Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Cell ; 163(1): 202-17, 2015 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-26388441

RESUMEN

Cancer cells acquire pathological phenotypes through accumulation of mutations that perturb signaling networks. However, global analysis of these events is currently limited. Here, we identify six types of network-attacking mutations (NAMs), including changes in kinase and SH2 modulation, network rewiring, and the genesis and extinction of phosphorylation sites. We developed a computational platform (ReKINect) to identify NAMs and systematically interpreted the exomes and quantitative (phospho-)proteomes of five ovarian cancer cell lines and the global cancer genome repository. We identified and experimentally validated several NAMs, including PKCγ M501I and PKD1 D665N, which encode specificity switches analogous to the appearance of kinases de novo within the kinome. We discover mutant molecular logic gates, a drift toward phospho-threonine signaling, weakening of phosphorylation motifs, and kinase-inactivating hotspots in cancer. Our method pinpoints functional NAMs, scales with the complexity of cancer genomes and cell signaling, and may enhance our capability to therapeutically target tumor-specific networks.


Asunto(s)
Neoplasias Ováricas/metabolismo , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Transducción de Señal , Femenino , Humanos , Almacenamiento y Recuperación de la Información , Modelos Moleculares , Mutación Puntual , Proteínas Quinasas/química , Programas Informáticos
2.
Apoptosis ; 17(7): 666-78, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22488300

RESUMEN

Acquisition of resistance to anchorage dependant cell death, a process termed anoikis, is a requirement for cancer cell metastasis. However, the molecular determinants of anoikis resistance and sensitivity are poorly understood. To better understand resistance to anoikis we conducted a genome wide lentiviral shRNA screen to identify genes whose knockdown render anoikis-sensitive RWPE-1 prostate cells resistant to anoikis. RWPE-1 cells were infected with a pooled lentiviral shRNA library with 54,021 shRNA targeting 11,255 genes. After infection, an anoikis-resistant cell population was selected and shRNA sequences were amplified and sequenced. Thirty-four shRNA sequences reproducibly protected RWPE-1 cells from anoikis after culture under suspension conditions including the top validated hit, α/ß hydrolase domain containing 4 (ABHD4). In validation studies, ABHD4 knockdown inhibited anoikis in RWPE-1 cells as well as anoikis sensitive NP69 nasopharyngeal and OVCAR3 ovarian cancer cells, while over-expression of the gene increased sensitivity. Induction of anoikis after ABHD4 knockdown was associated with cleavage of PARP and activation of caspases-3, but was independent in changes of FLIP, FAK and Src expression. Interestingly, induction of anoikis after ABHD4 knockdown was independent of the known role of ABHD4 in the anandamide synthesis pathway and the generation of glycerophospho-N-acyl ethanolamines. Thus, ABHD4 is a novel genetic regulator of anoikis sensitivity.


Asunto(s)
Anoicis/genética , Pruebas Genéticas , Genoma Humano/genética , Hidrolasas/metabolismo , ARN Interferente Pequeño/metabolismo , Ácidos Araquidónicos/biosíntesis , Ácidos Araquidónicos/química , Línea Celular Tumoral , Endocannabinoides/biosíntesis , Endocannabinoides/química , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Hidrolasas/genética , Lentivirus/genética , Lisofosfolipasa , Masculino , Alcamidas Poliinsaturadas/química , Estructura Terciaria de Proteína
3.
Blood ; 115(23): 4824-33, 2010 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-20348394

RESUMEN

On-patent and off-patent drugs with previously unrecognized anticancer activity could be rapidly repurposed for this new indication given their prior toxicity testing. To identify such compounds, we conducted chemical screens and identified the antihelmintic flubendazole. Flubendazole induced cell death in leukemia and myeloma cell lines and primary patient samples at nanomolar concentrations. Moreover, it delayed tumor growth in leukemia and myeloma xenografts without evidence of toxicity. Mechanistically, flubendazole inhibited tubulin polymerization by binding tubulin at a site distinct from vinblastine. In addition, cells resistant to vinblastine because of overexpression of P-glycoprotein remained fully sensitive to flubendazole, indicating that flubendazole can overcome some forms of vinblastine resistance. Given the different mechanisms of action, we evaluated the combination of flubendazole and vinblastine in vitro and in vivo. Flubendazole synergized with vinblastine to reduce the viability of OCI-AML2 cells. In addition, combinations of flubendazole with vinblastine or vincristine in a leukemia xenograft model delayed tumor growth more than either drug alone. Therefore, flubendazole is a novel microtubule inhibitor that displays preclinical activity in leukemia and myeloma.


Asunto(s)
Antinematodos/farmacología , Leucemia/tratamiento farmacológico , Mebendazol/análogos & derivados , Microtúbulos/metabolismo , Mieloma Múltiple/tratamiento farmacológico , Alcaloides de la Vinca/farmacología , Animales , Antinematodos/agonistas , Antinematodos/uso terapéutico , Antineoplásicos Fitogénicos/agonistas , Antineoplásicos Fitogénicos/farmacología , Antineoplásicos Fitogénicos/uso terapéutico , Muerte Celular , Supervivencia Celular , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Femenino , Células HeLa , Humanos , Leucemia/metabolismo , Masculino , Mebendazol/agonistas , Mebendazol/farmacología , Mebendazol/uso terapéutico , Ratones , Mieloma Múltiple/metabolismo , Células U937 , Vinblastina/agonistas , Vinblastina/farmacología , Vinblastina/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
4.
Oncogene ; 41(14): 2079-2094, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35181737

RESUMEN

The endocytic adaptor protein Numb acts as a tumor suppressor through downregulation of oncogenic pathways in multiple cancer types. The identification of splicing alterations giving rise to changes in Numb protein isoform expression indicate that Numb also has tumor promoting activity, though the underlying mechanisms are unknown. Here we report that NUMB exon 9 inclusion, which results in production of a protein isoform with an additional 49 amino acids, is a feature of multiple cancer types including all subtypes of breast cancer and correlates with worse progression-free survival. Specific deletion of exon 9-included Numb isoforms (Exon9in) from breast cancer cells reduced cell growth and prevents spontaneous lung metastasis in a mouse model. Quantitative proteome profiling showed that loss of Exon9in causes downregulation of membrane receptors and adhesion molecules, as well as proteins involved in extracellular matrix organization and the epithelial-mesenchymal transition (EMT) state. In addition, exon 9 deletion caused remodeling of the endocytic network, decreased ITGß5 surface localization, cell spreading on vitronectin and downstream signaling to ERK and SRC. Together these observations suggest that Exon9in isoform expression disrupts the endocytic trafficking functions of Numb, resulting in increased surface expression of ITGß5 as well as other plasma membrane proteins to promote cell adhesion, EMT, and tumor metastasis.


Asunto(s)
Neoplasias de la Mama , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Exones/genética , Femenino , Genes Supresores de Tumor , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Proteínas del Tejido Nervioso/metabolismo
5.
J Mol Biol ; 433(8): 166880, 2021 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-33617900

RESUMEN

CBL is a RING type E3 ubiquitin ligase that functions as a negative regulator of tyrosine kinase signaling and loss of CBL E3 function is implicated in several forms of leukemia. The Src-like adaptor proteins (SLAP/SLAP2) bind to CBL and are required for CBL-dependent downregulation of antigen receptor, cytokine receptor, and receptor tyrosine kinase signaling. Despite the established role of SLAP/SLAP2 in regulating CBL activity, the nature of the interaction and the mechanisms involved are not known. To understand the molecular basis of the interaction between SLAP/SLAP2 and CBL, we solved the crystal structure of CBL tyrosine kinase binding domain (TKBD) in complex with SLAP2. The carboxy-terminal region of SLAP2 adopts an α-helical structure which binds in a cleft between the 4H, EF-hand, and SH2 domains of the TKBD. This SLAP2 binding site is remote from the canonical TKBD phospho-tyrosine peptide binding site but overlaps with a region important for stabilizing CBL in its autoinhibited conformation. In addition, binding of SLAP2 to CBL in vitro activates the ubiquitin ligase function of autoinhibited CBL. Disruption of the CBL/SLAP2 interface through mutagenesis demonstrated a role for this protein-protein interaction in regulation of CBL E3 ligase activity in cells. Our results reveal that SLAP2 binding to a regulatory cleft of the TKBD provides an alternative mechanism for activation of CBL ubiquitin ligase function.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Proto-Oncogénicas c-cbl/química , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/química , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Ubiquitina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Sitios de Unión , Regulación hacia Abajo , Humanos , Conformación Molecular , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-cbl/genética , Proteínas Proto-Oncogénicas pp60(c-src)/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Alineación de Secuencia , Transducción de Señal , Ubiquitina-Proteína Ligasas/metabolismo , Dominios Homologos src
6.
Cancer Res ; 67(17): 8307-15, 2007 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-17804746

RESUMEN

Malignant epithelial cells with metastatic potential resist apoptosis that normally occurs upon loss of anchorage from the extracellular matrix, a process termed "anoikis." Resistance to anoikis enables malignant cells to survive in an anchorage-independent manner, which leads to the formation of distant metastases. To understand the regulation of anoikis, we designed, automated, and conducted a high-throughput chemical screen for anoikis sensitizers. PPC-1 anoikis-resistant prostate cancer cells were seeded in hydrogel-coated ultralow binding plates for suspension conditions and standard tissue culture plates to promote adhesion. After seeding, cells were treated with aliquots from a library of previously characterized small molecules, and viability was assessed using the 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt, assay. From this chemical screen, we identified anisomycin that induced apoptosis in suspension conditions, but was not toxic to these cells grown under adherent conditions. Anisomycin sensitized cells to anoikis by decreasing levels of the caspase-8 inhibitor FLIP and subsequently activating the death receptor pathway of caspase activation. Although anisomycin activated c-Jun-NH(2)-kinase and p38, these kinases were not functionally important for the effect of anisomycin on anoikis and FLIP. Rather, anisomycin decreased FLIP and sensitized cells to anoikis by inhibiting its protein synthesis. Finally, we showed that anisomycin decreased distal tumor formation in a mouse model of prostate cancer metastases. Thus, a novel chemical screen identified anisomycin as an anoikis sensitizer that acts by decreasing FLIP protein synthesis. Our results suggest that FLIP is a suppressor of anoikis and inhibiting FLIP protein synthesis may be a useful antimetastatic strategy.


Asunto(s)
Anisomicina/farmacología , Anisomicina/uso terapéutico , Anoicis/efectos de los fármacos , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/biosíntesis , Metástasis de la Neoplasia/prevención & control , Biosíntesis de Proteínas/efectos de los fármacos , Animales , Caspasas/metabolismo , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Masculino , Ratones , Modelos Biológicos , Células Neoplásicas Circulantes/efectos de los fármacos , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Receptores de Muerte Celular/metabolismo , Células Tumorales Cultivadas
7.
Mol Cancer Ther ; 7(11): 3546-55, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19001437

RESUMEN

Evasion of death receptor ligand-induced apoptosis is an important contributor to cancer development and progression. Therefore, molecules that restore sensitivity to death receptor stimuli would be important tools to better understand this biological pathway and potential leads for therapeutic adjuncts. Previously, the small-molecule N-[4-chloro-3-(trifluoromethyl)phenyl]-3-oxobutanamide (fasentin) was identified as a chemical sensitizer to the death receptor stimuli FAS and tumor necrosis factor apoptosis-inducing ligand, but its mechanism of action was unknown. Here, we determined that fasentin alters expression of genes associated with nutrient and glucose deprivation. Consistent with this finding, culturing cells in low-glucose medium recapitulated the effects of fasentin and sensitized cells to FAS. Moreover, we showed that fasentin inhibited glucose uptake. Using virtual docking studies with a homology model of the glucose transport protein GLUT1, fasentin interacted with a unique site in the intracellular channel of this protein. Additional chemical studies with other GLUT inhibitors and analogues of fasentin supported a role for partial inhibition of glucose transport as a mechanism to sensitize cells to death receptor stimuli. Thus, fasentin is a novel inhibitor of glucose transport that blocks glucose uptake and highlights a new mechanism to sensitize cells to death ligands.


Asunto(s)
Anilidas/farmacología , Antineoplásicos/farmacología , Apoptosis , Glucosa/metabolismo , Receptor fas/metabolismo , Anilidas/síntesis química , Antineoplásicos/síntesis química , Antineoplásicos/química , Sitios de Unión , Transporte Biológico/efectos de los fármacos , Ciclo Celular , Línea Celular Tumoral , Proteína Ligando Fas/metabolismo , Perfilación de la Expresión Génica , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Transportador de Glucosa de Tipo 1/antagonistas & inhibidores , Transportador de Glucosa de Tipo 1/metabolismo , Humanos , Masculino , Receptores de Muerte Celular/antagonistas & inhibidores , Receptores de Muerte Celular/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo
8.
Sci Rep ; 9(1): 10865, 2019 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-31350469

RESUMEN

Small molecule inhibitors of BRAF and MEK have proven effective at inhibiting tumor growth in melanoma patients, however this efficacy is limited due to the almost universal development of drug resistance. To provide advanced insight into the signaling responses that occur following kinase inhibition we have performed quantitative (phospho)-proteomics of human melanoma cells treated with either dabrafenib, a BRAF inhibitor; trametinib, a MEK inhibitor or SCH772984, an ERK inhibitor. Over nine experiments we identified 7827 class I phosphorylation sites on 4960 proteins. This included 54 phosphorylation sites that were significantly down-modulated after exposure to all three inhibitors, 34 of which have not been previously reported. Functional analysis of these novel ERK targets identified roles for them in GTPase activity and regulation, apoptosis and cell-cell adhesion. Comparison of the results presented here with previously reported phosphorylation sites downstream of ERK showed a limited degree of overlap suggesting that ERK signaling responses may be highly cell line and cue specific. In addition we identified 26 phosphorylation sites that were only responsive to dabrafenib. We provide further orthogonal experimental evidence for 3 of these sites in human embryonic kidney cells over-expressing BRAF as well as further computational insights using KinomeXplorer. The validated phosphorylation sites were found to be involved in actin regulation, which has been proposed as a novel mechanism for inhibiting resistance development. These results would suggest that the linearity of the BRAF-MEK-ERK module is at least context dependent.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Imidazoles/farmacología , Indazoles/farmacología , Melanoma/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Oximas/farmacología , Piperazinas/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Piridonas/farmacología , Pirimidinonas/farmacología , Neoplasias Cutáneas/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Melanoma/patología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Fosforilación/efectos de los fármacos , Proteoma , Proteómica/métodos , Proteínas Proto-Oncogénicas B-raf/metabolismo , Neoplasias Cutáneas/patología
9.
Elife ; 82019 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-31124786

RESUMEN

Human NimA-related kinases (Neks) have multiple mitotic and non-mitotic functions, but few substrates are known. We systematically determined the phosphorylation-site motifs for the entire Nek kinase family, except for Nek11. While all Nek kinases strongly select for hydrophobic residues in the -3 position, the family separates into four distinct groups based on specificity for a serine versus threonine phospho-acceptor, and preference for basic or acidic residues in other positions. Unlike Nek1-Nek9, Nek10 is a dual-specificity kinase that efficiently phosphorylates itself and peptide substrates on serine and tyrosine, and its activity is enhanced by tyrosine auto-phosphorylation. Nek10 dual-specificity depends on residues in the HRD+2 and APE-4 positions that are uncommon in either serine/threonine or tyrosine kinases. Finally, we show that the phosphorylation-site motifs for the mitotic kinases Nek6, Nek7 and Nek9 are essentially identical to that of their upstream activator Plk1, suggesting that Nek6/7/9 function as phospho-motif amplifiers of Plk1 signaling.


Asunto(s)
Quinasas Relacionadas con NIMA/metabolismo , Transducción de Señal , Especificidad por Sustrato , Humanos , Quinasas Relacionadas con NIMA/química , Fosforilación , Serina/metabolismo , Treonina/metabolismo
10.
J Med Chem ; 48(7): 2266-9, 2005 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-15801818

RESUMEN

A new class of selective FXIIIa inhibitors with a bicyclic [1,2,4]-thiadiazole pharmacophore is described. At 160 muM, compound 8 caused 50% reduction in fibrin gamma-chain cross-linking and suppressed the polymerization of alpha chains in platelet-depleted human plasma clots. Fibrinolysis rates in response to tissue plasminogen activator were directly proportional to the concentration of 8 in plasma at the time of clotting.


Asunto(s)
Factor XIII/antagonistas & inhibidores , Fibrinolíticos/síntesis química , Imidazoles/síntesis química , Tiadiazoles/síntesis química , Animales , Factor XIII/química , Fibrinolíticos/química , Fibrinolíticos/farmacología , Cobayas , Humanos , Imidazoles/química , Imidazoles/farmacología , Técnicas In Vitro , Cinética , Relación Estructura-Actividad , Tiadiazoles/química , Tiadiazoles/farmacología
11.
PLoS One ; 7(7): e41102, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22829912

RESUMEN

Tumor necrosis factor-related apoptosis inducing ligand (TRAIL) induces apoptosis in a variety of cancer cell lines with little or no effect on normal cells. However, its effect is limited as some cancers including pancreatic cancer show de novo resistance to TRAIL induced apoptosis. In this study we report that GSK-3 inhibition using the pharmacologic agent AR-18, enhanced TRAIL sensitivity in a range of pancreatic and prostate cancer cell lines. This sensitization was found to be caspase-dependent, and both pharmacological and genetic knock-down of GSK-3 isoforms resulted in apoptotic features as shown by cleavage of PARP and caspase-3. Elevated levels of reactive oxygen intermediates and disturbance of mitochondrial membrane potential point to a mitochondrial amplification loop for TRAIL-induced apoptosis after GSK-3 inhibition. Consistent with this, overexpression of anti-apoptotic mitochondrial targets such as Bcl-XL, Mcl-1, and Bcl-2 rescued PANC-1 and PPC-1 cells from TRAIL sensitization. However, overexpression of the caspase-8 inhibitor CrmA also inhibited the sensitizing effects of GSK-3 inhibitor, suggesting an additional role for GSK-3 that inhibits death receptor signaling. Acute treatment of mice bearing PANC-1 xenografts with a combination of AR-18 and TRAIL also resulted in a significant increase in apoptosis, as measured by caspase-3 cleavage. Sensitization to TRAIL occurred despite an increase in ß-catenin due to GSK-3 inhibition, suggesting that the approach might be effective even in cancers with dysregulated ß-catenin. These results suggest that GSK-3 inhibitors might be effectively combined with TRAIL for the treatment of pancreatic cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/metabolismo , Neoplasias Pancreáticas/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Apoptosis/genética , Línea Celular Tumoral , Inhibidores Enzimáticos/farmacología , Humanos , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
12.
PLoS One ; 7(9): e43721, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22957033

RESUMEN

Oncogenic signaling promotes tumor invasion and metastasis, in part, by increasing the expression of tri- and tetra- branched N-glycans. The branched N-glycans bind to galectins forming a multivalent lattice that enhances cell surface residency of growth factor receptors, and focal adhesion turnover. N-acetylglucosaminyltransferase I (MGAT1), the first branching enzyme in the pathway, is required for the addition of all subsequent branches. Here we have introduced MGAT1 shRNA into human HeLa cervical and PC-3-Yellow prostate tumor cells lines, generating cell lines with reduced transcript, enzyme activity and branched N-glycans at the cell surface. MGAT1 knockdown inhibited HeLa cell migration and invasion, but did not alter cell proliferation rates. Swainsonine, an inhibitor of α-mannosidase II immediately downstream of MGAT1, also inhibited cell invasion and was not additive with MGAT1 shRNA, consistent with a common mechanism of action. Focal adhesion and microfilament organization in MGAT1 knockdown cells also indicate a less motile phenotype. In vivo, MGAT1 knockdown in the PC-3-Yellow orthotopic prostate cancer xenograft model significantly decreased primary tumor growth and the incidence of lung metastases. Our results demonstrate that blocking MGAT1 is a potential target for anti-cancer therapy.


Asunto(s)
N-Acetilglucosaminiltransferasas/genética , Neoplasias/genética , Neoplasias/terapia , ARN Interferente Pequeño/metabolismo , Animales , Línea Celular Tumoral , Membrana Celular/metabolismo , Progresión de la Enfermedad , Inhibidores Enzimáticos/farmacología , Femenino , Células HeLa , Humanos , Masculino , Ratones , N-Acetilglucosaminiltransferasas/metabolismo , Metástasis de la Neoplasia , Trasplante de Neoplasias , Fenotipo , Polisacáridos/química , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/terapia , Transducción de Señal , Swainsonina/farmacología
13.
Mol Cancer Ther ; 9(1): 246-56, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20053768

RESUMEN

Evasion of death receptor ligand-induced apoptosis represents an important contributor to cancer development and progression. Therefore, molecules that restore sensitivity to death receptor stimuli would be important tools to better understand this biological pathway and potential leads for therapeutic adjuncts. Previously, the small-molecule 4-(4-chloro-2-methylphenoxy)-N-hydroxybutanamide (that we propose be named droxinostat) was identified as a chemical sensitizer to death receptor stimuli, decreasing the expression of the caspase-8 inhibitor FLIP. However, the direct targets of droxinostat were unknown. To better understand the mechanism of action of droxinostat and highlight new strategies to restore sensitivity to death receptor ligands, we analyzed changes in gene expression using the Connectivity Map after treating cells with droxinostat. Changes in gene expression after droxinostat treatment resembled changes observed after treatment with histone deacetylase (HDAC) inhibitors. Therefore, we examined the effects of droxinostat on HDAC activity and showed that it selectively inhibited HDAC3, HDAC6, and HDAC8 and that inhibition of these HDACs was functionally important for its ability to sensitize cells to death ligands. Thus, we have identified a selective HDAC inhibitor and showed that selective HDAC inhibition sensitizes cells to death ligands, thereby highlighting a new mechanism to overcome resistance to death receptor ligands.


Asunto(s)
Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Neoplasias/enzimología , Neoplasias/patología , Receptores de Muerte Celular/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Técnicas de Silenciamiento del Gen , Inhibidores de Histona Desacetilasas/química , Humanos , Ácidos Hidroxámicos/farmacología , Ligandos , Modelos Moleculares , Receptor fas/metabolismo
14.
Cancer Res ; 69(7): 2739-47, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19293189

RESUMEN

Normal epithelial cells undergo apoptosis upon detachment from the extracellular matrix, a process termed "anoikis." However, malignant epithelial cells with metastatic potential resist anoikis and can survive in an anchorage-independent fashion. Molecules that sensitize resistant cells to anoikis will be useful chemical probes to understand this pathway. To identify novel anoikis sensitizers in anoikis-resistant PPC-1 prostate adenocarcinoma cells, a library of 2,000 off-patent drugs and natural products was screened for their ability to preferentially induce cell death in suspension over adherent culture conditions. This screen identified five members of the family of cardiac glycosides as anoikis sensitizers, including ouabain, peruvoside, digoxin, digitoxin, and strophanthidin. We conducted further studies with ouabain to discern the mechanism of cardiac glycoside-induced anoikis sensitization. Ouabain initiated anoikis through the mitochondrial pathway of caspase activation. In addition, ouabain sensitized cells to anoikis by inhibiting its known target, the Na(+)/K(+) ATPase pump, and inducing hypoosmotic stress. Resistance to anoikis permits cancer cells to survive in the circulation and facilitates their metastasis to distant organs, so we tested the effects of Na(+)/K(+) ATPase inhibition on distant tumor formation in mouse models. In these mouse models, ouabain inhibited tumor metastases but did not alter the growth of subcutaneous tumors. Thus, we have identified a novel mechanism to sensitize resistant cells to anoikis and decrease tumor metastasis. These results suggest a potential mechanism for the observed clinical reduction in metastasis and relapse in breast cancer patients who have undergone treatments with cardiac glycosides.


Asunto(s)
Anoicis/efectos de los fármacos , Glicósidos Cardíacos/farmacología , Inhibidores Enzimáticos/farmacología , Neoplasias/tratamiento farmacológico , ATPasa Intercambiadora de Sodio-Potasio/antagonistas & inhibidores , Animales , Anoicis/fisiología , Inhibidores de Caspasas , Caspasas/metabolismo , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Activación Enzimática/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Metástasis de la Neoplasia , Neoplasias/enzimología , Neoplasias/patología , Presión Osmótica/efectos de los fármacos , Ouabaína/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Cancer Lett ; 272(2): 177-85, 2008 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-18579285

RESUMEN

As a barrier to metastases, cells normally undergo apoptosis after they lose contact with their extra cellular matrix or their neighbouring cells. This cell death process has been termed "anoikis". Tumour cells that acquire malignant potential have developed mechanisms to resist anoikis and thereby survive after detachment from their primary site and while travelling through the lymphatic and circulatory systems. Defects in the death receptor pathway of caspase activation, such as the over-expression of the caspase-8 inhibitor FLIP, can render cells resistant to anoikis. Likewise, roadblocks in the mitochondrial pathway, such as over-expression of the Bcl-2 family of anti-apoptotic proteins, can also confer resistance to anoikis. This review will focus on the roles of the death receptor and mitochondrial pathways in anoikis and anoikis resistance and how targeting defects in these pathways can restore sensitivity to anoikis and serve as the basis for therapeutic adjuncts that prevent metastasis.


Asunto(s)
Anoicis , Metástasis de la Neoplasia , Caspasas/metabolismo , Activación Enzimática , Humanos , Mitocondrias/enzimología , Mitocondrias/metabolismo , Receptores de Muerte Celular/metabolismo
16.
Cancer Res ; 68(16): 6688-97, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18701493

RESUMEN

Aberrant N-linked glycans promote the malignant potential of cells by enhancing the epithelial-to-mesenchymal transition and the invasive phenotype. To identify small molecule inhibitors of N-glycan biosynthesis, we developed a chemical screen based on the ability of the tetravalent plant lectin L-phytohemagglutinin (L-PHA) to bind and crosslink surface glycoproteins with beta1,6GlcNAc-branched complex type N-glycans and thereby induce agglutination and cell death. In this screen, Jurkat cells were treated with a library of off-patent chemicals (n = 1,280) to identify molecules that blocked L-PHA-induced death. The most potent hit from this screen was the cardiac glycoside (CG) dihydroouabain. In secondary assays, a panel of CGs was tested for their effects on L-PHA-induced agglutination and cell death. All of the CGs tested inhibited L-PHA-induced death in Jurkat cells, and the most potent CG tested was digoxin with an EC(50) of 60 +/- 20 nmol/L. Digoxin also increased the fraction of some concanavalin A-binding N-glycans. Using matrix-assisted laser desorption/ionization time-of-flight mass spectrometry, digoxin specifically increased GlcNAc(1)Man(3)GlcNAc(2)Fuc(1) and GlcNAc(2)Man(3)GlcNAc(2)Fuc(1) oligosaccharides demonstrating an impairment of the N-glycan pathway. Consistent with this effect on the N-glycan pathway, digoxin inhibited N-glycosylation-mediated processes of tumor cell migration and invasion. Furthermore, digoxin prevented distant tumor formation in two mouse models of metastatic prostate cancer. Thus, taken together, our high throughput screen identified CGs as modifiers of the N-glycan pathway. These molecules can be used as tools to better understand the role of N-glycans in normal and malignant cells. Moreover, these results may partly explain the anticancer effect of CGs in cardiovascular patients.


Asunto(s)
Glicósidos Digitálicos/farmacología , Inhibidores Enzimáticos/farmacología , Neoplasias/tratamiento farmacológico , Polisacáridos/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/antagonistas & inhibidores , Animales , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Técnicas Químicas Combinatorias , Glicopéptidos/metabolismo , Glicosilación , Humanos , Masculino , Ratones , Ratones SCID , Neoplasias/enzimología , Neoplasias/patología , Fitohemaglutininas/farmacología , ARN Interferente Pequeño/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Transfección , Células Tumorales Cultivadas , Cicatrización de Heridas
17.
J Natl Cancer Inst ; 99(10): 811-22, 2007 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-17505076

RESUMEN

BACKGROUND: Normal epithelial cells undergo anoikis, or apoptosis on loss of anchorage to the extracellular matrix, by initiating the death receptor pathway of caspase activation. However, malignant epithelial cells with metastatic potential resist anoikis and can survive in an anchorage-independent fashion. We hypothesized that c-Fas-associated death domain-like interleukin-1-converting enzyme-like inhibitory protein (FLIP), an endogenous inhibitor of death receptor signaling, may suppress anoikis. METHODS: We assessed viability and apoptosis of PPC-1 prostate cancer cells cultured in adherent and suspension conditions using 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium inner salt and Annexin V staining assays. Expression of the death receptor Fas and activation of caspase 8 were measured using flow cytometry. Expression of Fas ligand was measured by reverse transcription-polymerase chain reaction. FLIP protein expression was measured by immunoblotting. Small-molecule inhibitors of FLIP (including the death receptor sensitizer 5809354) and small-interfering (si) RNA directed against FLIP were used to assess the effects of FLIP inhibition on anoikis of prostate cancer cells in vitro and in vivo. All statistical tests were two-sided. RESULTS: PPC-1 cells cultured in suspension resisted anoikis, despite increased expression of Fas (0 versus 8 hours, mean relative percent expression = 100% versus 135%, difference = 35%, 95% confidence interval [CI] = 10% to 61%; P = .02) and Fas L (0 versus 24 hours, mean relative percent expression = 100% versus 208%, difference = 108%, 95% CI = 18% to 197%; P = .02). Knockdown of FLIP expression by siRNA or treatment with 5809354 sensitized prostate cancer cells to anoikis (control siRNA versus FLIP siRNA at 10 nM, mean relative percent viability = 95% versus 51%, difference = 44%, 95% CI = 34% to 54%; P<.001; control versus 5809354 at 20 microM, mean relative percent viability = 96% versus 52%, difference = 44%, 95% CI = 13% to 75%; P = .015). Inhibition of FLIP expression specifically activated caspase 8 in PPC-1 cells grown in suspension but not adherent conditions and decreased the metastatic potential of circulating PPC-1 cells in vivo. CONCLUSIONS: FLIP may be a suppressor of anoikis and therefore a possible target for antimetastatic therapeutic strategies.


Asunto(s)
Anoicis/fisiología , Metástasis de la Neoplasia , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Animales , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Caspasa 8/biosíntesis , Línea Celular Tumoral , Citometría de Flujo , Humanos , Procesamiento de Imagen Asistido por Computador , Imagenología Tridimensional , Immunoblotting , Masculino , Ratones , Ratones SCID , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Receptor fas/biosíntesis
18.
J Org Chem ; 70(16): 6230-41, 2005 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-16050682

RESUMEN

A versatile synthetic route to potentially useful fused-ring [1,2,4]thiadiazole scaffolds (e.g., 7a and 10b) via exchange reactions of the precursor [1,2,4]thiadiazol-3-(2H)one derivatives (e.g., 6 and 9) with appropriately substituted nitriles (e.g., cyanogen bromide or p-toluenesulfonyl cyanide) under mild conditions is described. For example, the tricyclic 3-bromo [1,2,4]THD derivative (7a) underwent S(N)Ar substitution with a variety of nucleophiles, which included amines, malonate esters and alcohols. Likewise, the bicyclic 3-p-tosyl [1,2,4]THD (10b) was employed as a template in reaction with diamines, and the resulting substituted diamines (e.g., 12a or 12e) were further selectively derivatized at the N1 and/or N2 positions in a linear fashion. The X-ray crystal structure of the 3-methyl bicyclic [1,2,4]THD (21) was obtained, and selective methylation at the N1 position via a protection-alkylation-deprotection protocol, as illustrated in Scheme 6, was confirmed. Alternatively, a short convergent synthesis of N1-functionalized derivatives from the reaction of 10b with appropriately substituted secondary amines was also developed. Hence, these synthetic strategies were advantageously exploited to provide access to a variety of diversely derivatized 3-substituted fused-ring [1,2,4]thiadiazole derivatives.


Asunto(s)
Benceno/química , Imidazoles/química , Tiadiazoles/química , Aminas/química , Cristalografía por Rayos X , Ciclización , Cisteína/química , Estructura Molecular , Tiadiazoles/síntesis química , Tiadiazoles/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA