Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 33(2): 145-6, 2009 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-19187757

RESUMEN

Activation of Bax and Bak by BH3-only molecules triggers mitochondrial apoptosis. In a recent issue of Molecular Cell, Fu et al. (2009) identify a constitutively active isoform of Bax, Baxbeta, whose activity is tightly controlled by the ubiquitin-proteasome system.


Asunto(s)
Empalme Alternativo/fisiología , Proteína X Asociada a bcl-2/metabolismo , Apoptosis , Mitocondrias/metabolismo , Modelos Biológicos , Complejo de la Endopetidasa Proteasomal/metabolismo , Isoformas de Proteínas/metabolismo , Ubiquitinas/metabolismo
2.
Mol Cell ; 36(3): 487-99, 2009 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-19917256

RESUMEN

While activation of BAX/BAK by BH3-only molecules (BH3s) is essential for mitochondrial apoptosis, the underlying mechanisms remain unsettled. Here we demonstrate that BAX undergoes stepwise structural reorganization leading to mitochondrial targeting and homo-oligomerization. The alpha1 helix of BAX keeps the alpha9 helix engaged in the dimerization pocket, rendering BAX as a monomer in cytosol. The activator BH3s, tBID/BIM/PUMA, attack and expose the alpha1 helix of BAX, resulting in secondary disengagement of the alpha9 helix and thereby mitochondrial insertion. Activator BH3s remain associated with the N-terminally exposed BAX through the BH1 domain to drive homo-oligomerization. BAK, an integral mitochondrial membrane protein, has bypassed the first activation step, explaining why its killing kinetics are faster than those of BAX. Furthermore, death signals initiated at ER induce BIM and PUMA to activate mitochondrial apoptosis. Accordingly, deficiency of Bim/Puma impedes ER stress-induced BAX/BAK activation and apoptosis. Our study provides mechanistic insights regarding the spatiotemporal execution of BAX/BAK-governed cell death.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/genética , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/genética , Proteína 11 Similar a Bcl2 , Células Cultivadas , Etopósido/farmacología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Immunoblotting , Inmunoprecipitación , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Microscopía Fluorescente , Mitocondrias/metabolismo , Modelos Biológicos , Mutación , Unión Proteica/efectos de los fármacos , Multimerización de Proteína , Proteínas Proto-Oncogénicas/genética , Estaurosporina/farmacología , Tapsigargina/farmacología , Proteínas Supresoras de Tumor/genética , Tunicamicina/farmacología , Proteína Destructora del Antagonista Homólogo bcl-2/química , Proteína Destructora del Antagonista Homólogo bcl-2/genética , Proteína X Asociada a bcl-2/química , Proteína X Asociada a bcl-2/genética
3.
Nature ; 467(7313): 343-6, 2010 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-20818375

RESUMEN

Cell cycle checkpoints are implemented to safeguard the genome, avoiding the accumulation of genetic errors. Checkpoint loss results in genomic instability and contributes to the evolution of cancer. Among G1-, S-, G2- and M-phase checkpoints, genetic studies indicate the role of an intact S-phase checkpoint in maintaining genome integrity. Although the basic framework of the S-phase checkpoint in multicellular organisms has been outlined, the mechanistic details remain to be elucidated. Human chromosome-11 band-q23 translocations disrupting the MLL gene lead to poor prognostic leukaemias. Here we assign MLL as a novel effector in the mammalian S-phase checkpoint network and identify checkpoint dysfunction as an underlying mechanism of MLL leukaemias. MLL is phosphorylated at serine 516 by ATR in response to genotoxic stress in the S phase, which disrupts its interaction with, and hence its degradation by, the SCF(Skp2) E3 ligase, leading to its accumulation. Stabilized MLL protein accumulates on chromatin, methylates histone H3 lysine 4 at late replication origins and inhibits the loading of CDC45 to delay DNA replication. Cells deficient in MLL showed radioresistant DNA synthesis and chromatid-type genomic abnormalities, indicative of S-phase checkpoint dysfunction. Reconstitution of Mll(-/-) (Mll also known as Mll1) mouse embryonic fibroblasts with wild-type but not S516A or ΔSET mutant MLL rescues the S-phase checkpoint defects. Moreover, murine myeloid progenitor cells carrying an Mll-CBP knock-in allele that mimics human t(11;16) leukaemia show a severe radioresistant DNA synthesis phenotype. MLL fusions function as dominant negative mutants that abrogate the ATR-mediated phosphorylation/stabilization of wild-type MLL on damage to DNA, and thus compromise the S-phase checkpoint. Together, our results identify MLL as a key constituent of the mammalian DNA damage response pathway and show that deregulation of the S-phase checkpoint incurred by MLL translocations probably contributes to the pathogenesis of human MLL leukaemias.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Fase S/fisiología , Alelos , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Línea Celular , Cromatina/metabolismo , Daño del ADN , Replicación del ADN/fisiología , Genes Dominantes/genética , Inestabilidad Genómica/fisiología , N-Metiltransferasa de Histona-Lisina , Histonas/química , Histonas/metabolismo , Humanos , Leucemia/genética , Lisina/metabolismo , Metilación , Ratones , Células Progenitoras Mieloides/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/química , Proteína de la Leucemia Mieloide-Linfoide/deficiencia , Proteína de la Leucemia Mieloide-Linfoide/genética , Fosforilación , Fosfoserina/metabolismo , Unión Proteica , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Transducción de Señal , Translocación Genética/genética
4.
Blood ; 118(25): 6544-52, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22012064

RESUMEN

The mixed-lineage leukemia (MLL) H3K4 methyltransferase protein, and the heterodimeric RUNX1/CBFß transcription factor complex, are critical for definitive and adult hematopoiesis, and both are frequently targeted in human acute leukemia. We identified a physical and functional interaction between RUNX1 (AML1) and MLL and show that both are required to maintain the histone lysine 4 trimethyl mark (H3K4me3) at 2 critical regulatory regions of the AML1 target gene PU.1. Similar to CBFß, we show that MLL binds to AML1 abrogating its proteasome-dependent degradation. Furthermore, a subset of previously uncharacterized frame-shift and missense mutations at the N terminus of AML1, found in MDS and AML patients, impairs its interaction with MLL, resulting in loss of the H3K4me3 mark within PU.1 regulatory regions, and decreased PU.1 expression. The interaction between MLL and AML1 provides a mechanism for the sequence-specific binding of MLL to DNA, and identifies RUNX1 target genes as potential effectors of MLL function.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Histonas/metabolismo , Mutación , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas Proto-Oncogénicas/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Transactivadores/genética , Enfermedad Aguda , Western Blotting , Línea Celular , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad beta del Factor de Unión al Sitio Principal/genética , Subunidad beta del Factor de Unión al Sitio Principal/metabolismo , Expresión Génica , Células HEK293 , N-Metiltransferasa de Histona-Lisina , Humanos , Leucemia Mieloide/genética , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patología , Lisina/metabolismo , Metilación , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/metabolismo , Síndromes Mielodisplásicos/patología , Proteína de la Leucemia Mieloide-Linfoide/genética , Unión Proteica , Proteínas Proto-Oncogénicas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transactivadores/metabolismo
5.
Nat Cell Biol ; 8(12): 1348-58, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17115033

RESUMEN

Although the BCL-2 family constitutes a crucial checkpoint in apoptosis, the intricate interplay between these family members remains elusive. Here, we demonstrate that BIM and PUMA, similar to truncated BID (tBID), directly activate BAX-BAK to release cytochrome c. Conversely, anti-apoptotic BCL-2-BCL-X(L)-MCL-1 sequesters these 'activator' BH3-only molecules into stable complexes, thus preventing the activation of BAX-BAK. Extensive mutagenesis of BAX-BAK indicates that their activity is not kept in check by BCL-2-BCL-X(L)-MCL-1. Anti-apoptotic BCL-2 members are differentially inactivated by the remaining 'inactivator' BH3-only molecules including BAD, NOXA, BMF, BIK/BLK and HRK/DP5. BAD displaces tBID, BIM or PUMA from BCL-2-BCL-X(L) to activate BAX-BAK, whereas NOXA specifically antagonizes MCL-1. Coexpression of BAD and NOXA killed wild-type but not Bax, Bak doubly deficient cells or Puma deficient cells with Bim knockdown, indicating that activator BH3-only molecules function downstream of inactivator BH3-only molecules to activate BAX-BAK. Our data establish a hierarchical regulation of mitochondrion-dependent apoptosis by various BCL-2 subfamilies.


Asunto(s)
Apoptosis , Mitocondrias/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Proteína 11 Similar a Bcl2 , Citocromos c/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Proteína bcl-X/metabolismo
6.
Proc Natl Acad Sci U S A ; 106(4): 1093-8, 2009 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-19144918

RESUMEN

Three forms of cell death have been described: apoptosis, autophagic cell death, and necrosis. Although genetic and biochemical studies have formulated a detailed blueprint concerning the apoptotic network, necrosis is generally perceived as a passive cellular demise resulted from unmanageable physical damages. Here, we conclude an active de novo genetic program underlying DNA damage-induced necrosis, thus assigning necrotic cell death as a form of "programmed cell death." Cells deficient of the essential mitochondrial apoptotic effectors, BAX and BAK, ultimately succumbed to DNA damage, exhibiting signature necrotic characteristics. Importantly, this genotoxic stress-triggered necrosis was abrogated when either transcription or translation was inhibited. We pinpointed the p53-cathepsin axis as the quintessential framework underlying necrotic cell death. p53 induces cathepsin Q that cooperates with reactive oxygen species (ROS) to execute necrosis. Moreover, we presented the in vivo evidence of p53-activated necrosis in tumor allografts. Current study lays the foundation for future experimental and therapeutic discoveries aimed at "programmed necrotic death."


Asunto(s)
Catepsinas/metabolismo , Cisteína Endopeptidasas/metabolismo , Daño del ADN , Necrosis/patología , Especies Reactivas de Oxígeno/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Catepsinas/genética , Línea Celular Tumoral , Cisteína Endopeptidasas/genética , Fibroblastos/patología , Fibroblastos/ultraestructura , Humanos , Ratones , Trasplante de Neoplasias , Neoplasias/patología , Neoplasias/ultraestructura , Activación Transcripcional/genética , Trasplante Homólogo , Proteína Destructora del Antagonista Homólogo bcl-2/deficiencia , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/deficiencia , Proteína X Asociada a bcl-2/metabolismo
7.
Blood ; 113(7): 1395-6, 2009 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-19221041

RESUMEN

MLL5 is an MLL family protein and a candidate tumor suppressor located within the human chromosome band 7q22 that is frequently deleted in myeloid malignancies. In this issue of Blood, 3 independent studies report the first genetic analysis of MLL5 deficiency in mice. All 3 strains of MLL5 knockout mice exhibited defects in hematopoiesis, highlighting the critical role of MLL5 in hematopoietic stem cell functions.

8.
Mol Cell Biol ; 27(20): 7063-72, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17698583

RESUMEN

Site-specific proteolytic processing plays important roles in the regulation of cellular activities. The histone modification activity of the human trithorax group mixed-lineage leukemia (MLL) protein and the cell cycle regulatory activity of the cell proliferation factor herpes simplex virus host cell factor 1 (HCF-1) are stimulated by cleavage of precursors that generates stable heterodimeric complexes. MLL is processed by a protease called taspase 1, whereas the precise mechanisms of HCF-1 maturation are unclear, although they are known to depend on a series of sequence repeats called HCF-1(PRO) repeats. We demonstrate here that the Drosophila homologs of MLL and HCF-1, called Trithorax and dHCF, are both cleaved by Drosophila taspase 1. Although highly related, the human and Drosophila taspase 1 proteins display cognate species specificity. Thus, human taspase 1 preferentially cleaves MLL and Drosophila taspase 1 preferentially cleaves Trithorax, consistent with coevolution of taspase 1 and MLL/Trithorax proteins. HCF proteins display even greater species-specific divergence in processing: whereas dHCF is cleaved by the Drosophila taspase 1, human and mouse HCF-1 maturation is taspase 1 independent. Instead, human and Xenopus HCF-1PRO repeats are cleaved in vitro by a human proteolytic activity with novel properties. Thus, from insects to humans, HCF proteins have conserved proteolytic maturation but evolved different mechanisms.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Drosophila/metabolismo , Endopeptidasas/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Precursores de Proteínas/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Cromosómicas no Histona/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Endopeptidasas/genética , Estabilidad de Enzimas , Evolución Molecular , Células HeLa , Humanos , Ratones , Datos de Secuencia Molecular , Proteína de la Leucemia Mieloide-Linfoide/genética , Inhibidores de Proteasas/metabolismo , Precursores de Proteínas/genética , Interferencia de ARN , Alineación de Secuencia , Especificidad por Sustrato
9.
Mol Cell Biol ; 26(7): 2728-35, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16537915

RESUMEN

In higher eukaryotes, the large subunit of the general transcription factor TFIIA is encoded by the single TFIIAalphabeta gene and posttranslationally cleaved into alpha and beta subunits. The molecular mechanisms and biological significance of this proteolytic process have remained obscure. Here, we show that TFIIA is a substrate of taspase 1 as reported for the trithorax group mixed-lineage leukemia protein. We demonstrate that recombinant taspase 1 cleaves TFIIA in vitro. Transfected taspase 1 enhances cleavage of TFIIA, and RNA interference knockdown of endogenous taspase 1 diminishes cleavage of TFIIA in vivo. In taspase 1-/- MEF cells, only uncleaved TFIIA is detected. In Xenopus laevis embryos, knockdown of TFIIA results in phenotype and expression defects. Both defects can be rescued by expression of an uncleavable TFIIA mutant. Our study shows that uncleaved TFIIA is transcriptionally active and that cleavage of TFIIA does not serve to render TFIIA competent for transcription. We propose that cleavage fine tunes the transcription regulation of a subset of genes during differentiation and development.


Asunto(s)
Endopeptidasas/metabolismo , Procesamiento Proteico-Postraduccional , Factor de Transcripción TFIIA/metabolismo , Transcripción Genética/genética , Secuencia de Aminoácidos , Animales , Extractos Celulares , Núcleo Celular/metabolismo , Embrión no Mamífero/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células HeLa , Humanos , Datos de Secuencia Molecular , Mutación/genética , Péptido Hidrolasas/metabolismo , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato , Factor de Transcripción TFIIA/química , Xenopus
10.
Bioorg Med Chem Lett ; 19(17): 5086-90, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19631530

RESUMEN

Taspase1 is a threonine protease responsible for cleaving MLL (Mixed-Lineage Leukemia) to achieve proper HOX gene expression. Subsequent studies identified additional Taspase1 substrates including Transcription Factor IIA (TFIIA) and Drosophila HCF. Taspase1 is essential for cell proliferation and is overexpressed in many cancer cell lines. Currently no small molecule inhibitors of this enzyme have been described. Here, we report the synthesis and evaluation of vinyl sulfone, vinyl ketone, epoxy ketone, and boronic acid inhibitors designed based on the preferred Taspase1 cleavage site (Ac-Ile-Ser-Gln-Leu-Asp). Specifically, we evaluated compounds in which the reactive warhead is positioned in place of the P1 aspartic acid side chain as well as at the C-terminus of the peptide. Interestingly, both classes of inhibitors were effective and vinyl ketones and vinyl sulfones showed the greatest potency for the target protease. These results suggest that Taspase1 has unique substrate recognition properties that could potentially be exploited in the design of potent and selective inhibitors of this enzyme.


Asunto(s)
Ácidos Borónicos/síntesis química , Endopeptidasas/química , Cetonas/síntesis química , Inhibidores de Proteasas/síntesis química , Sulfonas/síntesis química , Secuencia de Aminoácidos , Ácidos Borónicos/química , Ácidos Borónicos/farmacología , Diseño de Fármacos , Endopeptidasas/metabolismo , Humanos , Cetonas/química , Cetonas/farmacología , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacología , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/metabolismo , Sulfonas/química , Sulfonas/farmacología
11.
Mol Cell Biol ; 23(1): 186-94, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12482972

RESUMEN

The mixed-lineage leukemia gene (MLL, ALL1, HRX) encodes a 3,969-amino-acid nuclear protein homologous to Drosophila trithorax and is required to maintain proper Hox gene expression. Chromosome translocations in human leukemia disrupt MLL (11q23), generating chimeric proteins between the N terminus of MLL and multiple translocation partners. Here we report that MLL is normally cleaved at two conserved sites (D/GADD and D/GVDD) and that mutation of these sites abolishes the proteolysis. MLL cleavage generates N-terminal p320 (N320) and C-terminal p180 (C180) fragments, which form a stable complex that localizes to a subnuclear compartment. The FYRN domain of N320 directly interacts with the FYRC and SET domains of C180. Disrupting the interaction between N320 and C180 leads to a marked decrease in the level of N320 and a redistribution of C180 to a diffuse nuclear pattern. These data suggest a model in which a dynamic post-cleavage association confers stability to N320 and correct nuclear sublocalization of the complex, to control the availability of N320 for target genes. This predicts that MLL fusion proteins of leukemia which would lose the ability to complex with C180 have their stability conferred instead by the fusion partners, thus providing one mechanism for altered target gene expression.


Asunto(s)
Estructuras del Núcleo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Proto-Oncogenes , Factores de Transcripción , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Células Cultivadas , Secuencia Conservada , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , N-Metiltransferasa de Histona-Lisina , Humanos , Mutación , Proteína de la Leucemia Mieloide-Linfoide , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Estructura Terciaria de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
12.
Sci Signal ; 6(268): ra20, 2013 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-23532334

RESUMEN

The clinical efficacy of tyrosine kinase inhibitors supports the dependence of distinct subsets of cancers on specific driver mutations for survival, a phenomenon called "oncogene addiction." We demonstrate that PUMA and BIM are the key apoptotic effectors of tyrosine kinase inhibitors in breast cancers with amplification of the gene encoding human epidermal growth factor receptor 2 (HER2) and lung cancers with epidermal growth factor receptor (EGFR) mutants. The BH3 domain containing proteins BIM and PUMA can directly activate the proapoptotic proteins BAX and BAK to permeabilize mitochondria, leading to caspase activation and apoptosis. We delineated the signal transduction pathways leading to the induction of BIM and PUMA by tyrosine kinase inhibitors. Inhibition of the mitogen-activated or extracellular signal-regulated protein kinase kinase (MEK)-extracellular signal-regulated kinase (ERK) pathway caused increased abundance of BIM, whereas antagonizing the phosphoinositide 3-kinase (PI3K)-AKT pathway triggered nuclear translocation of the FOXO transcription factors, which directly activated the PUMA promoter. In a mouse breast tumor model, the abundance of PUMA and BIM was increased after inactivation of HER2. Moreover, deficiency of Bim or Puma impaired caspase activation and reduced tumor regression caused by inactivation of HER2. Similarly, deficiency of Puma impeded the regression of EGFR(L858R)-driven mouse lung tumors upon inactivation of the EGFR-activating mutant. Overall, our study identified PUMA and BIM as the sentinels that interconnect kinase signaling networks and the mitochondrion-dependent apoptotic program, which offers therapeutic insights for designing novel cell death mechanism-based anticancer strategies.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/genética , Neoplasias de la Mama/metabolismo , Silenciador del Gen/fisiología , Proteínas de la Membrana/metabolismo , Oncogenes/fisiología , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal/fisiología , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteína 11 Similar a Bcl2 , Compuestos de Bifenilo/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Receptores ErbB/metabolismo , Femenino , Citometría de Flujo , Humanos , Immunoblotting , Inmunohistoquímica , Lapatinib , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas de la Membrana/genética , Ratones , Nitrofenoles/farmacología , Oncogenes/genética , Piperazinas/farmacología , Plásmidos/genética , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Quinazolinas , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptor ErbB-2/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Sulfonamidas/farmacología
13.
Nat Med ; 19(1): 50-6, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23223005

RESUMEN

Inactivation of the von Hippel-Lindau tumor suppressor gene, VHL, is an archetypical tumor-initiating event in clear cell renal carcinoma (ccRCC) that leads to the activation of hypoxia-inducible transcription factors (HIFs). However, VHL mutation status in ccRCC is not correlated with clinical outcome. Here we show that during ccRCC progression, cancer cells exploit diverse epigenetic alterations to empower a branch of the VHL-HIF pathway for metastasis, and the strength of this activation is associated with poor clinical outcome. By analyzing metastatic subpopulations of VHL-deficient ccRCC cells, we discovered an epigenetically altered VHL-HIF response that is specific to metastatic ccRCC. Focusing on the two most prominent pro-metastatic VHL-HIF target genes, we show that loss of Polycomb repressive complex 2 (PRC2)-dependent histone H3 Lys27 trimethylation (H3K27me3) activates HIF-driven chemokine (C-X-C motif) receptor 4 (CXCR4) expression in support of chemotactic cell invasion, whereas loss of DNA methylation enables HIF-driven cytohesin 1 interacting protein (CYTIP) expression to protect cancer cells from death cytokine signals. Thus, metastasis in ccRCC is based on an epigenetically expanded output of the tumor-initiating pathway.


Asunto(s)
Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/secundario , Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Receptores CXCR4/metabolismo , Factores de Transcripción/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Animales , Secuencia de Bases , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Histonas/metabolismo , Humanos , Neoplasias Renales/genética , Ratones , Ratones Endogámicos NOD , Ratones SCID , Metástasis de la Neoplasia , Complejo Represivo Polycomb 2/genética , Receptores CXCR4/genética , Análisis de Secuencia de ADN , Factores de Transcripción/genética
14.
Cancer Res ; 72(3): 736-46, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22166309

RESUMEN

The threonine endopeptidase Taspase1 has a critical role in cancer cell proliferation and apoptosis. In this study, we developed and evaluated small molecule inhibitors of Taspase1 as a new candidate class of therapeutic modalities. Genetic deletion of Taspase1 in the mouse produced no overt deficiencies, suggesting the possibility of a wide therapeutic index for use of Taspase1 inhibitors in cancers. We defined the peptidyl motifs recognized by Taspase1 and conducted a cell-based dual-fluorescent proteolytic screen of the National Cancer Institute diversity library to identify Taspase1 inhibitors (TASPIN). On the basis of secondary and tertiary screens the 4-[(4-arsonophenyl)methyl]phenyl] arsonic acid NSC48300 was determined to be the most specific active compound. Structure-activity relationship studies indicated a crucial role for the arsenic acid moiety in mediating Taspase1 inhibition. Additional fluorescence resonance energy transfer-based kinetic analysis characterized NSC48300 as a reversible, noncompetitive inhibitor of Taspase1 (K(i) = 4.22 µmol/L). In the MMTV-neu mouse model of breast cancer and the U251 xenograft model of brain cancer, NSC48300 produced effective tumor growth inhibition. Our results offer an initial preclinical proof-of-concept to develop TASPINs for cancer therapy.


Asunto(s)
Arsenicales/farmacología , Neoplasias Encefálicas/prevención & control , Neoplasias de la Mama/prevención & control , Endopeptidasas/metabolismo , Inhibidores de Proteasas/farmacología , Secuencia de Aminoácidos , Aminoácidos/genética , Aminoácidos/metabolismo , Animales , Sitios de Unión/genética , Neoplasias Encefálicas/enzimología , Neoplasias Encefálicas/patología , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Endopeptidasas/genética , Células HEK293 , Humanos , Cinética , Masculino , Neoplasias Mamarias Experimentales/enzimología , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/prevención & control , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , Bibliotecas de Moléculas Pequeñas , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Cancer Res ; 70(13): 5358-67, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20516119

RESUMEN

Taspase1, the mixed lineage leukemia and TFIIAalpha-beta cleaving protease, enables cell proliferation and permits oncogenic initiation. Here, we show its critical role in cancer maintenance and thus offer a new anticancer target. Taspase1 is overexpressed in primary human cancers, and deficiency of Taspase1 in cancer cells not only disrupts proliferation but also enhances apoptosis. Mechanistically, loss of Taspase1 induces the levels of CDK inhibitors (CDKI: p16, p21, and p27) and reduces the level of antiapoptotic MCL-1. Therapeutically, deficiency of Taspase1 synergizes with chemotherapeutic agents and ABT-737, an inhibitor of BCL-2/BCL-X(L), to kill cancer cells. Taspase1 alone or in conjunction with MYC, RAS, or E1A fails to transform NIH/3T3 cells or primary mouse embryonic fibroblasts, respectively, but plays critical roles in cancer initiation and maintenance. Therefore, Taspase1 is better classified as a "non-oncogene addiction" protease, the inhibition of which may offer a novel anticancer therapeutic strategy. The reliance of oncogenes on subordinate non-oncogenes during tumorigenesis underscores the non-oncogene addiction hypothesis in which a large class of non-oncogenes functions to maintain cancer phenotypes and presents attractive anticancer therapeutic targets. The emergence of successful cancer therapeutics targeting non-oncogenes to which cancers are addicted supports the future development and potential application of small-molecule Taspase1 inhibitors for cancer therapy.


Asunto(s)
Endopeptidasas/genética , Glioblastoma/genética , Melanoma/genética , Animales , Antineoplásicos/farmacología , Apoptosis/genética , Compuestos de Bifenilo/farmacología , Procesos de Crecimiento Celular/genética , Línea Celular Transformada , Línea Celular Tumoral , Endopeptidasas/deficiencia , Endopeptidasas/metabolismo , Genes myc , Genes ras , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Masculino , Melanoma/metabolismo , Melanoma/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Células 3T3 NIH , Nitrofenoles/farmacología , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Sulfonamidas/farmacología , Transducción Genética , Transfección
16.
Science ; 330(6009): 1390-3, 2010 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-21127253

RESUMEN

Although the proteins BAX and BAK are required for initiation of apoptosis at the mitochondria, how BAX and BAK are activated remains unsettled. We provide in vivo evidence demonstrating an essential role of the proteins BID, BIM, and PUMA in activating BAX and BAK. Bid, Bim, and Puma triple-knockout mice showed the same developmental defects that are associated with deficiency of Bax and Bak, including persistent interdigital webs and imperforate vaginas. Genetic deletion of Bid, Bim, and Puma prevented the homo-oligomerization of BAX and BAK, and thereby cytochrome c-mediated activation of caspases in response to diverse death signals in neurons and T lymphocytes, despite the presence of other BH3-only molecules. Thus, many forms of apoptosis require direct activation of BAX and BAK at the mitochondria by a member of the BID, BIM, or PUMA family of proteins.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Proteínas de la Membrana/metabolismo , Neuronas/fisiología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteínas Reguladoras de la Apoptosis/genética , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/deficiencia , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/genética , Proteína 11 Similar a Bcl2 , Caspasas/metabolismo , Células Cultivadas , Cerebelo/citología , Citocromos c/metabolismo , Membranas Intracelulares/metabolismo , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Modelos Biológicos , Permeabilidad , Multimerización de Proteína , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , Estrés Fisiológico , Linfocitos T/fisiología , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética , Proteína Destructora del Antagonista Homólogo bcl-2/química , Proteína Destructora del Antagonista Homólogo bcl-2/genética , Proteína X Asociada a bcl-2/química , Proteína X Asociada a bcl-2/genética
17.
Cancer Biol Ther ; 8(13): 1204-11, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19729989

RESUMEN

Human leukemias with chromosomal band 11q23 aberrations that disrupt the MLL/HRX/ALL-1 gene portend poor prognosis. MLL associated leukemias account for the majority of infant leukemia, approximately 10% of adult de novo leukemia and approximately 33% of therapy related acute leukemia with a balanced chromosome translocation. The 500 kD MLL precursor is processed by Taspase1 to generate mature MLL(N320/C180), which orchestrates many aspects of biology such as embryogenesis, cell cycle, cell fate and stem cell maintenance. Leukemogenic MLL translocations fuse the common MLL N-terminus (approximately 1,400 aa) in frame with more than 60 translocation partner genes (TPGs). Recent studies on MLL and MLL leukemia have greatly advanced our knowledge concerning the normal function of MLL and its deregulation in leukemogenesis. Here, we summarize the critical biological and pathological activities of MLL and MLL fusions, and discuss available models and potential therapeutic targets of MLL associated leukemias.


Asunto(s)
Leucemia/genética , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas de Fusión Oncogénica/genética , Translocación Genética , Animales , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Leucemia/patología , Modelos Biológicos , Transducción de Señal/genética
18.
Sci Signal ; 2(85): ra48, 2009 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-19706873

RESUMEN

The proapoptotic proteins BAX and BAK constitute the mitochondrial apoptotic gateway that executes cellular demise after integrating death signals. The lethal BAK is kept in check by voltage-dependent anion channel 2 (VDAC2), a mammalian-restricted VDAC isoform. Here, we provide evidence showing a critical role for the VADC2-BAK complex in determining thymocyte survival in vivo. Genetic depletion of Vdac2 in the thymus resulted in excessive cell death and hypersensitivity to diverse death stimuli including engagement of the T cell receptor. These phenotypes were completely rescued by the concurrent deletion of Bak but not that of Bax. Thus, the VDAC2-BAK axis provides a mechanism that governs the homeostasis of thymocytes. Our study reveals a sophisticated built-in rheostat that likely fine-tunes immune competence to balance autoimmunity and immunodeficiency.


Asunto(s)
Supresión Clonal/fisiología , Linfocitos T/citología , Canal Aniónico 2 Dependiente del Voltaje/fisiología , Proteína Destructora del Antagonista Homólogo bcl-2/fisiología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Apoptosis/genética , Apoptosis/fisiología , Autoinmunidad/genética , Autoinmunidad/fisiología , Complejo CD3/inmunología , Supresión Clonal/genética , Dimerización , Femenino , Técnicas de Inactivación de Genes , Genotipo , Transporte Iónico/genética , Transporte Iónico/fisiología , Masculino , Ratones , Ratones Noqueados , Membranas Mitocondriales/fisiología , Timo/citología , Canal Aniónico 2 Dependiente del Voltaje/deficiencia , Canal Aniónico 2 Dependiente del Voltaje/genética , Proteína Destructora del Antagonista Homólogo bcl-2/deficiencia , Proteína Destructora del Antagonista Homólogo bcl-2/genética , Proteína X Asociada a bcl-2/fisiología
19.
Cell Cycle ; 7(4): 428-35, 2008 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18235233

RESUMEN

Discovered in 1992 from cloning the gene involved in human leukemias carrying chromosome band 11q23 translocations, the MLL/HRX/ALL-1 gene has since attracted scientists from various disciplines by its diverse functions in normal physiological and pathological processes. MLL is the human orthologue of Drosophila trithorax (trx)-the founding member of trithorax group proteins, Trx-G. Leukemogenic11q23 translocations fuse the common MLL N-terminal 1400aa in-frame with a wide variety of fusion partners that share no structural or functional homology. The 500 kD precursor MLL undergoes evolutionarily conserved site-specific cleavage mediated by Taspase1, generating the mature MLL(N320/C180) heterodimer which methylates histone H3 at lysine 4 with its carboxy-terminal SET domain. Extensive biochemical and genetic studies on MLL/trx have established its critical role in maintaining the expression of Hox/homeotic genes. By contrast, the involvement of MLL in many other essential cellular processes remains unclear. Recent reports including ours began to elucidate the intricate interplay between MLL and the cell cycle machinery, which ensures proper cell cycle phase transitions. Thus, this review will focus on this novel activity of MLL and discuss the implications of its deregulation in MLL leukemias.


Asunto(s)
Ciclo Celular/fisiología , Endopeptidasas/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Leucemia/metabolismo , Modelos Biológicos , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Transducción de Señal/fisiología , Humanos , Proteína de la Leucemia Mieloide-Linfoide/biosíntesis
20.
Genes Dev ; 21(19): 2385-98, 2007 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17908926

RESUMEN

Human chromosome 11q23 translocations disrupting MLL result in poor prognostic leukemias. It fuses the common MLL N-terminal approximately 1400 amino acids in-frame with >60 different partners without shared characteristics. In addition to the well-characterized activity of MLL in maintaining Hox gene expression, our recent studies established an MLL-E2F axis in orchestrating core cell cycle gene expression including Cyclins. Here, we demonstrate a biphasic expression of MLL conferred by defined windows of degradation mediated by specialized cell cycle E3 ligases. Specifically, SCF(Skp2) and APC(Cdc20) mark MLL for degradation at S phase and late M phase, respectively. Abolished peak expression of MLL incurs corresponding defects in G1/S transition and M-phase progression. Conversely, overexpression of MLL blocks S-phase progression. Remarkably, MLL degradation initiates at its N-terminal approximately 1400 amino acids, and tested prevalent MLL fusions are resistant to degradation. Thus, impaired degradation of MLL fusions likely constitutes the universal mechanism underlying all MLL leukemias. Our data conclude an essential post-translational regulation of MLL by the cell cycle ubiquitin/proteasome system (UPS) assures the temporal necessity of MLL in coordinating cell cycle progression.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencia de Aminoácidos , Proteínas Cdc20 , Ciclo Celular , Células HeLa , N-Metiltransferasa de Histona-Lisina , Humanos , Leucemia Mieloide/etiología , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas de Fusión Oncogénica/genética , Procesamiento Proteico-Postraduccional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA