Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Mol Cell ; 67(1): 128-138.e7, 2017 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-28648777

RESUMEN

Mutations in cancer reprogram amino acid metabolism to drive tumor growth, but the molecular mechanisms are not well understood. Using an unbiased proteomic screen, we identified mTORC2 as a critical regulator of amino acid metabolism in cancer via phosphorylation of the cystine-glutamate antiporter xCT. mTORC2 phosphorylates serine 26 at the cytosolic N terminus of xCT, inhibiting its activity. Genetic inhibition of mTORC2, or pharmacologic inhibition of the mammalian target of rapamycin (mTOR) kinase, promotes glutamate secretion, cystine uptake, and incorporation into glutathione, linking growth factor receptor signaling with amino acid uptake and utilization. These results identify an unanticipated mechanism regulating amino acid metabolism in cancer, enabling tumor cells to adapt to changing environmental conditions.


Asunto(s)
Sistema de Transporte de Aminoácidos y+/metabolismo , Neoplasias Encefálicas/enzimología , Cisteína/metabolismo , Glioblastoma/enzimología , Glutamina/metabolismo , Complejos Multiproteicos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Células A549 , Sistema de Transporte de Aminoácidos y+/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Glioblastoma/genética , Glioblastoma/patología , Glutatión/biosíntesis , Células HEK293 , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos/genética , Mutación , Fosforilación , Unión Proteica , Proteómica/métodos , Interferencia de ARN , Serina , Serina-Treonina Quinasas TOR/genética , Espectrometría de Masas en Tándem , Factores de Tiempo , Transfección , Microambiente Tumoral
2.
Genes Dev ; 31(12): 1212-1227, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28724615

RESUMEN

In glioblastoma (GBM), heterogeneous expression of amplified and mutated epidermal growth factor receptor (EGFR) presents a substantial challenge for the effective use of EGFR-directed therapeutics. Here we demonstrate that heterogeneous expression of the wild-type receptor and its constitutively active mutant form, EGFRvIII, limits sensitivity to these therapies through an interclonal communication mechanism mediated by interleukin-6 (IL-6) cytokine secreted from EGFRvIII-positive tumor cells. IL-6 activates a NF-κB signaling axis in a paracrine and autocrine manner, leading to bromodomain protein 4 (BRD4)-dependent expression of the prosurvival protein survivin (BIRC5) and attenuation of sensitivity to EGFR tyrosine kinase inhibitors (TKIs). NF-κB and survivin are coordinately up-regulated in GBM patient tumors, and functional inhibition of either protein or BRD4 in in vitro and in vivo models restores sensitivity to EGFR TKIs. These results provide a rationale for improving anti-EGFR therapeutic efficacy through pharmacological uncoupling of a convergence point of NF-κB-mediated survival that is leveraged by an interclonal circuitry mechanism established by intratumoral mutational heterogeneity.


Asunto(s)
Resistencia a Antineoplásicos/genética , Glioblastoma/fisiopatología , FN-kappa B/genética , FN-kappa B/metabolismo , Transducción de Señal/genética , Animales , Comunicación Celular , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Interleucina-6/metabolismo , Ratones , Ratones Desnudos , Mutación , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
3.
Mol Cell ; 60(2): 307-18, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26455392

RESUMEN

Epidermal growth factor receptor (EGFR) gene amplification and mutations are the most common oncogenic events in glioblastoma (GBM), but the mechanisms by which they promote aggressive tumor growth are not well understood. Here, through integrated epigenome and transcriptome analyses of cell lines, genotyped clinical samples, and TCGA data, we show that EGFR mutations remodel the activated enhancer landscape of GBM, promoting tumorigenesis through a SOX9 and FOXG1-dependent transcriptional regulatory network in vitro and in vivo. The most common EGFR mutation, EGFRvIII, sensitizes GBM cells to the BET-bromodomain inhibitor JQ1 in a SOX9, FOXG1-dependent manner. These results identify the role of transcriptional/epigenetic remodeling in EGFR-dependent pathogenesis and suggest a mechanistic basis for epigenetic therapy.


Asunto(s)
Neoplasias Encefálicas/genética , Epigénesis Genética , Receptores ErbB/genética , Factores de Transcripción Forkhead/genética , Glioblastoma/genética , Proteínas del Tejido Nervioso/genética , Factor de Transcripción SOX9/genética , Adulto , Animales , Azepinas/farmacología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Niño , Receptores ErbB/metabolismo , Factores de Transcripción Forkhead/metabolismo , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Ratones , Ratones Desnudos , Mutación , Trasplante de Neoplasias , Proteínas del Tejido Nervioso/metabolismo , Factor de Transcripción SOX9/metabolismo , Transducción de Señal , Transcriptoma , Triazoles/farmacología
4.
J Biol Chem ; 292(47): 19209-19225, 2017 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-28972160

RESUMEN

The ubiquitin-proteasome system (UPS) is responsible for most selective protein degradation in eukaryotes and regulates numerous cellular processes, including cell cycle control and protein quality control. A component of this system, the deubiquitinating enzyme USP14, associates with the proteasome where it can rescue substrates from degradation by removal of the ubiquitin tag. We previously found that a small-molecule inhibitor of USP14, known as IU1, can increase the rate of degradation of a subset of proteasome substrates. We report here the synthesis and characterization of 87 variants of IU1, which resulted in the identification of a 10-fold more potent USP14 inhibitor that retains specificity for USP14. The capacity of this compound, IU1-47, to enhance protein degradation in cells was tested using as a reporter the microtubule-associated protein tau, which has been implicated in many neurodegenerative diseases. Using primary neuronal cultures, IU1-47 was found to accelerate the rate of degradation of wild-type tau, the pathological tau mutants P301L and P301S, and the A152T tau variant. We also report that a specific residue in tau, lysine 174, is critical for the IU1-47-mediated tau degradation by the proteasome. Finally, we show that IU1-47 stimulates autophagic flux in primary neurons. In summary, these findings provide a powerful research tool for investigating the complex biology of USP14.


Asunto(s)
Embrión de Mamíferos/metabolismo , Inhibidores Enzimáticos/farmacología , Fibroblastos/metabolismo , Neuronas/metabolismo , Pirroles/farmacología , Ubiquitina Tiolesterasa/fisiología , Proteínas tau/metabolismo , Animales , Células Cultivadas , Citoplasma/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/efectos de los fármacos , Inhibidores Enzimáticos/síntesis química , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/citología , Neuronas/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Pirroles/síntesis química , Ratas Sprague-Dawley , Ubiquitina/metabolismo , Ubiquitinación
5.
Cancers (Basel) ; 14(23)2022 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-36497283

RESUMEN

Breast cancer is a common cancer in women. Breast cancer cells synthesize large amounts of hyaluronan to assist their proliferation, survival, migration and invasion. Accumulation of hyaluronan and overexpression of its receptor CD44 and hyaluronidase TMEM2 in breast tumors correlate with tumor progression and reduced overall survival of patients. Currently, the only known small molecule inhibitor of hyaluronan synthesis is 4-methyl-umbelliferone (4-MU). Due to the importance of hyaluronan for breast cancer progression, our aim was to identify new, potent and chemically distinct inhibitors of its synthesis. Here, we report a new small molecule inhibitor of hyaluronan synthesis, the thymidine analog 5'-Deoxy-5'-(1,3-Diphenyl-2-Imidazolidinyl)-Thymidine (DDIT). This compound is more potent than 4-MU and displays significant anti-tumorigenic properties. Specifically, DDIT inhibits breast cancer cell proliferation, migration, invasion and cancer stem cell self-renewal by suppressing HAS-synthesized hyaluronan. DDIT appears as a promising lead compound for the development of inhibitors of hyaluronan synthesis with potential usefulness in breast cancer treatment.

6.
Bioorg Med Chem Lett ; 21(22): 6888-94, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21986586

RESUMEN

We have identified and synthesized a series of imidazole containing dimerization inhibitors of inducible nitric oxide synthase (iNOS). The necessity of key imidazole and piperonyl functionality was demonstrated and SAR studies led to the identification of compound 35, which showed a dose dependant inhibition in multiple pain models, including tactile allodynia induced by spinal nerve ligation (Chung model).


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/uso terapéutico , Hiperalgesia/tratamiento farmacológico , Imidazoles/química , Imidazoles/uso terapéutico , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Dolor/tratamiento farmacológico , Multimerización de Proteína/efectos de los fármacos , Animales , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Imidazoles/farmacología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Ratas , Ratas Endogámicas Lew
7.
Cancer Cell ; 35(3): 504-518.e7, 2019 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-30827889

RESUMEN

Ionizing radiation (IR) and chemotherapy are standard-of-care treatments for glioblastoma (GBM) patients and both result in DNA damage, however, the clinical efficacy is limited due to therapeutic resistance. We identified a mechanism of such resistance mediated by phosphorylation of PTEN on tyrosine 240 (pY240-PTEN) by FGFR2. pY240-PTEN is rapidly elevated and bound to chromatin through interaction with Ki-67 in response to IR treatment and facilitates the recruitment of RAD51 to promote DNA repair. Blocking Y240 phosphorylation confers radiation sensitivity to tumors and extends survival in GBM preclinical models. Y240F-Pten knockin mice showed radiation sensitivity. These results suggest that FGFR-mediated pY240-PTEN is a key mechanism of radiation resistance and is an actionable target for improving radiotherapy efficacy.


Asunto(s)
Neoplasias Encefálicas/terapia , Núcleo Celular/metabolismo , Glioma/terapia , Fosfohidrolasa PTEN/metabolismo , Pirimidinas/administración & dosificación , Tolerancia a Radiación/efectos de los fármacos , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Animales , Neoplasias Encefálicas/metabolismo , Reparación del ADN/efectos de los fármacos , Femenino , Glioma/metabolismo , Humanos , Masculino , Ratones , Fosforilación/efectos de los fármacos , Pirimidinas/farmacología , Recombinasa Rad51/metabolismo , Tirosina/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Sci Rep ; 6: 29868, 2016 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-27430378

RESUMEN

Plasticity in epithelial tissues relates to processes of embryonic development, tissue fibrosis and cancer progression. Pharmacological modulation of epithelial transitions during disease progression may thus be clinically useful. Using human keratinocytes and a robotic high-content imaging platform, we screened for chemical compounds that reverse transforming growth factor ß (TGF-ß)-induced epithelial-mesenchymal transition. In addition to TGF-ß receptor kinase inhibitors, we identified small molecule epithelial plasticity modulators including a naturally occurring hydroxysterol agonist of the liver X receptors (LXRs), members of the nuclear receptor transcription factor family. Endogenous and synthetic LXR agonists tested in diverse cell models blocked α-smooth muscle actin expression, myofibroblast differentiation and function. Agonist-dependent LXR activity or LXR overexpression in the absence of ligand counteracted TGF-ß-mediated myofibroblast terminal differentiation and collagen contraction. The protective effect of LXR agonists against TGF-ß-induced pro-fibrotic activity raises the possibility that anti-lipidogenic therapy may be relevant in fibrotic disorders and advanced cancer.


Asunto(s)
Diferenciación Celular/genética , Receptores X del Hígado/genética , Miofibroblastos/citología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Diferenciación Celular/efectos de los fármacos , Colágeno/metabolismo , Desarrollo Embrionario/genética , Células Epiteliales/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Queratinocitos/efectos de los fármacos , Receptores X del Hígado/agonistas , Ratones , Receptores de Factores de Crecimiento Transformadores beta , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Factor de Crecimiento Transformador beta/genética
9.
Mol Cancer Ther ; 15(6): 1217-26, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26983878

RESUMEN

Inhibitors of the bromodomain and extraterminal domain (BET) protein family attenuate the proliferation of several tumor cell lines. These effects are mediated, at least in part, through repression of c-MYC. In colorectal cancer, overexpression of c-MYC due to hyperactive WNT/ß-catenin/TCF signaling is a key driver of tumor progression; however, effective strategies to target this oncogene remain elusive. Here, we investigated the effect of BET inhibitors (BETi) on colorectal cancer cell proliferation and c-MYC expression. Treatment of 20 colorectal cancer cell lines with the BETi JQ1 identified a subset of highly sensitive lines. JQ1 sensitivity was higher in cell lines with microsatellite instability but was not associated with the CpG island methylator phenotype, c-MYC expression or amplification status, BET protein expression, or mutation status of TP53, KRAS/BRAF, or PIK3CA/PTEN Conversely, JQ1 sensitivity correlated significantly with the magnitude of c-MYC mRNA and protein repression. JQ1-mediated c-MYC repression was not due to generalized attenuation of ß-catenin/TCF-mediated transcription, as JQ1 had minimal effects on other ß-catenin/TCF target genes or ß-catenin/TCF reporter activity. BETi preferentially target super-enhancer-regulated genes, and a super-enhancer in c-MYC was recently identified in HCT116 cells to which BRD4 and effector transcription factors of the WNT/ß-catenin/TCF and MEK/ERK pathways are recruited. Combined targeting of c-MYC with JQ1 and inhibitors of these pathways additively repressed c-MYC and proliferation of HCT116 cells. These findings demonstrate that BETi downregulate c-MYC expression and inhibit colorectal cancer cell proliferation and identify strategies for enhancing the effects of BETi on c-MYC repression by combinatorial targeting the c-MYC super-enhancer. Mol Cancer Ther; 15(6); 1217-26. ©2016 AACR.


Asunto(s)
Azepinas/administración & dosificación , Neoplasias Colorrectales/tratamiento farmacológico , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-myc/genética , Piridonas/administración & dosificación , Pirimidinonas/administración & dosificación , Triazoles/administración & dosificación , Vía de Señalización Wnt/efectos de los fármacos , Animales , Azepinas/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HCT116 , Humanos , Ratones , Trasplante de Neoplasias , Proteínas Proto-Oncogénicas c-myc/metabolismo , Piridonas/farmacología , Pirimidinonas/farmacología , Triazoles/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Cancer Cell ; 30(5): 683-693, 2016 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-27746144

RESUMEN

Small-molecule inhibitors targeting growth factor receptors have failed to show efficacy for brain cancers, potentially due to their inability to achieve sufficient drug levels in the CNS. Targeting non-oncogene tumor co-dependencies provides an alternative approach, particularly if drugs with high brain penetration can be identified. Here we demonstrate that the highly lethal brain cancer glioblastoma (GBM) is remarkably dependent on cholesterol for survival, rendering these tumors sensitive to Liver X receptor (LXR) agonist-dependent cell death. We show that LXR-623, a clinically viable, highly brain-penetrant LXRα-partial/LXRß-full agonist selectively kills GBM cells in an LXRß- and cholesterol-dependent fashion, causing tumor regression and prolonged survival in mouse models. Thus, a metabolic co-dependency provides a pharmacological means to kill growth factor-activated cancers in the CNS.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Colesterol/metabolismo , Glioblastoma/tratamiento farmacológico , Indazoles/administración & dosificación , Receptores X del Hígado/metabolismo , Animales , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Glioblastoma/metabolismo , Humanos , Indazoles/farmacología , Ratones , Resultado del Tratamiento
11.
Science ; 348(6239): 1155-60, 2015 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-25931445

RESUMEN

Centrioles are ancient organelles that build centrosomes, the major microtubule-organizing centers of animal cells. Extra centrosomes are a common feature of cancer cells. To investigate the importance of centrosomes in the proliferation of normal and cancer cells, we developed centrinone, a reversible inhibitor of Polo-like kinase 4 (Plk4), a serine-threonine protein kinase that initiates centriole assembly. Centrinone treatment caused centrosome depletion in human and other vertebrate cells. Centrosome loss irreversibly arrested normal cells in a senescence-like G1 state by a p53-dependent mechanism that was independent of DNA damage, stress, Hippo signaling, extended mitotic duration, or segregation errors. In contrast, cancer cell lines with normal or amplified centrosome numbers could proliferate indefinitely after centrosome loss. Upon centrinone washout, each cancer cell line returned to an intrinsic centrosome number "set point." Thus, cells with cancer-associated mutations fundamentally differ from normal cells in their response to centrosome loss.


Asunto(s)
Centriolos/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Pirimidinas/farmacología , Sulfonas/farmacología , Animales , Línea Celular Tumoral , Proliferación Celular , Humanos , Ratones , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/química , Pirimidinas/química , Sulfonas/química
12.
Front Oncol ; 5: 285, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26732741

RESUMEN

Aurora kinases are essential for cell division and are frequently misregulated in human cancers. Based on their potential as cancer therapeutics, a plethora of small molecule Aurora kinase inhibitors have been developed, with a subset having been adopted as tools in cell biology. Here, we fill a gap in the characterization of Aurora kinase inhibitors by using biochemical and cell-based assays to systematically profile a panel of 10 commercially available compounds with reported selectivity for Aurora A (MLN8054, MLN8237, MK-5108, MK-8745, Genentech Aurora Inhibitor 1), Aurora B (Hesperadin, ZM447439, AZD1152-HQPA, GSK1070916), or Aurora A/B (VX-680). We quantify the in vitro effect of each inhibitor on the activity of Aurora A alone, as well as Aurora A and Aurora B bound to fragments of their activators, TPX2 and INCENP, respectively. We also report kinome profiling results for a subset of these compounds to highlight potential off-target effects. In a cellular context, we demonstrate that immunofluorescence-based detection of LATS2 and histone H3 phospho-epitopes provides a facile and reliable means to assess potency and specificity of Aurora A versus Aurora B inhibition, and that G2 duration measured in a live imaging assay is a specific readout of Aurora A activity. Our analysis also highlights variation between HeLa, U2OS, and hTERT-RPE1 cells that impacts selective Aurora A inhibition. For Aurora B, all four tested compounds exhibit excellent selectivity and do not significantly inhibit Aurora A at effective doses. For Aurora A, MK-5108 and MK-8745 are significantly more selective than the commonly used inhibitors MLN8054 and MLN8237. A crystal structure of an Aurora A/MK-5108 complex that we determined suggests the chemical basis for this higher specificity. Taken together, our quantitative biochemical and cell-based analyses indicate that AZD1152-HQPA and MK-8745 are the best current tools for selectively inhibiting Aurora B and Aurora A, respectively. However, MK-8745 is not nearly as ideal as AZD1152-HQPA in that it requires high concentrations to achieve full inhibition in a cellular context, indicating a need for more potent Aurora A-selective inhibitors. We conclude with a set of "good practice" guidelines for the use of Aurora inhibitors in cell biology experiments.

15.
Curr Chem Genomics ; 2: 1-9, 2008 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-20161838

RESUMEN

The transcription of inducible nitric oxide synthase (iNOS) is activated by a network of proinflammatory signaling pathways. Here we describe the identification of a small molecule that downregulates the expression of iNOS mRNA and protein in cytokine-activated cells and suppresses nitric oxide production in vivo. Mechanistic analysis suggests that this small molecule, erstressin, also activates the unfolded protein response (UPR), a signaling pathway triggered by endoplasmic reticulum stress. Erstressin induces rapid phosphorylation of eIF2alpha and the alternative splicing of XBP-1, hallmark initiating events of the UPR. Further, erstressin activates the transcription of multiple genes involved in the UPR. These data suggest an inverse relationship between UPR activation and iNOS mRNA and protein expression under proinflammatory conditions.

16.
Bioorg Med Chem Lett ; 17(13): 3562-9, 2007 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-17498954

RESUMEN

We have identified and synthesized a series of thiophene containing inhibitors of kinesin spindle protein. SAR studies led to the synthesis of 33, which was co-crystallized with KSP and determined to bind to an allosteric pocket previously described for other known KSP inhibitors.


Asunto(s)
Química Farmacéutica/métodos , Cinesinas/antagonistas & inhibidores , Tiofenos/química , Sitio Alostérico , Amidas/química , Antimitóticos/farmacología , Línea Celular Tumoral , Cristalización , Cristalografía por Rayos X , Diseño de Fármacos , Humanos , Concentración 50 Inhibidora , Cinesinas/química , Modelos Químicos , Modelos Moleculares , Conformación Molecular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA