Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
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.

2.
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.

3.
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
4.
Mol Cell Biol ; 38(16)2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-29866656

RESUMEN

Although members of the Slfn family have been implicated in the regulation of type I interferon (IFN) responses, the mechanisms by which they mediate their effects remain unknown. In the present study, we provide evidence that targeted disruption of the Slfn2 gene leads to increased transcription of IFN-stimulated genes (ISGs) and enhanced type I IFN-mediated antiviral responses. We demonstrate that Slfn2 interacts with protein phosphatase 6 regulatory subunit 1 (PPP6R1), leading to reduced type I IFN-induced activation of nuclear factor kappa B (NF-κB) signaling, resulting in reduced expression of ISGs. Altogether, these data suggest a novel mechanism by which Slfn2 controls ISG expression and provide evidence for a critical role for Slfn2 in the regulation of IFN-mediated biological responses.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Interferón Tipo I/metabolismo , FN-kappa B/metabolismo , Animales , Sitios de Unión/genética , Proteínas de Ciclo Celular/deficiencia , Proteínas de Ciclo Celular/genética , Células Cultivadas , Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Células 3T3 NIH , Fosfoproteínas Fosfatasas/metabolismo , Regiones Promotoras Genéticas , Transducción de Señal
5.
Mol Cell Biol ; 35(15): 2684-98, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26012550

RESUMEN

We provide evidence that human SLFN5, an interferon (IFN)-inducible member of the Schlafen (SLFN) family of proteins, exhibits key roles in controlling motility and invasiveness of renal cell carcinoma (RCC) cells. Our studies define the mechanism by which this occurs, demonstrating that SLFN5 negatively controls expression of the matrix metalloproteinase 1 gene (MMP-1), MMP-13, and several other genes involved in the control of malignant cell motility. Importantly, our data establish that SLFN5 expression correlates with a better overall survival in a large cohort of patients with RCC. The inverse relationship between SLFN5 expression and RCC aggressiveness raises the possibility of developing unique therapeutic approaches in the treatment of RCC, by modulating SLFN5 expression.


Asunto(s)
Carcinoma de Células Renales/patología , Proteínas de Ciclo Celular/genética , Neoplasias Renales/patología , Metaloproteinasa 13 de la Matriz/biosíntesis , Metaloproteinasa 1 de la Matriz/biosíntesis , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/mortalidad , Proteínas de Ciclo Celular/biosíntesis , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Interferón-alfa/farmacología , Neoplasias Renales/genética , Neoplasias Renales/mortalidad , Invasividad Neoplásica/genética , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Interferente Pequeño
6.
Discov Med ; 20(112): 413-27, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26760985

RESUMEN

Statins, the most commonly prescribed class of drug, have demonstrated effects beyond cholesterol reduction including anti-tumor and immunomodulatory properties. Several epidemiological studies have suggested an anti-neoplastic effect of statins evidenced by reductions in cancer incidence and cancer-related mortality. Clinical trials on statins as part of therapy for cancer have generated interest in the oncology community. Statins have been investigated for a variety of cancers, early and late stage, and in combination with chemotherapy and radiation. So far promising results have been reported with statin use in pediatric brainstem tumors, early stage breast cancer, hepatocellular carcinoma (HCC), colorectal cancer (CRC), refractory or relapsed multiple myeloma (MM), and refractory acute myeloid leukemia (AML). There is still much investigation to be completed to determine which subtypes of patients benefit from statin therapy, how statins may potentiate other anticancer approaches, and the appropriate dosing schedule to use.


Asunto(s)
Antineoplásicos/uso terapéutico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/epidemiología , Investigación Biomédica Traslacional , Ensayos Clínicos como Asunto , Terapia Combinada , Humanos , Estadificación de Neoplasias , Neoplasias/patología , Neoplasias/radioterapia
7.
Clin Cancer Res ; 20(9): 2400-9, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24610825

RESUMEN

PURPOSE: To examine whether induction of autophagy is a mechanism of leukemic cell resistance to dual mTORC1/mTORC2 inhibitors in acute myelogenous leukemia (AML) leukemic progenitors. EXPERIMENTAL DESIGN: Combinations of different experimental approaches were used to assess induction of autophagy, including immunoblotting to detect effects on LC3II and p62/SQTM1 expression and on ULK1 phosphorylation, immunofluorescence, and electron microscopy. Functional responses were assessed using cell viability and apoptosis assays, and clonogenic leukemic progenitor assays in methylcellulose. RESULTS: We provide evidence that treatment of AML cells with catalytic mTOR inhibitors results in induction of autophagy, which acts as a regulatory mechanism to promote leukemic cell survival. Such induction of autophagy by dual mTORC1/mTORC2 inhibitors partially protects primitive leukemic precursors from the inhibitory effects of such agents and limits their activities. Simultaneous blockade of the autophagic process using chloroquine or by knockdown of ULK1 results in enhanced antileukemic responses. CONCLUSIONS: Dual targeting of mTORC2 and mTORC1 results in induction of autophagy in AML cells. Combinations of catalytic mTOR targeting agents and autophagy inhibitors may provide a unique approach to target primitive leukemic precursors in AML.


Asunto(s)
Autofagia/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Leucemia Mieloide Aguda/metabolismo , Complejos Multiproteicos/antagonistas & inhibidores , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Línea Celular Tumoral , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Células Madre Neoplásicas/ultraestructura
8.
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
9.
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
10.
Leuk Lymphoma ; 54(12): 2601-5, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23540342

RESUMEN

Statins are 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors that act on the mevalonate pathway and inhibit synthesis of cholesterol, geranylgeranyl pyrophosphate (GGPP) and farnesyl pyrophosphate (FPP). In preclinical studies, these agents have been shown to inhibit proliferation, trigger apoptosis and promote cell differentiation of leukemia. Proposed mechanisms include cholesterol deprivation and inhibition of isoprenylation of important signaling molecules. Case reports and early clinical studies suggest a therapeutic potential for statins in acute myeloid leukemia (AML). In the other leukemias there are limited clinical data, but in vitro studies provide a strong rationale for future studies involving statins. The effects of statins on the immune system may lend these agents to a role in allogeneic stem cell transplant. While many of the studies are early, statins have the future potential to be integrated into conventional chemotherapy regimens with limited side effects.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Leucemia/tratamiento farmacológico , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Humanos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico
11.
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
12.
Cancer Biol Ther ; 14(5): 411-6, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23377826

RESUMEN

Arsenic Trioxide (As2O3) is one of the most effective agents in the treatment of acute promyelocytic leukemia (APL), but has no significant efficacy in other forms of AML. The mechanisms of relative resistance of non-APL cells are not well understood, but emerging evidence suggests that activation of negative feedback regulatory loops and pathways contributes to such resistance. We provide evidence that a signaling cascade involving the kinase RSK1 is engaged in a negative feedback manner during arsenic-treatment of cells and exhibits regulatory effects on growth and survival of AML cells in response to treatment with As2O3. Our data demonstrate that pharmacological inhibition or molecular disruption of expression of RSK1 enhances As2O3-dependent apoptosis and/or growth inhibition of AML cells. Importantly, combination of a pharmacological inhibitor of RSK and As2O3 results in enhanced suppression of primary AML leukemic progenitors. Altogether, our findings suggest an important regulatory role for RSK1 in the generation of the effects of As2O3 in AML cells. They also raise the potential of RSK1 targeting in combination with As2O3 as a novel approach to promote antileukemic responses.


Asunto(s)
Arsenicales/farmacología , Leucemia Mieloide Aguda/tratamiento farmacológico , Óxidos/farmacología , Proteínas Quinasas S6 Ribosómicas 90-kDa/antagonistas & inhibidores , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Apoptosis/efectos de los fármacos , Trióxido de Arsénico , Línea Celular Tumoral , Electroforesis en Gel de Poliacrilamida , Humanos , Leucemia Mieloide Aguda/sangre , Leucemia Mieloide Aguda/enzimología , Leucemia Mieloide Aguda/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Células U937
13.
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
14.
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
15.
Leuk Lymphoma ; 53(9): 1779-84, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22356114

RESUMEN

Statins are HMG-CoA (3-hydroxy-3-methyl-glutaryl-coenzyme A) reductase inhibitors, which block the conversion of HMG-CoA to mevalonate and have potent cholesterol lowering properties. Beyond their importance in the generation of lipid lowering effects, the regulatory effects of statins on the mevalonate pathway have a significant impact on multiple other cellular functions. There is now extensive evidence that statins have anti-inflammatory and anti-neoplastic properties, but the precise mechanisms by which such responses are generated are not well understood. In the present study we demonstrate that statins engage a member of the protein kinase C (PKC) family of proteins, PKCδ, in acute promyelocytic leukemia (APL) cells. Our study shows that atorvastatin and fluvastatin induce proteolytic activation of PKCδ in the APL NB4 cell line, which expresses the t(15;17) translocation. Such engagement of PKCδ results in induction of its kinase domain and downstream regulation of pathways important for statin-dependent leukemia cell differentiation. Our research shows that the function of PKCδ is essential for statin-induced leukemic cell differentiation, as demonstrated by studies involving selective targeting of PKCδ using siRNAs. We also demonstrate that the potent enhancing effects of statins on all-trans retinoic acid (ATRA)-induced gene expression for CCL3 and CCL4 requires the function of PKCδ, suggesting a mechanism by which statins may promote ATRA-induced antileukemic responses. Altogether, our data establish a novel function for PKCδ as a mediator of statin-induced differentiation of APL cells and antileukemic effects.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Ácidos Grasos Monoinsaturados/farmacología , Ácidos Heptanoicos/farmacología , Indoles/farmacología , Proteína Quinasa C-delta/metabolismo , Pirroles/farmacología , Antineoplásicos/farmacología , Atorvastatina , Diferenciación Celular/genética , Línea Celular Tumoral , Quimiocina CCL3/genética , Quimiocina CCL3/metabolismo , Quimiocina CCL4/genética , Quimiocina CCL4/metabolismo , Sinergismo Farmacológico , Activación Enzimática/efectos de los fármacos , Fluvastatina , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Immunoblotting , Leucemia Promielocítica Aguda/enzimología , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/patología , Proteína Quinasa C-delta/genética , Proteolisis/efectos de los fármacos , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Tretinoina/farmacología
16.
Oncotarget ; 2(6): 510-7, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21680954

RESUMEN

Despite recent advances in the field, the treatment of patients with acute myeloid leukemia (AML) remains challenging and difficult. Although chemotherapeutic agents induce remissions in a large number of patients, many of them eventually relapse and die. A major goal for the development of new approaches for the treatment of AML is to enhance the antileukemic effects of standard chemotherapeutics and to design effective combinations targeting non-overlapping cellular pathways. The PI3K/Akt/mTOR signaling pathway plays a critical role in survival and growth of malignant cells and its targeting has been the focus of extensive work and research efforts over the last two decades. It now appears possible that a major limitation of the first generation of mTOR inhibitors can be overcome by a new class of catalytic inhibitors of mTOR. There is emerging evidence that such compounds target both TORC1 and TORC2 and elicit much more potent responses against early leukemic precursors in vitro. In addition, recent studies have shown that combinations of such agents with cytarabine result in enhanced antileukemic responses in vitro, raising the prospect and potential of use of these agents in combination regimens for the treatment of AML.


Asunto(s)
Antineoplásicos/uso terapéutico , Drogas en Investigación/uso terapéutico , Leucemia Mieloide Aguda/tratamiento farmacológico , Terapia Molecular Dirigida , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Drogas en Investigación/farmacología , Humanos , Oncología Médica/métodos , Oncología Médica/tendencias , Modelos Biológicos , Terapia Molecular Dirigida/métodos
17.
Clin Cancer Res ; 17(13): 4378-88, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21415215

RESUMEN

PURPOSE: To determine whether mTORC2 and rapamycin-insensitive (RI)-mTORC1 complexes are present in acute myeloid leukemia (AML) cells and to examine the effects of dual mTORC2/mTORC1 inhibition on primitive AML leukemic progenitors. EXPERIMENTAL DESIGN: Combinations of different experimental approaches were used, including immunoblotting to detect phosphorylated/activated forms of elements of the mTOR pathway in leukemic cell lines and primary AML blasts; cell-proliferation assays; direct assessment of mRNA translation in polysomal fractions of leukemic cells; and clonogenic assays in methylcellulose to evaluate leukemic progenitor-colony formation. RESULTS: mTORC2 complexes are active in AML cells and play critical roles in leukemogenesis. RI-mTORC1 complexes are also formed and regulate the activity of the translational repressor 4E-BP1 in AML cells. OSI-027 blocks mTORC1 and mTORC2 activities and suppresses mRNA translation of cyclin D1 and other genes that mediate proliferative responses in AML cells. Moreover, OSI-027 acts as a potent suppressor of primitive leukemic precursors from AML patients and is much more effective than rapamycin in eliciting antileukemic effects in vitro. CONCLUSIONS: Dual targeting of mTORC2 and mTORC1 results in potent suppressive effects on primitive leukemic progenitors from AML patients. Inhibition of the mTOR catalytic site with OSI-027 results in suppression of both mTORC2 and RI-mTORC1 complexes and elicits much more potent antileukemic responses than selective mTORC1 targeting with rapamycin.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Leucemia Mieloide Aguda/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Proteínas/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Proteínas Reguladoras de la Apoptosis/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina D1/genética , Ciclina D1/metabolismo , Relación Dosis-Respuesta a Droga , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Células HL-60 , Humanos , Leucemia Mieloide Aguda/genética , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos , Células Madre Neoplásicas/metabolismo , Proteína Oncogénica v-akt/metabolismo , Fosforilación/efectos de los fármacos , Proteínas de Unión al ARN/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Sirolimus/farmacología , Serina-Treonina Quinasas TOR , Células U937
18.
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
19.
Autophagy ; 6(7): 966-7, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20699667

RESUMEN

In recent years, there have been substantial research advances on the mechanisms by which BCR-ABL transforms hematopoietic cells and promotes leukemic cell growth and survival. Among the diverse signaling cascades activated by BCR-ABL, the mTOR pathway plays a critical role in mRNA translation of genes that promote leukemogenesis and mitogenic responses. We have recently shown that dual targeting of mTORC1 and mTORC2 complexes using a catalytic mTOR inhibitor, OSI-027, results in generation of potent antileukemic effects against BCR-ABL transformed cells. Such effects were also seen in cells expressing the T315I mutation, which is resistant to all currently approved BCR-ABL kinase inhibitors. Our studies also demonstrate that such dual catalytic inhibition of mTORC2 and mTORC1 complexes in BCR-ABL-expressing K562 cells results in induction of autophagy, and that inhibition of the autophagic process using chloroquine promotes apoptosis of these cells. Altogether, our studies suggest that autophagy may be a limiting factor for the induction of apoptosis during dual mTORC2-mTORC1 targeting, in at least some types of BCR-ABL-expressing cells and have raised the potential of combinations of catalytic inhibitors of mTOR with autophagy inhibitors for the treatment of refractory Ph(+) leukemias.


Asunto(s)
Autofagia/fisiología , Proteínas de Fusión bcr-abl/metabolismo , Proteínas/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Antirreumáticos/farmacología , Apoptosis/fisiología , Autofagia/efectos de los fármacos , Cloroquina/farmacología , Proteínas de Fusión bcr-abl/genética , Humanos , Células K562 , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos , Serina-Treonina Quinasas TOR
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...