. We also demonstrate that such complexes exhibit regulatory effects on other IFN-dependent mammalian target of rapamycin-mediated signaling events, likely via engagement of the AKT/mTORC1 axis, including IFN-induced phosphorylation of S6 kinase and its effector rpS6, as well as phosphorylation of the translational repressor 4E-binding protein 1. We also show that induction of ISG protein expression and the generation of antiviral responses are defective in Rictor and mLST8-KO cells. Together, our data provide evidence for unique functions of mTORC2 complexes in the induction of type I IFN responses and suggest a critical role for mTORC2-mediated signals in IFN signaling.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Interferones/metabolismo , Complejos Multiproteicos/metabolismo , Transducción de Señal/inmunología , Serina-Treonina Quinasas TOR/metabolismo , Transactivadores/metabolismo , Animales , Proteínas Portadoras/genética , Células HeLa , Humanos , Immunoblotting , Interferones/inmunología , Luciferasas , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína Asociada al mTOR Insensible a la Rapamicina , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína S6 Ribosómica/metabolismo , Proteínas Quinasas S6 Ribosómicas/metabolismo
4.
J Biol Chem ; 288(46): 33006-15, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24089532

RESUMEN

There is emerging evidence that the IFN-inducible family of Slfn genes and proteins play important roles in cell cycle progression and control of cellular proliferation, but the precise functional roles of different Slfn members in the regulation of tumorigenesis remain unclear. In the present study, we undertook a systematic analysis on the expression and functional relevance of different mouse Slfn genes in malignant melanoma and renal cell carcinoma cells. Our studies demonstrate that several mouse Slfn genes are up-regulated in response to IFN treatment of mouse melanoma and renal cell carcinoma cells, including Slfn1, Slfn2, Slfn4, Slfn5, and Slfn8. Our data show that Slfn2 and Slfn3 play essential roles in the control of mouse malignant melanoma cell proliferation and/or anchorage-independent growth, suggesting key and non-overlapping roles for these genes in the control of malignant melanoma tumorigenesis. In renal cell carcinoma cells, in addition to Slfn2 and Slfn3, Slfn5 also exhibits important antineoplastic effects. Altogether, our findings indicate important functions for distinct mouse Slfn genes in the control of tumorigenesis and provide evidence for differential involvement of distinct members of this gene family in controlling tumorigenesis. They also raise the potential of future therapeutic approaches involving modulation of expression of members of this family of genes in malignant melanoma and renal cell carcinoma.


Asunto(s)
Carcinoma de Células Renales/metabolismo , Proteínas de Ciclo Celular/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Renales/metabolismo , Melanoma/metabolismo , Proteínas de Neoplasias/metabolismo , Animales , Antivirales/farmacología , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Interferón gamma/farmacología , Neoplasias Renales/genética , Neoplasias Renales/patología , Melanoma/genética , Melanoma/patología , Ratones , Proteínas de Neoplasias/genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
5.
bioRxiv ; 2024 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-38712152

RESUMEN

Cancer progression is an evolutionary process driven by the selection of cells adapted to gain growth advantage. We present the first formal study on the adaptation of gene expression in subclonal evolution. We model evolutionary changes in gene expression as stochastic Ornstein-Uhlenbeck processes, jointly leveraging the evolutionary history of subclones and single-cell expression data. Applying our model to sublines derived from single cells of a mouse melanoma revealed that sublines with distinct phenotypes are underlined by different patterns of gene expression adaptation, indicating non-genetic mechanisms of cancer evolution. Interestingly, sublines previously observed to be resistant to anti-CTLA-4 treatment showed adaptive expression of genes related to invasion and non-canonical Wnt signaling, whereas sublines that responded to treatment showed adaptive expression of genes related to proliferation and canonical Wnt signaling. Our results suggest that clonal phenotypes emerge as the result of specific adaptivity patterns of gene expression.

6.
J Biol Chem ; 287(50): 42352-60, 2012 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-23074222

RESUMEN

Interferons (IFNs) have important antiviral and antineoplastic properties, but the precise mechanisms required for generation of these responses remain to be defined. We provide evidence that during engagement of the Type I IFN receptor (IFNR), there is up-regulation of expression of Sprouty (Spry) proteins 1, 2, and 4. Our studies demonstrate that IFN-inducible up-regulation of Spry proteins is Mnk kinase-dependent and results in suppressive effects on the IFN-activated p38 MAP kinase (MAPK), the function of which is required for transcription of interferon-stimulated genes (ISGs). Our data establish that ISG15 mRNA expression and IFN-dependent antiviral responses are enhanced in Spry1,2,4 triple knock-out mouse embryonic fibroblasts, consistent with negative feedback regulatory roles for Spry proteins in IFN-mediated signaling. In other studies, we found that siRNA-mediated knockdown of Spry1, Spry2, or Spry4 promotes IFN-inducible antileukemic effects in vitro and results in enhanced suppressive effects on malignant hematopoietic progenitors from patients with polycythemia vera. Altogether, our findings demonstrate that Spry proteins are potent regulators of Type I IFN signaling and negatively control induction of Type I IFN-mediated biological responses.


Asunto(s)
Interferón Tipo I/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Fosfoproteínas/metabolismo , Receptor de Interferón alfa y beta/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Interferón Tipo I/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Fosfoproteínas/genética , Policitemia Vera/genética , Policitemia Vera/metabolismo , Policitemia Vera/patología , Proteínas Serina-Treonina Quinasas , Receptor de Interferón alfa y beta/genética , Células U937 , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
Proc Natl Acad Sci U S A ; 107(28): 12469-74, 2010 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-20616057

RESUMEN

mTOR-generated signals play critical roles in growth of leukemic cells by controlling mRNA translation of genes that promote mitogenic responses. Despite extensive work on the functional relevance of rapamycin-sensitive mTORC1 complexes, much less is known on the roles of rapamycin-insensitive (RI) complexes, including mTORC2 and RI-mTORC1, in BCR-ABL-leukemogenesis. We provide evidence for the presence of mTORC2 complexes in BCR-ABL-transformed cells and identify phosphorylation of 4E-BP1 on Thr37/46 and Ser65 as RI-mTORC1 signals in primary chronic myelogenous leukemia (CML) cells. Our studies establish that a unique dual mTORC2/mTORC1 inhibitor, OSI-027, induces potent suppressive effects on primitive leukemic progenitors from CML patients and generates antileukemic responses in cells expressing the T315I-BCR-ABL mutation, which is refractory to all BCR-ABL kinase inhibitors currently in clinical use. Induction of apoptosis by OSI-027 appears to negatively correlate with induction of autophagy in some types of BCR-ABL transformed cells, as shown by the induction of autophagy during OSI-027-treatment and the potentiation of apoptosis by concomitant inhibition of such autophagy. Altogether, our studies establish critical roles for mTORC2 and RI-mTORC1 complexes in survival and growth of BCR-ABL cells and suggest that dual therapeutic targeting of such complexes may provide an approach to overcome leukemic cell resistance in CML and Ph+ ALL.


Asunto(s)
Proteínas de Fusión bcr-abl/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Apoptosis/efectos de los fármacos , Apoptosis/genética , Estructuras Celulares/metabolismo , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Proteínas de Fusión bcr-abl/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/farmacología , Péptidos y Proteínas de Señalización Intracelular/uso terapéutico , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Mutación/efectos de los fármacos , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/uso terapéutico , Transducción de Señal/genética , Sirolimus/farmacología , Sirolimus/uso terapéutico , Serina-Treonina Quinasas TOR
8.
bioRxiv ; 2023 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-37333132

RESUMEN

Intratumoral heterogeneity (ITH) can promote cancer progression and treatment failure, but the complexity of the regulatory programs and contextual factors involved complicates its study. To understand the specific contribution of ITH to immune checkpoint blockade (ICB) response, we generated single cell-derived clonal sublines from an ICB-sensitive and genetically and phenotypically heterogeneous mouse melanoma model, M4. Genomic and single cell transcriptomic analyses uncovered the diversity of the sublines and evidenced their plasticity. Moreover, a wide range of tumor growth kinetics were observed in vivo , in part associated with mutational profiles and dependent on T cell-response. Further inquiry into melanoma differentiation states and tumor microenvironment (TME) subtypes of untreated tumors from the clonal sublines demonstrated correlations between highly inflamed and differentiated phenotypes with the response to anti-CTLA-4 treatment. Our results demonstrate that M4 sublines generate intratumoral heterogeneity at both levels of intrinsic differentiation status and extrinsic TME profiles, thereby impacting tumor evolution during therapeutic treatment. These clonal sublines proved to be a valuable resource to study the complex determinants of response to ICB, and specifically the role of melanoma plasticity in immune evasion mechanisms.

9.
J Biol Chem ; 285(51): 40333-41, 2010 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-20956525

RESUMEN

IFNα exerts potent inhibitory activities against malignant melanoma cells in vitro and in vivo, but the mechanisms by which it generates its antitumor effects remain unknown. We examined the effects of interferon α (IFNα) on the expression of human members of the Schlafen (SLFN) family of genes, a group of cell cycle regulators that mediate growth-inhibitory responses. Using quantitative RT-real time PCR, we found detectable basal expression of all the different human SLFN genes examined (SLFN5, SLFN11, SLFN12, SLFN13, and SLFN14), in malignant melanoma cells and primary normal human melanocytes, but SLFN5 basal expression was suppressed in all analyzed melanoma cell lines. Treatment of melanoma cells with IFNα resulted in induction of expression of SLFN5 in malignant cells, suggesting a potential involvement of this gene in the antitumor effects of IFNα. Importantly, stable knockdown of SLFN5 in malignant melanoma cells resulted in increased anchorage-independent growth, as evidenced by enhanced colony formation in soft agar assays. Moreover, SLFN5 knockdown also resulted in increased invasion in three-dimensional collagen, suggesting a dual role for SLFN5 in the regulation of invasion and anchorage-independent growth of melanoma cells. Altogether, our findings suggest an important role for the SLFN family of proteins in the generation of the anti-melanoma effects of IFNα and for the first time directly implicate a member of the human SLFN family in the regulation of cell invasion.


Asunto(s)
Proteínas de Ciclo Celular/biosíntesis , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Factores Inmunológicos/farmacología , Interferón-alfa/farmacología , Melanocitos/metabolismo , Melanoma/metabolismo , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Melanocitos/patología , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/patología , Invasividad Neoplásica
10.
Proc Natl Acad Sci U S A ; 105(12): 4808-13, 2008 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-18339807

RESUMEN

Multiple signaling pathways are engaged by the type I and II IFN receptors, but their specific roles and possible coordination in the generation of IFN-mediated biological responses remain unknown. We provide evidence that activation of Akt kinases is required for IFN-inducible engagement of the mTOR/p70 S6 kinase pathway. Our data establish that Akt activity is essential for up-regulation of key IFN-alpha- and IFN-gamma-inducible proteins, which have important functional consequences in the induction of IFN responses. Such effects of the Akt pathway are unrelated to regulatory activities on IFN-dependent STAT phosphorylation/activation or transcriptional regulation. By contrast, they reflect regulatory activities on mRNA translation via direct control of the mTOR pathway. In studies using Akt1 and Akt2 double knockout cells, we found that the absence of Akt kinases results in dramatic reduction in IFN-induced antiviral responses, establishing a critical role of the Akt pathway in IFN signaling. Thus, activation of the Akt pathway by the IFN receptors complements the function of IFN-activated JAK-STAT pathways, by allowing mRNA translation of IFN-stimulated genes and, ultimately, the induction of the biological effects of IFNs.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Interferón-alfa/farmacología , Interferón gamma/farmacología , Biosíntesis de Proteínas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Línea Celular , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Citocinas/genética , Citocinas/metabolismo , Virus de la Encefalomiocarditis/efectos de los fármacos , Virus de la Encefalomiocarditis/inmunología , Activación Enzimática/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Fibroblastos/virología , Ratones , Fosforilación/efectos de los fármacos , Proteínas Quinasas/metabolismo , Factores de Transcripción STAT/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR , Transcripción Genética/efectos de los fármacos , Ubiquitinas/genética , Ubiquitinas/metabolismo
11.
Mol Pharmacol ; 77(5): 828-35, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20159944

RESUMEN

Arsenic trioxide (As(2)O(3)) has potent antileukemic properties in vitro and in vivo, but the mechanisms by which it generates its effects on target leukemic cells are not well understood. Understanding cellular mechanisms and pathways that are activated in leukemic cells to control the generation of As(2)O(3) responses should have important implications in the development of novel approaches using As(2)O(3) for the treatment of leukemias. In this study, we used immunoblotting and immune complex kinase assays to provide evidence that the kinases thousand-and-one amino acid kinase 2 (TAO2) and transforming growth factor-beta-activated kinase 1 (TAK1) are rapidly activated in response to treatment of acute leukemia cells with As(2)O(3). Such activation occurs after the generation of reactive oxygen species and regulates downstream engagement of the p38 mitogen-activated protein kinase. Our studies demonstrate that siRNA-mediated knockdown of TAO2 or TAK1 or pharmacological inhibition of TAK1 enhances the suppressive effects of As(2)O(3) on KT-1-derived leukemic progenitor colony formation and on primary leukemic progenitors from patients with acute myelogenous leukemia. These results indicate key negative-feedback regulatory roles for these kinases in the generation of the antileukemic effects of As(2)O(3). Thus, molecular or pharmacological targeting of these kinases may provide a novel approach to enhance the generation of arsenic-dependent antileukemic responses.


Asunto(s)
Arsenicales/farmacología , Quinasas Quinasa Quinasa PAM/metabolismo , Óxidos/farmacología , Acetilcisteína/farmacología , Trióxido de Arsénico , Línea Celular Tumoral , Ditiotreitol/farmacología , Activación Enzimática , Variación Genética , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/fisiología , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva , Quinasas Quinasa Quinasa PAM/efectos de los fármacos , Quinasas Quinasa Quinasa PAM/genética , Fosforilación , Proteínas Quinasas/efectos de los fármacos , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , ARN Interferente Pequeño/genética , Especies Reactivas de Oxígeno/metabolismo , Tretinoina/farmacología , Células U937
12.
Mol Pharmacol ; 78(4): 778-84, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20664001

RESUMEN

Mnk kinases are downstream effectors of mitogen-activated protein kinase pathways and mediate phosphorylation of the eukaryotic initiation factor (eIF4E), a protein that plays a key role in the regulation of mRNA translation and is up-regulated in acute myeloid leukemia (AML). We determined the effects of chemotherapy (cytarabine) on the activation status of Mnk in AML cells and its role in the generation of antileukemic responses. A variety of experimental approaches were used, including immunoblotting, apoptosis assays, small interfering RNA (siRNA)-mediated knockdown of proteins, and clonogenic hematopoietic progenitor assays in methylcellulose. Cytarabine induced phosphorylation/activation of Mnk and Mnk-mediated phosphorylation of eIF4E on Ser209, as evidenced by studies involving pharmacological inhibition of Mnk or experiments using cells with targeted disruption of Mnk1 and Mnk2 genes. To assess the functional relevance of cytarabine-inducible engagement of Mnk/eIF4E pathway, the effects of pharmacological inhibition of Mnk on cytarabine-mediated suppression of primitive leukemic progenitors [leukemic colony forming unit (CFU-L)] were examined. Concomitant treatment of cells with a pharmacological inhibitor of Mnk or siRNA-mediated knockdown of Mnk1/2 strongly enhanced the suppressive effects of low cytarabine concentrations on CFU-L. It is noteworthy that the mammalian target of rapamycin (mTOR) inhibitor rapamycin also induced phosphorylation of eIF4E in a Mnk-dependent manner, whereas inhibition strongly enhanced its antileukemic effects. These data demonstrate that Mnk kinases are activated in a negative-feedback regulatory manner in response to chemotherapy and impair the generation of antileukemic responses. They also identify this pathway as a novel target for the design of new approaches to enhance the antileukemic effects of chemotherapy or mTOR inhibitors in AML.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Citarabina/farmacología , Leucemia/tratamiento farmacológico , Leucemia/enzimología , Proteínas Serina-Treonina Quinasas/fisiología , Animales , Antimetabolitos Antineoplásicos/uso terapéutico , Línea Celular Transformada , Células Cultivadas , Citarabina/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Células K562 , Leucemia/patología , Ratones , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Células U937
13.
J Biol Chem ; 284(37): 25051-64, 2009 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-19592487

RESUMEN

The precise STAT-regulated gene targets that inhibit cell growth and generate the antitumor effects of Type I interferons (IFNs) remain unknown. We provide evidence that Type I IFNs regulate expression of Schlafens (SLFNs), a group of genes involved in the control of cell cycle progression and growth inhibitory responses. Using cells with targeted disruption of different STAT proteins and/or the p38 MAP kinase, we demonstrate that the IFN-dependent expression of distinct Schlafen genes is differentially regulated by STAT complexes and the p38 MAP kinase pathway. We also provide evidence for a key functional role of a member of the SLFN family, SLFN2, in the induction of the growth-suppressive effects of IFNs. This is shown in studies demonstrating that knockdown of SLFN2 enhances hematopoietic progenitor colony formation and reverses the growth-suppressive effects of IFNalpha on normal hematopoietic progenitors. Importantly, NIH3T3 or L929 cells with stable knockdown of SLFN2 form more colonies in soft agar, implicating this protein in the regulation of anchorage-independent growth. Altogether, our data implicate SLFN2 as a negative regulator of the metastatic and growth potential of malignant cells and strongly suggest a role for the SLFN family of proteins in the generation of the antiproliferative effects of Type I IFNs.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Interferón-alfa/metabolismo , Animales , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Proliferación Celular , Islas de CpG , Citocinas/metabolismo , Células Madre Hematopoyéticas/citología , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Transgénicos , Células 3T3 NIH , Factor de Transcripción STAT1/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
Br J Haematol ; 150(2): 158-69, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20507312

RESUMEN

Mantle cell lymphoma (MCL) is characterized by translocation t(11;14)(q13;q32), aggressive clinical behaviour, and poor patient outcomes following conventional chemotherapy. New treatment approaches are needed that target novel biological pathways. All trans retinoic acid (ATRA) is a key retinoid that acts through nuclear receptors that function as ligand-inducible transcription factors. The present study evaluated cell killing effects of ATRA-enriched nanoscale delivery particles, termed nanodisks (ND), on MCL cell lines. Results show that ATRA-ND induced cell death more effectively than naked ATRA (dimethyl sulphoxide) or empty ND. ATRA-ND induced reactive oxygen species (ROS) generation to a greater extent than naked ATRA. The antioxidant, N-acetylcysteine, inhibited ATRA-ND induced apoptosis. Compared to naked ATRA, ATRA-ND enhanced G1 growth arrest, up-regulated p21and p27, and down regulated cyclin D1. At ATRA concentrations that induced apoptosis, expression levels of retinoic acid receptor-alpha (RARalpha) and retinoid X receptor-gamma (RXRgamma) were increased. Compared to naked ATRA, ATRA-ND significantly stimulated transcriptional activity of RARA in a model carcinoma cell line. Furthermore, the RAR antagonist, Ro 41-5253, inhibited ATRA-ND induced ROS generation and prevented ATRA-ND induced cell growth arrest and apoptosis. In summary, incorporation of ATRA into ND enhanced the biological activity of this retinoid in cell culture models of MCL.


Asunto(s)
Antineoplásicos/farmacología , Linfoma de Células del Manto/patología , Receptores de Ácido Retinoico/efectos de los fármacos , Tretinoina/farmacología , Antineoplásicos/administración & dosificación , Apoptosis/efectos de los fármacos , Benzoatos/farmacología , Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/efectos de los fármacos , Cromanos/farmacología , Sistemas de Liberación de Medicamentos , Evaluación Preclínica de Medicamentos , Factores de Intercambio de Guanina Nucleótido/biosíntesis , Factores de Intercambio de Guanina Nucleótido/efectos de los fármacos , Humanos , Linfoma de Células del Manto/metabolismo , Nanopartículas , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/efectos de los fármacos , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Receptores de Ácido Retinoico/antagonistas & inhibidores , Receptores de Ácido Retinoico/metabolismo , Receptores X Retinoide/efectos de los fármacos , Receptores X Retinoide/metabolismo , Transcripción Genética/efectos de los fármacos , Tretinoina/administración & dosificación , Células Tumorales Cultivadas
15.
J Immunol ; 181(10): 7316-23, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18981154

RESUMEN

PI3K is activated by the type I and II IFN receptors, but its precise role in the generation of IFN responses is not well understood. In the present study we used embryonic fibroblasts from mice with targeted disruption of the genes encoding for both the p85alpha and p85beta regulatory subunits of PI3'-kinase (p85alpha(-/-)beta(-/-)) to precisely define the role of PI3K in the control of IFN-induced biological responses. Our data demonstrate that PI3K plays dual regulatory roles in the induction of IFN responses by controlling both IFN-alpha- and IFN-gamma-dependent transcriptional regulation of IFN-sensitive genes and simultaneously regulating the subsequent initiation of mRNA translation for such genes. These processes include the Isg15, Cxcl10, and/or Irf7 genes, whose functions are important in the generation of the biological effects of IFNs. Consistent with this, the induction of IFN antiviral responses is defective in double p85alpha/p85beta knockout cells. Thus, integration of signals via PI3K is a critical event during engagement of the IFN receptors that complements both the transcriptional activity of Jak-STAT pathways and controls initiation of mRNA translation.


Asunto(s)
Fibroblastos/metabolismo , Regulación de la Expresión Génica , Interferones/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/inmunología , Animales , Western Blotting , Infecciones por Cardiovirus/inmunología , Virus de la Encefalomiocarditis/inmunología , Fibroblastos/inmunología , Fibroblastos/virología , Expresión Génica , Interferones/inmunología , Ratones , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/inmunología , Fosforilación , Biosíntesis de Proteínas , Proteínas Proto-Oncogénicas c-akt , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética
16.
Mol Cancer Ther ; 8(3): 615-25, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19240159

RESUMEN

There is emerging evidence that, beyond their cholesterol-lowering properties, statins exhibit important antileukemic effects in vitro and in vivo, but the precise mechanisms by which they generate such responses remain to be determined. We have previously shown that statins promote differentiation of acute promyelocytic leukemia cells and enhance generation of all-trans retinoic acid (ATRA)-dependent antileukemic responses. We now provide evidence that statin-dependent leukemic cell differentiation requires engagement and activation of the c-Jun NH2-terminal kinase kinase pathway. In addition, in experiments, to define the molecular targets and mediators of statin-induced differentiation, we found a remarkable effect of statins on ATRA-dependent gene transcription, evidenced by the selective induction of over 400 genes by the combination of atorvastatin and ATRA. Altogether, our studies identify novel statin molecular targets linked to differentiation, establish that statins modulate ATRA-dependent transcription, and suggest that combined use of statins with retinoids may provide a novel approach to enhance antileukemic responses in acute promyelocytic leukemia and possibly other leukemias.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Leucemia/patología , Retinoides/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Atorvastatina , Diferenciación Celular/genética , Sinergismo Farmacológico , Perfilación de la Expresión Génica , Redes Reguladoras de Genes/efectos de los fármacos , Ácidos Heptanoicos/administración & dosificación , Ácidos Heptanoicos/farmacología , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Leucemia/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Pirroles/administración & dosificación , Pirroles/farmacología , Retinoides/administración & dosificación , Factores de Tiempo , Tretinoina/administración & dosificación , Tretinoina/farmacología , Células Tumorales Cultivadas
17.
Nat Commun ; 11(1): 333, 2020 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-31949145

RESUMEN

Cutaneous malignant melanoma is an aggressive cancer of melanocytes with a strong propensity to metastasize. We posit that melanoma cells acquire metastatic capability by adopting an embryonic-like phenotype, and that a lineage approach would uncover metastatic melanoma biology. Using a genetically engineered mouse model to generate a rich melanoblast transcriptome dataset, we identify melanoblast-specific genes whose expression contribute to metastatic competence and derive a 43-gene signature that predicts patient survival. We identify a melanoblast gene, KDELR3, whose loss impairs experimental metastasis. In contrast, KDELR1 deficiency enhances metastasis, providing the first example of different disease etiologies within the KDELR-family of retrograde transporters. We show that KDELR3 regulates the metastasis suppressor, KAI1, and report an interaction with the E3 ubiquitin-protein ligase gp78, a regulator of KAI1 degradation. Our work demonstrates that the melanoblast transcriptome can be mined to uncover targetable pathways for melanoma therapy.


Asunto(s)
Perfilación de la Expresión Génica , Melanoma/genética , Melanoma/metabolismo , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Transcriptoma , Animales , Línea Celular Tumoral , Retículo Endoplásmico , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Proteína Kangai-1/genética , Proteína Kangai-1/metabolismo , Pulmón/patología , Melanocitos/metabolismo , Melanoma/patología , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia/genética , Neoplasias Primarias Secundarias/patología , Fenotipo , Receptores de Péptidos/genética , Receptores de Péptidos/metabolismo , Neoplasias Cutáneas/patología , Ubiquitina-Proteína Ligasas/metabolismo , Melanoma Cutáneo Maligno
18.
Clin Cancer Res ; 14(14): 4640-9, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18628479

RESUMEN

PURPOSE: Statins are pharmacologic inhibitors of the 3-hydroxy-3-methylglutaryl-coenzyme A reductase with potent regulatory effects on cholesterol biosynthesis in vitro and in vivo. There is accumulating evidence that, beyond their cholesterol-lowering properties, statins inhibit cell proliferation and promote apoptosis of malignant cells in vitro, but the mechanisms by which they generate such responses remain to be defined. EXPERIMENTAL DESIGN: Combinations of experimental approaches were used, including immunoblotting and cell proliferation and apoptosis assays. RESULTS: We provide evidence that fluvastatin is a potent inducer of apoptosis and suppresses proliferation of renal cell carcinoma (RCC) cells in vitro. Such effects are mediated by direct targeting of the Akt/mammalian target of rapamycin (mTOR) pathway, as evidenced by the suppression of phosphorylation/activation of Akt, resulting in inhibition of its downstream effectors, mTOR and p70 S6 kinase. In addition, fluvastatin blocks the mTOR-dependent phosphorylation/deactivation of the translational repressor eukaryotic initiation factor 4E (eIF4E)-binding protein, leading to the formation of eIF4E-binding protein-eIF4E complexes that suppress initiation of cap-dependent mRNA translation. Importantly, inhibition of p70 S6 kinase activity by fluvastatin results in the up-regulation of expression of programmed cell death 4 (PDCD4), a tumor suppressor protein with inhibitory effects on the translation initiation factor eIF4A, suggesting a mechanism for the generation of antitumor responses. CONCLUSIONS: Altogether, our findings establish that fluvastatin exhibits potent anti-RCC activities via inhibitory effects on the Akt/mTOR pathway and raise the possibility that combinations of statins and Akt inhibitors may be of future therapeutic value in the treatment of RCC.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/efectos de los fármacos , Carcinoma de Células Renales/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Neoplasias Renales/metabolismo , Proteínas Quinasas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas de Unión al ARN/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Línea Celular Tumoral , Electroforesis en Gel de Poliacrilamida , Citometría de Flujo , Humanos , Immunoblotting , Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas de Unión al ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR , Regulación hacia Arriba
19.
Cancer Res ; 67(9): 4524-32, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17483369

RESUMEN

The family of statins includes pharmacologic inhibitors of the 3-hydroxy-3-methylglutaryl-CoA reductase that are potent regulators of cholesterol biosynthesis. In addition to their cholesterol-lowering effects, statins inhibit cell proliferation and promote apoptosis of malignant cells in vitro, but their potential therapeutic roles in the treatment of malignancies remain to be defined. We examined the effects of statins on the growth and differentiation of acute myeloid leukemia (AML) cells. Atorvastatin and fluvastatin were found to be potent inducers of cell differentiation and apoptosis of the NB4 acute promyelocytic leukemia (APL) cell line. Such effects correlated with activation of the small G-proteins Rac1/Cdc42 and downstream engagement of the c-Jun NH(2)-terminal kinase kinase pathway, whose function was found to be essential for the generation of proapoptotic responses. Importantly, different statins were found to enhance all-trans-retinoic acid (ATRA)-dependent differentiation of APL blasts and reverse resistance to the antileukemic effects of ATRA. In addition, fluvastatin exhibited growth-inhibitory properties on primary bone marrow-derived leukemic progenitors from patients with AML and enhanced the suppressive effects of ATRA on leukemic progenitor colony formation. Altogether, these studies establish that statins exhibit potent antileukemic properties in vitro and raise the possibility that combinations of statins with ATRA may be an effective approach to overcome the development of ATRA resistance by the leukemic cells.


Asunto(s)
Ácidos Grasos Monoinsaturados/farmacología , Ácidos Heptanoicos/farmacología , Indoles/farmacología , Leucemia Promielocítica Aguda/tratamiento farmacológico , Pirroles/farmacología , Atorvastatina , Diferenciación Celular/efectos de los fármacos , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Sinergismo Farmacológico , Fluvastatina , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Leucemia Promielocítica Aguda/patología , Tretinoina/farmacología , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/metabolismo
20.
Biochem Biophys Res Commun ; 368(4): 983-9, 2008 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-18280804

RESUMEN

The mechanisms by which retinoids regulate initiation of mRNA translation for proteins that mediate their biological effects are not known. We have previously shown that all-trans-retinoic acid (ATRA) induces mTOR-mediated activation of the p70 S6 kinase, suggesting the existence of a mechanism by which retinoids may regulate mRNA translation. We now demonstrate that treatment of acute promyelocytic leukemia (APL)-derived NB4 cells with ATRA results in dissociation of the translational repressor 4E-BP1 from the eukaryotic initiation factor eIF4E, and subsequent formation of eIF4G-eIF4E complexes. We also show that siRNA-mediated inhibition of 4E-BP1 expression enhances ATRA-dependent upregulation of p21(Waf1/Cip1), a protein that plays a key role in the induction of retinoid-dependent responses. Our data also establish that ATRA- or cis-RA-dependent p21(Waf1/Cip1) protein expression is enhanced in mouse embryonic fibroblasts with targeted disruption of the 4e-bp1 gene, in the absence of any effects on the transcriptional regulation of the p21(Waf1/Cip1) gene. Moreover, generation of ATRA- or cis-retinoic acid (cis-RA)-antiproliferative responses is enhanced in 4E-BP1 knockout cells. Altogether, these findings strongly suggest a key regulatory role for the translational repressor 4E-BP1 in the generation of retinoid-dependent functional responses.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Fosfoproteínas/fisiología , Tretinoina/farmacología , Animales , Proteínas de Ciclo Celular , Línea Celular Tumoral , Factor 4E Eucariótico de Iniciación/metabolismo , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Humanos , Leucemia Promielocítica Aguda , Ratones , Ratones Noqueados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA