Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Cell ; 140(2): 280-93, 2010 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-20141841

RESUMEN

SIRT6 is a member of a highly conserved family of NAD(+)-dependent deacetylases with various roles in metabolism, stress resistance, and life span. SIRT6-deficient mice develop normally but succumb to a lethal hypoglycemia early in life; however, the mechanism underlying this hypoglycemia remained unclear. Here, we demonstrate that SIRT6 functions as a histone H3K9 deacetylase to control the expression of multiple glycolytic genes. Specifically, SIRT6 appears to function as a corepressor of the transcription factor Hif1alpha, a critical regulator of nutrient stress responses. Consistent with this notion, SIRT6-deficient cells exhibit increased Hif1alpha activity and show increased glucose uptake with upregulation of glycolysis and diminished mitochondrial respiration. Our studies uncover a role for the chromatin factor SIRT6 as a master regulator of glucose homeostasis and may provide the basis for novel therapeutic approaches against metabolic diseases, such as diabetes and obesity.


Asunto(s)
Glucosa/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Sirtuinas/metabolismo , Animales , Respiración de la Célula , Transportador de Glucosa de Tipo 1 , Glucólisis , Ratones , Ratones Noqueados , Sirtuinas/genética
2.
EMBO J ; 31(5): 1266-78, 2012 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-22246181

RESUMEN

The cellular response to p53 activation varies greatly in a stimulus- and cell type-specific manner. Dissecting the molecular mechanisms defining these cell fate choices will assist the development of effective p53-based cancer therapies and also illuminate fundamental processes by which gene networks control cellular behaviour. Using an experimental system wherein stimulus-specific p53 responses are elicited by non-genotoxic versus genotoxic agents, we discovered a novel mechanism that determines whether cells undergo proliferation arrest or cell death. Strikingly, we observe that key mediators of cell-cycle arrest (p21, 14-3-3σ) and apoptosis (PUMA, BAX) are equally activated regardless of outcome. In fact, arresting cells display strong translocation of PUMA and BAX to the mitochondria, yet fail to release cytochrome C or activate caspases. Surprisingly, the key differential events in apoptotic cells are p53-dependent activation of the DR4 death receptor pathway, caspase 8-mediated cleavage of BID, and BID-dependent activation of poised BAX at the mitochondria. These results reveal a previously unappreciated role for DR4 and the extrinsic apoptotic pathway in cell fate choice following p53 activation.


Asunto(s)
Apoptosis , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Caspasa 8/metabolismo , Proliferación Celular , Humanos , Mitocondrias/metabolismo , Transporte de Proteínas , Proteínas Proto-Oncogénicas/metabolismo
3.
Genome Res ; 22(8): 1426-36, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22684280

RESUMEN

CBX5, CBX1, and CBX3 (HP1α, ß, and γ, respectively) play an evolutionarily conserved role in the formation and maintenance of heterochromatin. In addition, CBX5, CBX1, and CBX3 may also participate in transcriptional regulation of genes. Recently, CBX3 binding to the bodies of a subset of genes has been observed in human and murine cells. However, the generality of this phenomenon and the role CBX3 may play in this context are unknown. Genome-wide localization analysis reveals CBX3 binding at genic regions, which strongly correlates with gene activity across multiple cell types. Depletion of CBX3 resulted in down-regulation of a subset of target genes. Loss of CBX3 binding leads to a more dramatic accumulation of unspliced nascent transcripts. In addition, we observed defective recruitment of splicing factors, including SNRNP70, to CBX3 target genes. Collectively, our data suggest a role for CBX3 in aiding in efficient cotranscriptional RNA processing.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Genoma Humano , Heterocromatina/metabolismo , Procesamiento Postranscripcional del ARN , Sitios de Unión , Inmunoprecipitación de Cromatina , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/genética , Exones , Regulación de la Expresión Génica , Células HCT116 , Heterocromatina/genética , Humanos , Células K562 , Unión Proteica , Ribonucleoproteína Nuclear Pequeña U1/genética , Ribonucleoproteína Nuclear Pequeña U1/metabolismo , Transcripción Genética , Activación Transcripcional
4.
Nat Chem Biol ; 8(7): 646-54, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22660439

RESUMEN

The p53 tumor suppressor orchestrates alternative stress responses including cell cycle arrest and apoptosis, but the mechanisms defining cell fate upon p53 activation are poorly understood. Several small-molecule activators of p53 have been developed, including Nutlin-3, but their therapeutic potential is limited by the fact that they induce reversible cell cycle arrest in most cancer cell types. We report here the results of a genome-wide short hairpin RNA screen for genes that are lethal in combination with p53 activation by Nutlin-3, which showed that the ATM and MET kinases govern cell fate choice upon p53 activation. Genetic or pharmacological interference with ATM or MET activity converts the cellular response from cell cycle arrest into apoptosis in diverse cancer cell types without affecting expression of key p53 target genes. ATM and MET inhibitors also enable Nutlin-3 to kill tumor spheroids. These results identify new pathways controlling the cellular response to p53 activation and aid in the design of p53-based therapies.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Genes p53 , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Apoptosis/genética , Proteínas de la Ataxia Telangiectasia Mutada , Ciclo Celular , Proteínas de Ciclo Celular/genética , Línea Celular , Proteínas de Unión al ADN/genética , Genes Letales , Genes Sintéticos , Humanos , Imidazoles/metabolismo , Piperazinas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Supresoras de Tumor/genética
5.
Biochim Biophys Acta ; 1825(2): 229-44, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22333261

RESUMEN

The p53 tumor suppressor is embedded in a large gene network controlling diverse cellular and organismal phenotypes. Multiple signaling pathways converge onto p53 activation, mostly by relieving the inhibitory effects of its repressors, MDM2 and MDM4. In turn, signals originating from increased p53 activity diverge into distinct effector pathways to deliver a specific cellular response to the activating stimuli. Much attention has been devoted to dissecting how the various input pathways trigger p53 activation and how the activity of the p53 protein itself can be modulated by a plethora of co-factors and post-translational modifications. In this review we will focus instead on the multiple configurations of the effector pathways. We will discuss how p53-generated signals are transmitted, amplified, resisted and eventually integrated by downstream gene circuits operating at the transcriptional, post-transcriptional and post-translational levels. We will also discuss how context-dependent variations in these gene circuits define the cellular response to p53 activation and how they may impact the clinical efficacy of p53-based targeted therapies.


Asunto(s)
Genes p53 , Proteína p53 Supresora de Tumor/genética , Animales , Apoptosis/genética , Puntos de Control del Ciclo Celular/genética , Humanos , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo
6.
Biotechnol Bioeng ; 110(11): 2826-35, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23719862

RESUMEN

The eukaryotic green algae, Chlamydomonas reinhardtii has been shown to be capable of producing a variety of recombinant proteins, but the true potential of this platform remains largely unexplored. To assess the potential of algae for the production of novel recombinant proteins, we generated a series of chimeric proteins containing a single chain antibody (scFv) targeting the B-cell surface antigen CD22, genetically fused to the eukaryotic ribosome inactivating protein, gelonin, from Gelonium multiflorm. These unique molecules, termed immunotoxins, are encoded as a single gene that produces an antibody--toxin chimeric protein capable of delivering a cytotoxic molecule to targeted B-cells. We show that the addition of an Fc domain of a human IgG1 to these fusion proteins results in the production of assembled dimeric immunotoxins, containing two cell binding scFvs and two gelonin molecules. Additionally, we demonstrate that these algal expressed proteins are capable of binding and reducing the viability of B-cell lymphomas, while treatment of T-cells, that lack the CD22 antigen, had no impact on cell viability. Since other protein expression platforms are incapable of folding and accumulating these complex immunotoxins as soluble and enzymatically active proteins, our studies document a novel and efficient method for immunotoxin production.


Asunto(s)
Antineoplásicos/metabolismo , Chlamydomonas reinhardtii/metabolismo , Inmunotoxinas/metabolismo , Proteínas Inactivadoras de Ribosomas/metabolismo , Linfocitos B/efectos de los fármacos , Linfocitos B/fisiología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Chlamydomonas reinhardtii/genética , Humanos , Inmunoglobulina G/genética , Inmunoglobulina G/metabolismo , Inmunotoxinas/genética , Datos de Secuencia Molecular , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Inactivadoras de Ribosomas/genética , Proteínas Inactivadoras de Ribosomas Tipo 1/genética , Proteínas Inactivadoras de Ribosomas Tipo 1/metabolismo , Análisis de Secuencia de ADN , Lectina 2 Similar a Ig de Unión al Ácido Siálico , Linfocitos T/efectos de los fármacos , Linfocitos T/fisiología
7.
BMC Biotechnol ; 9: 26, 2009 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-19323825

RESUMEN

BACKGROUND: Expression of recombinant proteins in green algal chloroplast holds substantial promise as a platform for the production of human therapeutic proteins. A number of proteins have been expressed in the chloroplast of Chlamydomonas reinhardtii, including complex mammalian proteins, but many of these proteins accumulate to significantly lower levels than do endogenous chloroplast proteins. We examined if recombinant protein accumulation could be enhanced by genetically fusing the recombinant reporter protein, luciferase, to the carboxy-terminal end of an abundant endogenous protein, the large subunit of ribulose bisphosphate carboxylase (Rubisco LSU). Additionally, as recombinant proteins fused to endogenous proteins are of little clinical or commercial value, we explored the possibility of engineering our recombinant protein to be cleavable from the endogenous protein in vivo. This strategy would obviate the need for further in vitro processing steps in order to produce the desired recombinant protein. To achieve this, a native protein-processing site from preferredoxin (preFd) was placed between the Rubisco LSU and luciferase coding regions in the fusion protein construct. RESULTS: The luciferase from the fusion protein accumulated to significantly higher levels than luciferase expressed alone. By eliminating the endogenous Rubisco large subunit gene (rbcL), we achieved a further increase in luciferase accumulation with respect to luciferase expression in the WT background. Importantly, near-wild type levels of functional Rubisco holoenzyme were generated following the proteolytic removal of the fused luciferase, while luciferase activity for the fusion protein was almost ~33 times greater than luciferase expressed alone. These data demonstrate the utility of using fusion proteins to enhance recombinant protein accumulation in algal chloroplasts, and also show that engineered proteolytic processing sites can be used to liberate the exogenous protein from the endogenous fusion partner, allowing for the purification of the intended mature protein. CONCLUSION: These results demonstrate the utility of fusion proteins in algal chloroplast as a method to increase accumulation of recombinant proteins that are difficult to express. Since Rubisco is ubiquitous to land plants and green algae, this strategy may also be applied to higher plant transgenic expression systems.


Asunto(s)
Chlamydomonas reinhardtii/metabolismo , Cloroplastos/metabolismo , Proteínas Recombinantes de Fusión/biosíntesis , Ribulosa-Bifosfato Carboxilasa/metabolismo , Animales , Chlamydomonas reinhardtii/genética , Cloroplastos/genética , Expresión Génica , Genes Reporteros , Vectores Genéticos , Luciferasas/metabolismo , Modelos Moleculares , Organismos Modificados Genéticamente/genética , Organismos Modificados Genéticamente/metabolismo , Procesamiento Postranscripcional del ARN , Transformación Genética
8.
Oncotarget ; 7(36): 57651-57670, 2016 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-27472392

RESUMEN

Lung cancer is the most common cause of cancer death globally with a significant, unmet need for more efficacious treatments. The receptor tyrosine kinase MET has been implicated as an oncogene in numerous cancer subtypes, including non-small cell lung cancer (NSCLC). Here we explore the therapeutic potential of savolitinib (volitinib, AZD6094, HMPL-504), a potent and selective MET inhibitor, in NSCLC. In vitro, savolitinib inhibits MET phosphorylation with nanomolar potency, which correlates with blockade of PI3K/AKT and MAPK signaling as well as MYC down-regulation. In vivo, savolitinib causes inhibition of these pathways and significantly decreases growth of MET-dependent xenografts. To understand resistance mechanisms, we generated savolitinib resistance in MET-amplified NSCLC cell lines and analyzed individual clones. We found that upregulation of MYC and constitutive mTOR pathway activation is a conserved feature of resistant clones that can be overcome by knockdown of MYC or dual mTORC1/2 inhibition. Lastly, we demonstrate that mechanisms of resistance are heterogeneous, arising via a switch to EGFR dependence or by a requirement for PIM signaling. This work demonstrates the efficacy of savolitinib in NSCLC and characterizes acquired resistance, identifying both known and novel mechanisms that may inform combination strategies in the clinic.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Resistencia a Antineoplásicos , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-myc/metabolismo , Pirazinas/química , Serina-Treonina Quinasas TOR/metabolismo , Triazinas/química , Animales , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Regulación hacia Abajo , Receptores ErbB/metabolismo , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-met/metabolismo
9.
Clin Cancer Res ; 21(12): 2811-9, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25779944

RESUMEN

PURPOSE: Papillary renal cell carcinoma (PRCC) is the second most common cancer of the kidney and carries a poor prognosis for patients with nonlocalized disease. The HGF receptor MET plays a central role in PRCC and aberrations, either through mutation, copy number gain, or trisomy of chromosome 7 occurring in the majority of cases. The development of effective therapies in PRCC has been hampered in part by a lack of available preclinical models. We determined the pharmacodynamic and antitumor response of the selective MET inhibitor AZD6094 in two PRCC patient-derived xenograft (PDX) models. EXPERIMENTAL DESIGN: Two PRCC PDX models were identified and MET mutation status and copy number determined. Pharmacodynamic and antitumor activity of AZD6094 was tested using a dose response up to 25 mg/kg daily, representing clinically achievable exposures, and compared with the activity of the RCC standard-of-care sunitinib (in RCC43b) or the multikinase inhibitor crizotinib (in RCC47). RESULTS: AZD6094 treatment resulted in tumor regressions, whereas sunitinib or crizotinib resulted in unsustained growth inhibition. Pharmacodynamic analysis of tumors revealed that AZD6094 could robustly suppress pMET and the duration of target inhibition was dose related. AZD6094 inhibited multiple signaling nodes, including MAPK, PI3K, and EGFR. Finally, at doses that induced tumor regression, AZD6094 resulted in a dose- and time-dependent induction of cleaved PARP, a marker of cell death. CONCLUSIONS: Data presented provide the first report testing therapeutics in preclinical in vivo models of PRCC and support the clinical development of AZD6094 in this indication.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Papilar/metabolismo , Carcinoma Papilar/patología , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Pirazinas/farmacología , Triazinas/farmacología , Animales , Antineoplásicos/administración & dosificación , Carcinoma Papilar/tratamiento farmacológico , Carcinoma Papilar/genética , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/genética , Línea Celular Tumoral , Crizotinib , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Indoles/farmacología , Inhibidores de Proteínas Quinasas/administración & dosificación , Proteínas Proto-Oncogénicas c-met/genética , Pirazinas/administración & dosificación , Pirazoles/farmacología , Piridinas/farmacología , Pirroles/farmacología , Sunitinib , Triazinas/administración & dosificación , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Cell Cycle ; 7(15): 2427-33, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18677110

RESUMEN

The cellular response to Nutlin-3, a small-molecule inhibitor of the p53 repressor MDM2, varies widely among human cancer-derived cell types. Whereas HCT116 colorectal carcinoma cells display sustained cell cycle arrest, BV173 leukemia cells undergo rapid apoptosis and other cell lines show an intermediate response. We found that the expression of the p53 target genes p21, 14-3-3sigma and the microRNA miR-34a correlates tightly with the cell fate choice adopted. All three genes were strongly induced in arresting cells, but silenced in cells undergoing Nutlin-3-induced apoptosis. In contrast, key apoptotic p53 target genes were equally expressed in arresting and apoptotic cells. Interestingly, we establish that miR-34a cooperates with p21 and 14-3-3sigma to override the apoptotic signals generated by p53 activation. Strikingly, p53 binding to chromatin and p53-mediated recruitment of certain coactivators to all three target loci does not vary among cell types. Instead, the cell type-specific silencing of these genes is due to enhanced p21 mRNA degradation, 14-3-3sigma promoter DNA methylation and reduced processing of the miR-34a primary transcript. Thus, p53-independent events regulating expression of protein-coding genes and microRNAs within the network can define the cellular outcome of p53 activation.


Asunto(s)
Silenciador del Gen/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/fisiología , Proteínas 14-3-3/genética , Apoptosis/efectos de los fármacos , Apoptosis/genética , Carcinoma/genética , Carcinoma/patología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Regulación de la Expresión Génica , Células HCT116 , Humanos , Imidazoles/farmacología , MicroARNs/fisiología , Modelos Biológicos , Piperazinas/farmacología , Transducción de Señal/genética , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA