Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 175
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 55(4): 671-685.e10, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35417675

RESUMEN

Interferon-gamma (IFN-γ) has pleiotropic effects on cancer immune checkpoint blockade (ICB), including roles in ICB resistance. We analyzed gene expression in ICB-sensitive versus ICB-resistant tumor cells and identified a strong association between interferon-mediated resistance and expression of Ripk1, a regulator of tumor necrosis factor (TNF) superfamily receptors. Genetic interaction screening revealed that in cancer cells, RIPK1 diverted TNF signaling through NF-κB and away from its role in cell death. This promoted an immunosuppressive chemokine program by cancer cells, enhanced cancer cell survival, and decreased infiltration of T and NK cells expressing TNF superfamily ligands. Deletion of RIPK1 in cancer cells compromised chemokine secretion, decreased ARG1+ suppressive myeloid cells linked to ICB failure in mice and humans, and improved ICB response driven by CASP8-killing and dependent on T and NK cells. RIPK1-mediated resistance required its ubiquitin scaffolding but not kinase function. Thus, cancer cells co-opt RIPK1 to promote cell-intrinsic and cell-extrinsic resistance to immunotherapy.


Asunto(s)
Resistencia a Antineoplásicos , Inhibidores de Puntos de Control Inmunológico , Interferones , Neoplasias , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Animales , Inmunoterapia , Interferón gamma/metabolismo , Interferones/metabolismo , Ratones , FN-kappa B/metabolismo , Neoplasias/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo
2.
Nat Rev Genet ; 23(8): 461-466, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35534711

RESUMEN

Careers in biomedicine can take many forms, and one common career decision facing scientists is whether to pursue jobs in academia or industry. In this Viewpoint article, four leading scientists who have spent time in both academia and industry provide their perspectives on both types of workplace, such as whether the environments are really as distinct as they are often perceived to be, as well as how academia-industry collaborations can be a driving force in biomedical research and translation.


Asunto(s)
Investigación Biomédica , Industrias
3.
Cell ; 150(3): 575-89, 2012 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-22863010

RESUMEN

The mechanism by which cells decide to skip mitosis to become polyploid is largely undefined. Here we used a high-content image-based screen to identify small-molecule probes that induce polyploidization of megakaryocytic leukemia cells and serve as perturbagens to help understand this process. Our study implicates five networks of kinases that regulate the switch to polyploidy. Moreover, we find that dimethylfasudil (diMF, H-1152P) selectively increased polyploidization, mature cell-surface marker expression, and apoptosis of malignant megakaryocytes. An integrated target identification approach employing proteomic and shRNA screening revealed that a major target of diMF is Aurora kinase A (AURKA). We further find that MLN8237 (Alisertib), a selective inhibitor of AURKA, induced polyploidization and expression of mature megakaryocyte markers in acute megakaryocytic leukemia (AMKL) blasts and displayed potent anti-AMKL activity in vivo. Our findings provide a rationale to support clinical trials of MLN8237 and other inducers of polyploidization and differentiation in AMKL.


Asunto(s)
Azepinas/farmacología , Descubrimiento de Drogas , Leucemia Megacarioblástica Aguda/tratamiento farmacológico , Megacariocitos/metabolismo , Poliploidía , Pirimidinas/farmacología , Bibliotecas de Moléculas Pequeñas , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Animales , Aurora Quinasa A , Aurora Quinasas , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Leucemia Megacarioblástica Aguda/genética , Megacariocitos/citología , Megacariocitos/patología , Ratones , Ratones Endogámicos C57BL , Mapas de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Quinasas Asociadas a rho/metabolismo
4.
Cell ; 146(5): 697-708, 2011 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-21884932

RESUMEN

AKT activation is associated with many malignancies, where AKT acts, in part, by inhibiting FOXO tumor suppressors. We show a converse role for AKT/FOXOs in acute myeloid leukemia (AML). Rather than decreased FOXO activity, we observed that FOXOs are active in ∼40% of AML patient samples regardless of genetic subtype. We also observe this activity in human MLL-AF9 leukemia allele-induced AML in mice, where either activation of Akt or compound deletion of FoxO1/3/4 reduced leukemic cell growth, with the latter markedly diminishing leukemia-initiating cell (LIC) function in vivo and improving animal survival. FOXO inhibition resulted in myeloid maturation and subsequent AML cell death. FOXO activation inversely correlated with JNK/c-JUN signaling, and leukemic cells resistant to FOXO inhibition responded to JNK inhibition. These data reveal a molecular role for AKT/FOXO and JNK/c-JUN in maintaining a differentiation blockade that can be targeted to inhibit leukemias with a range of genetic lesions.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Animales , Antígenos CD34/metabolismo , Apoptosis , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Células Cultivadas , Modelos Animales de Enfermedad , Proteína Forkhead Box O3 , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/metabolismo
5.
Cell ; 137(5): 821-34, 2009 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-19490892

RESUMEN

An alternative to therapeutic targeting of oncogenes is to perform "synthetic lethality" screens for genes that are essential only in the context of specific cancer-causing mutations. We used high-throughput RNA interference (RNAi) to identify synthetic lethal interactions in cancer cells harboring mutant KRAS, the most commonly mutated human oncogene. We find that cells that are dependent on mutant KRAS exhibit sensitivity to suppression of the serine/threonine kinase STK33 irrespective of tissue origin, whereas STK33 is not required by KRAS-independent cells. STK33 promotes cancer cell viability in a kinase activity-dependent manner by regulating the suppression of mitochondrial apoptosis mediated through S6K1-induced inactivation of the death agonist BAD selectively in mutant KRAS-dependent cells. These observations identify STK33 as a target for treatment of mutant KRAS-driven cancers and demonstrate the potential of RNAi screens for discovering functional dependencies created by oncogenic mutations that may enable therapeutic intervention for cancers with "undruggable" genetic alterations.


Asunto(s)
Neoplasias/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular , Humanos , Ratones , Mutación , Células 3T3 NIH , Neoplasias/genética , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas p21(ras) , Interferencia de ARN , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo
6.
Proc Natl Acad Sci U S A ; 114(18): 4757-4762, 2017 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-28424250

RESUMEN

Fibrotic diseases are not well-understood. They represent a number of different diseases that are characterized by the development of severe organ fibrosis without any obvious cause, such as the devastating diseases idiopathic pulmonary fibrosis (IPF) and scleroderma. These diseases have a poor prognosis comparable with endstage cancer and are uncurable. Given the phenotypic differences, it was assumed that the different fibrotic diseases also have different pathomechanisms. Here, we demonstrate that many endstage fibrotic diseases, including IPF; scleroderma; myelofibrosis; kidney-, pancreas-, and heart-fibrosis; and nonalcoholic steatohepatosis converge in the activation of the AP1 transcription factor c-JUN in the pathologic fibroblasts. Expression of the related AP1 transcription factor FRA2 was restricted to pulmonary artery hypertension. Induction of c-Jun in mice was sufficient to induce severe fibrosis in multiple organs and steatohepatosis, which was dependent on sustained c-Jun expression. Single cell mass cytometry revealed that c-Jun activates multiple signaling pathways in mice, including pAkt and CD47, which were also induced in human disease. αCD47 antibody treatment and VEGF or PI3K inhibition reversed various organ c-Jun-mediated fibroses in vivo. These data suggest that c-JUN is a central molecular mediator of most fibrotic conditions.


Asunto(s)
Fibrosis Pulmonar Idiopática , Mielofibrosis Primaria , Proteínas Proto-Oncogénicas c-jun , Esclerodermia Sistémica , Factor de Transcripción AP-1 , Antígeno 2 Relacionado con Fos/genética , Antígeno 2 Relacionado con Fos/metabolismo , Humanos , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/metabolismo , Mielofibrosis Primaria/genética , Mielofibrosis Primaria/metabolismo , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Esclerodermia Sistémica/genética , Esclerodermia Sistémica/metabolismo , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo
7.
Blood ; 123(22): e123-33, 2014 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-24740812

RESUMEN

Genomic studies have identified somatic alterations in the majority of myeloproliferative neoplasms (MPN) patients, including JAK2 mutations in the majority of MPN patients and CALR mutations in JAK2-negative MPN patients. However, the role of JAK-STAT pathway activation in different MPNs, and in patients without JAK2 mutations, has not been definitively delineated. We used expression profiling, single nucleotide polymorphism arrays, and mutational profiling to investigate a well-characterized cohort of MPN patients. MPN patients with homozygous JAK2V617F mutations were characterized by a distinctive transcriptional profile. Notably, a transcriptional signature consistent with activated JAK2 signaling is seen in all MPN patients regardless of clinical phenotype or mutational status. In addition, the activated JAK2 signature was present in patients with somatic CALR mutations. Conversely, we identified a gene expression signature of CALR mutations; this signature was significantly enriched in JAK2-mutant MPN patients consistent with a shared mechanism of transformation by JAK2 and CALR mutations. We also identified a transcriptional signature of TET2 mutations in MPN patent samples. Our data indicate that MPN patients, regardless of diagnosis or JAK2 mutational status, are characterized by a distinct gene expression signature with upregulation of JAK-STAT target genes, demonstrating the central importance of the JAK-STAT pathway in MPN pathogenesis.


Asunto(s)
Genómica , Quinasas Janus/metabolismo , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/metabolismo , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Calreticulina , Estudios de Casos y Controles , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Análisis por Conglomerados , Femenino , Perfilación de la Expresión Génica , Homocigoto , Humanos , Janus Quinasa 2/genética , Quinasas Janus/genética , Masculino , Mutación , Factores de Transcripción STAT/genética , Transcriptoma
8.
Mol Cell ; 31(1): 134-42, 2008 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-18614052

RESUMEN

Genetic alterations causing constitutive tyrosine kinase activation are observed in a broad spectrum of cancers. Thus far, these mutant kinases have been localized to the plasma membrane or cytoplasm, where they engage proliferation and survival pathways. We report that the NUP214-ABL1 fusion is unique among these because of its requisite localization to the nuclear pore complex for its transforming potential. We show that NUP214-ABL1 displays attenuated transforming capacity as compared to BCR-ABL1 and that NUP214-ABL1 preferentially transforms T cells, which is in agreement with its unique occurrence in T cell acute lymphoblastic leukemia. Furthermore, NUP214-ABL1 differs from BCR-ABL1 in subcellular localization, initiation of kinase activity, and signaling and lacks phosphorylation on its activation loop. In addition to delineating an unusual mechanism for kinase activation, this study provides new insights into the spectrum of chromosomal translocations involving nucleoporins by indicating that the nuclear pore context itself may play a central role in transformation.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Poro Nuclear/enzimología , Proteínas de Fusión Oncogénica/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Animales , Línea Celular , Activación Enzimática , Humanos , Ratones , Proteínas de Complejo Poro Nuclear/metabolismo
9.
Nat Rev Cancer ; 7(9): 673-83, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17721432

RESUMEN

The myeloproliferative disorders polycythaemia vera (PV), essential thombocythaemia (ET), and primary myelofibrosis (PMF) are clonal disorders of multipotent haematopoietic progenitors. The genetic cause of these diseases was not known until 2005, when several independent groups demonstrated that most patients with PV, ET and PMF acquire a single point mutation in the cytoplasmic tyrosine kinase JAK2 (JAK2V617F). These discoveries have changed the landscape for diagnosis and classification of PV, ET and PMF, and show the ability of genomic technologies to identify new molecular targets in human malignancies with pathogenetic, diagnostic and therapeutic significance.


Asunto(s)
Janus Quinasa 2/genética , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/terapia , Activación Enzimática , Predicción , Humanos , Janus Quinasa 2/antagonistas & inhibidores , Janus Quinasa 2/fisiología , Mutación Puntual , Transducción de Señal , Activación Transcripcional
10.
Nat Chem Biol ; 9(12): 840-848, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24161946

RESUMEN

Efforts to develop more effective therapies for acute leukemia may benefit from high-throughput screening systems that reflect the complex physiology of the disease, including leukemia stem cells (LSCs) and supportive interactions with the bone marrow microenvironment. The therapeutic targeting of LSCs is challenging because LSCs are highly similar to normal hematopoietic stem and progenitor cells (HSPCs) and are protected by stromal cells in vivo. We screened 14,718 compounds in a leukemia-stroma co-culture system for inhibition of cobblestone formation, a cellular behavior associated with stem-cell function. Among those compounds that inhibited malignant cells but spared HSPCs was the cholesterol-lowering drug lovastatin. Lovastatin showed anti-LSC activity in vitro and in an in vivo bone marrow transplantation model. Mechanistic studies demonstrated that the effect was on target, via inhibition of HMG-CoA reductase. These results illustrate the power of merging physiologically relevant models with high-throughput screening.


Asunto(s)
Antineoplásicos/farmacología , Ensayos de Selección de Medicamentos Antitumorales/métodos , Leucemia , Células Madre Neoplásicas/efectos de los fármacos , Línea Celular Tumoral , Células Madre Hematopoyéticas , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Lovastatina/farmacología , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/fisiología
11.
Cancer Cell ; 12(4): 367-80, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17936561

RESUMEN

Despite their known transforming properties, the effects of leukemogenic FLT3-ITD mutations on hematopoietic stem and multipotent progenitor cells and on hematopoietic differentiation are not well understood. We report a mouse model harboring an ITD in the murine Flt3 locus that develops myeloproliferative disease resembling CMML and further identified FLT3-ITD mutations in a subset of human CMML. These findings correlated with an increase in number, cell cycling, and survival of multipotent stem and progenitor cells in an ITD dose-dependent manner in animals that exhibited alterations within their myeloid progenitor compartments and a block in normal B cell development. This model provides insights into the consequences of constitutive signaling by an oncogenic tyrosine kinase on hematopoietic progenitor quiescence, function, and cell fate.


Asunto(s)
Proliferación Celular , Células Madre Hematopoyéticas/metabolismo , Leucemia Mielomonocítica Crónica/metabolismo , Células Madre Multipotentes/metabolismo , Mutación , Trastornos Mieloproliferativos/metabolismo , Tirosina Quinasa 3 Similar a fms/metabolismo , Animales , Diferenciación Celular , Supervivencia Celular , Células Cultivadas , Exones , Regulación Neoplásica de la Expresión Génica , Genotipo , Células Madre Hematopoyéticas/patología , Humanos , Estimación de Kaplan-Meier , Leucemia Experimental/metabolismo , Leucemia Experimental/patología , Leucemia Mielomonocítica Crónica/genética , Leucemia Mielomonocítica Crónica/mortalidad , Leucemia Mielomonocítica Crónica/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Madre Multipotentes/patología , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/patología , Fenotipo , Transducción de Señal , Tirosina Quinasa 3 Similar a fms/genética
12.
Cancer Cell ; 12(3): 201-14, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17785202

RESUMEN

To better understand the signaling properties of oncogenic FGFR3, we performed phospho-proteomics studies to identify potential downstream signaling effectors that are tyrosine phosphorylated in hematopoietic cells expressing constitutively activated leukemogenic FGFR3 mutants. We found that FGFR3 directly tyrosine phosphorylates the serine/threonine kinase p90RSK2 at Y529, which consequently regulates RSK2 activation by facilitating inactive ERK binding to RSK2 that is required for ERK-dependent phosphorylation and activation of RSK2. Moreover, inhibition of RSK2 by siRNA or a specific RSK inhibitor fmk effectively induced apoptosis in FGFR3-expressing human t(4;14)-positive myeloma cells. Our findings suggest that FGFR3 mediates hematopoietic transformation by activating RSK2 in a two-step fashion, promoting both the ERK-RSK2 interaction and subsequent phosphorylation of RSK2 by ERK.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Sistema de Señalización de MAP Quinasas , Mieloma Múltiple/enzimología , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/fisiología , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Apoptosis , Sitios de Unión , Línea Celular Tumoral , Proliferación Celular , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Modelos Biológicos , Mieloma Múltiple/metabolismo , Fosforilación , Interferencia de ARN , Proteínas Quinasas S6 Ribosómicas 90-kDa/antagonistas & inhibidores , Tirosina/metabolismo
13.
Cancer Cell ; 12(6): 501-13, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18068628

RESUMEN

Mutations in the juxtamembrane and kinase domains of FLT3 are common in AML, but it is not known whether alterations outside these regions contribute to leukemogenesis. We used a high-throughput platform to interrogate the entire FLT3 coding sequence in AML patients without known FLT3 mutations and experimentally tested the consequences of each candidate leukemogenic allele. This approach identified gain-of-function mutations that activated downstream signaling and conferred sensitivity to FLT3 inhibition and alleles that were not associated with kinase activation, including mutations in the catalytic domain. These findings support the concept that acquired mutations in cancer may not contribute to malignant transformation and underscore the importance of functional studies to distinguish "driver" mutations underlying tumorigenesis from biologically neutral "passenger" alterations.


Asunto(s)
Alelos , Mutación/genética , Tirosina Quinasa 3 Similar a fms/genética , Adulto , Animales , Proliferación Celular/efectos de los fármacos , Análisis Mutacional de ADN , Activación Enzimática/efectos de los fármacos , Humanos , Leucemia Monocítica Aguda/enzimología , Leucemia Monocítica Aguda/genética , Leucemia Monocítica Aguda/patología , Ratones , Proteínas Mutantes/metabolismo , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Estructura Secundaria de Proteína , Transducción de Señal/efectos de los fármacos , Estaurosporina/análogos & derivados , Estaurosporina/farmacología , Tirosina Quinasa 3 Similar a fms/química
14.
Nature ; 462(7270): 182-8, 2009 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-19907488

RESUMEN

Direct inhibition of transcription factor complexes remains a central challenge in the discipline of ligand discovery. In general, these proteins lack surface involutions suitable for high-affinity binding by small molecules. Here we report the design of synthetic, cell-permeable, stabilized alpha-helical peptides that target a critical protein-protein interface in the NOTCH transactivation complex. We demonstrate that direct, high-affinity binding of the hydrocarbon-stapled peptide SAHM1 prevents assembly of the active transcriptional complex. Inappropriate NOTCH activation is directly implicated in the pathogenesis of several disease states, including T-cell acute lymphoblastic leukaemia (T-ALL). The treatment of leukaemic cells with SAHM1 results in genome-wide suppression of NOTCH-activated genes. Direct antagonism of the NOTCH transcriptional program causes potent, NOTCH-specific anti-proliferative effects in cultured cells and in a mouse model of NOTCH1-driven T-ALL.


Asunto(s)
Péptidos/farmacología , Receptor Notch1/antagonistas & inhibidores , Activación Transcripcional/efectos de los fármacos , Animales , Unión Competitiva , Línea Celular Tumoral , Permeabilidad de la Membrana Celular , Proliferación Celular/efectos de los fármacos , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Proteínas de Drosophila/química , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genoma/efectos de los fármacos , Genoma/genética , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Ratones , Modelos Moleculares , Proteínas Nucleares/química , Péptidos/síntesis química , Péptidos/química , Péptidos/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Unión Proteica/efectos de los fármacos , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptor Notch1/química , Receptor Notch1/metabolismo , Transducción de Señal/efectos de los fármacos , Especificidad por Sustrato , Factores de Transcripción/química , Factores de Transcripción/metabolismo
15.
Nature ; 462(7269): 108-12, 2009 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-19847166

RESUMEN

The proto-oncogene KRAS is mutated in a wide array of human cancers, most of which are aggressive and respond poorly to standard therapies. Although the identification of specific oncogenes has led to the development of clinically effective, molecularly targeted therapies in some cases, KRAS has remained refractory to this approach. A complementary strategy for targeting KRAS is to identify gene products that, when inhibited, result in cell death only in the presence of an oncogenic allele. Here we have used systematic RNA interference to detect synthetic lethal partners of oncogenic KRAS and found that the non-canonical IkappaB kinase TBK1 was selectively essential in cells that contain mutant KRAS. Suppression of TBK1 induced apoptosis specifically in human cancer cell lines that depend on oncogenic KRAS expression. In these cells, TBK1 activated NF-kappaB anti-apoptotic signals involving c-Rel and BCL-XL (also known as BCL2L1) that were essential for survival, providing mechanistic insights into this synthetic lethal interaction. These observations indicate that TBK1 and NF-kappaB signalling are essential in KRAS mutant tumours, and establish a general approach for the rational identification of co-dependent pathways in cancer.


Asunto(s)
Genes ras/genética , Proteína Oncogénica p21(ras)/genética , Proteína Oncogénica p21(ras)/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , Alelos , Apoptosis , Línea Celular Tumoral , Supervivencia Celular , Perfilación de la Expresión Génica , Genes Letales , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-rel/metabolismo , Transducción de Señal , Proteína bcl-X/metabolismo
16.
Blood ; 119(6): 1511-21, 2012 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-22160378

RESUMEN

KIT mutations are the most common secondary mutations in inv(16) acute myeloid leukemia (AML) patients and are associated with poor prognosis. It is therefore important to verify that KIT mutations cooperate with CBFB-MYH11, the fusion gene generated by inv(16), for leukemogenesis. Here, we transduced wild-type and conditional Cbfb-MYH11 knockin (KI) mouse bone marrow (BM) cells with KIT D816V/Y mutations. KIT transduction caused massive BM Lin(-) cell death and fewer colonies in culture that were less severe in the KI cells. D816Y KIT but not wild-type KIT enhanced proliferation in Lin(-) cells and led to more mixed lineage colonies from transduced KI BM cells. Importantly, 60% and 80% of mice transplanted with KI BM cells expressing D816V or D816Y KIT, respectively, died from leukemia within 9 months, whereas no control mice died. Results from limiting dilution transplantations indicate higher frequencies of leukemia-initiating cells in the leukemia expressing mutated KIT. Signaling pathway analysis revealed that p44/42 MAPK and Stat3, but not AKT and Stat5, were strongly phosphorylated in the leukemia cells. Finally, leukemia cells carrying KIT D816 mutations were sensitive to the kinase inhibitor PKC412. Our data provide clear evidence for cooperation between mutated KIT and CBFB-MYH11 during leukemogenesis.


Asunto(s)
Leucemia/genética , Mutación , Proteínas de Fusión Oncogénica/genética , Proteínas Proto-Oncogénicas c-kit/genética , Animales , Western Blotting , Trasplante de Médula Ósea , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Progresión de la Enfermedad , Femenino , Citometría de Flujo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Leucemia/metabolismo , Leucemia/patología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT5/metabolismo , Transducción de Señal , Estaurosporina/análogos & derivados , Estaurosporina/farmacología
17.
Cancer Cell ; 10(1): 1-2, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16843258

RESUMEN

In this issue of Cancer Cell, Kovacic and colleagues have reexamined the role of STAT1 in murine models of leukemogenesis. Their studies shed new light on the complex interplay between cell-autonomous contributions and host responsiveness to cancer and elucidate a previously unknown role of STAT1 in tumor progression.


Asunto(s)
Leucemia Experimental/patología , Factor de Transcripción STAT1/fisiología , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Leucemia Experimental/genética , Leucemia Experimental/metabolismo , Ratones , Ratones Noqueados , Factor de Transcripción STAT1/genética
18.
Cancer Cell ; 10(1): 65-75, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16843266

RESUMEN

Tyrosine kinases are aberrantly activated in numerous malignancies, including acute myeloid leukemia (AML). To identify tyrosine kinases activated in AML, we developed a screening strategy that rapidly identifies tyrosine-phosphorylated proteins using mass spectrometry. This allowed the identification of an activating mutation (A572V) in the JAK3 pseudokinase domain in the acute megakaryoblastic leukemia (AMKL) cell line CMK. Subsequent analysis identified two additional JAK3 alleles, V722I and P132T, in AMKL patients. JAK3(A572V), JAK3(V722I), and JAK3(P132T) each transform Ba/F3 cells to factor-independent growth, and JAK3(A572V) confers features of megakaryoblastic leukemia in a murine model. These findings illustrate the biological importance of gain-of-function JAK3 mutations in leukemogenesis and demonstrate the utility of proteomic approaches to identifying clinically relevant mutations.


Asunto(s)
Leucemia Experimental/genética , Leucemia Megacarioblástica Aguda/genética , Proteínas Tirosina Quinasas/genética , Alelos , Animales , Apoptosis/efectos de los fármacos , Benzamidas , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Mesilato de Imatinib , Janus Quinasa 2 , Janus Quinasa 3 , Células K562 , Leucemia Experimental/metabolismo , Leucemia Experimental/patología , Leucemia Megacarioblástica Aguda/metabolismo , Leucemia Megacarioblástica Aguda/patología , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Fosforilación/efectos de los fármacos , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Estructura Terciaria de Proteína/genética , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Pirimidinas/farmacología , ARN Interferente Pequeño/genética , TYK2 Quinasa
19.
Nature ; 455(7215): 975-8, 2008 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-18923525

RESUMEN

Neuroblastoma, an embryonal tumour of the peripheral sympathetic nervous system, accounts for approximately 15% of all deaths due to childhood cancer. High-risk neuroblastomas are rapidly progressive; even with intensive myeloablative chemotherapy, relapse is common and almost uniformly fatal. Here we report the detection of previously unknown mutations in the ALK gene, which encodes a receptor tyrosine kinase, in 8% of primary neuroblastomas. Five non-synonymous sequence variations were identified in the kinase domain of ALK, of which three were somatic and two were germ line. The most frequent mutation, F1174L, was also identified in three different neuroblastoma cell lines. ALK complementary DNAs encoding the F1174L and R1275Q variants, but not the wild-type ALK cDNA, transformed interleukin-3-dependent murine haematopoietic Ba/F3 cells to cytokine-independent growth. Ba/F3 cells expressing these mutations were sensitive to the small-molecule inhibitor of ALK, TAE684 (ref. 4). Furthermore, two human neuroblastoma cell lines harbouring the F1174L mutation were also sensitive to the inhibitor. Cytotoxicity was associated with increased amounts of apoptosis as measured by TdT-mediated dUTP nick end labelling (TUNEL). Short hairpin RNA (shRNA)-mediated knockdown of ALK expression in neuroblastoma cell lines with the F1174L mutation also resulted in apoptosis and impaired cell proliferation. Thus, activating alleles of the ALK receptor tyrosine kinase are present in primary neuroblastoma tumours and in established neuroblastoma cell lines, and confer sensitivity to ALK inhibition with small molecules, providing a molecular rationale for targeted therapy of this disease.


Asunto(s)
Mutación/genética , Neuroblastoma/genética , Neuroblastoma/terapia , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/genética , Alelos , Quinasa de Linfoma Anaplásico , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Activación Enzimática/genética , Genoma Humano/genética , Humanos , Hibridación Fluorescente in Situ , Etiquetado Corte-Fin in Situ , Ratones , Neuroblastoma/enzimología , Neuroblastoma/patología , Polimorfismo de Nucleótido Simple/genética , Estructura Terciaria de Proteína/genética , Proteínas Tirosina Quinasas/química , Proteínas Tirosina Quinasas/metabolismo , Proteínas Tirosina Quinasas Receptoras , Análisis de Secuencia de ADN
20.
Nat Rev Cancer ; 5(4): 311-21, 2005 04.
Artículo en Inglés | MEDLINE | ID: mdl-15803157

RESUMEN

Many cancers seem to depend on a small population of 'cancer stem cells' for their continued growth and propagation. The leukaemia stem cell (LSC) was the first such cell to be described. The origins of these cells are controversial, and their biology - like that of their normal-tissue counterpart, the haematopoietic stem cell (HSC) - is still not fully elucidated. However, the LSC is likely to be the most crucial target in the treatment of leukaemias, and a thorough understanding of its biology - particularly of how the LSC differs from the HSC - might allow it to be selectively targeted, improving therapeutic outcome.


Asunto(s)
Leucemia/patología , Células Madre Neoplásicas/fisiología , Animales , Biomarcadores de Tumor , División Celular , Sistemas de Liberación de Medicamentos , Células Madre Hematopoyéticas/fisiología , Historia del Siglo XIX , Historia del Siglo XX , Humanos , Leucemia/genética , Leucemia/historia , Ratones , Modelos Biológicos , Investigación , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA