Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Pharmacol Rev ; 65(4): 1162-97, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23943849

RESUMEN

Autophagy, a process of self-digestion of the cytoplasm and organelles through which cellular components are recycled for reuse or energy production, is an evolutionarily conserved response to metabolic stress found in eukaryotes from yeast to mammals. It is noteworthy that autophagy is also associated with various pathophysiologic conditions in which this cellular process plays either a cytoprotective or cytopathic role in response to a variety of stresses such as metabolic, inflammatory, neurodegenerative, and therapeutic stress. It is now generally believed that modulating the activity of autophagy through targeting specific regulatory molecules in the autophagy machinery may impact disease processes, thus autophagy may represent a new pharmacologic target for drug development and therapeutic intervention of various human disorders. Induction or inhibition of autophagy using small molecule compounds has shown promise in the treatment of diseases such as cancer. Depending on context, induction or suppression of autophagy may exert therapeutic effects via promoting either cell survival or death, two major events targeted by therapies for various disorders. A better understanding of the biology of autophagy and the pharmacology of autophagy modulators has the potential for facilitating the development of autophagy-based therapeutic interventions for several human diseases.


Asunto(s)
Autofagia/fisiología , Animales , Autofagia/efectos de los fármacos , Enfermedad , Quimioterapia , Humanos
2.
Biochem Biophys Res Commun ; 424(2): 308-14, 2012 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-22749997

RESUMEN

Eukaryotic elongation factor-2 kinase (eEF-2K) is a Ca(2+)/calmodulin-dependent enzyme that negatively regulates protein synthesis. eEF-2K has been shown to be up-regulated in cancer, and to play an important role in cell survival through inhibition of protein synthesis. Post-translational modification of protein synthesis machinery is important for its regulation and could be critical for survival of cancer cells encountering stress. The purpose of our study was to examine the regulation of eEF-2K during stress with a focus on the roles of phosphorylation in determining the stability of eEF-2K. We found that stress conditions (nutrient deprivation and hypoxia) increase eEF-2K protein. mRNA levels are only transiently increased and shortly return to normal, while eEF-2K protein levels continue to increase after further exposure to stress. A seemingly paradoxical decrease in eEF-2K stability was found when glioma cells were subjected to stress despite increased protein expression. We further demonstrated that phosphorylation of eEF-2K differentially affects the enzyme's turnover under both normal and stress conditions, as evidenced by the different half-lives of phosphorylation-defective mutants of eEF-2K. We further found that the eEF-2K site (Ser398) phosphorylated by AMPK is pivotal to the protein's stability, as the half-life of S398A mutant increases to greater than 24h under both normal and stress conditions. These data indicate that eEF-2K is regulated at multiple levels with phosphorylation playing a critical role in the enzyme's turnover under stressful conditions. The complexity of eEF-2K phosphorylation highlights the intricacies of protein synthesis control during cellular stress.


Asunto(s)
Quinasa del Factor 2 de Elongación/metabolismo , Procesamiento Proteico-Postraduccional , Estrés Fisiológico , Línea Celular Tumoral , Quinasa del Factor 2 de Elongación/genética , Estabilidad de Enzimas , Humanos , Mutación , Fosforilación , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores
3.
Cancer Res ; 67(12): 5831-9, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17575151

RESUMEN

A single nucleotide polymorphism (SNP) SNP309 (T-->G) in the murine double minute 2 (MDM2) promoter creates a high-affinity Sp1 binding site and increases the expression of MDM2 mRNA and protein. Approximately 40% of the populations harbor at least one variant allele and 12% to 17% are homozygous G/G at codon 309. This MDM2 SNP increases susceptibility to cancer and decreases the response of cancer cells to certain forms of treatment, such as radiation therapy and DNA-damaging drugs. Topoisomerase II (TopoII)-targeting agents are commonly used chemotherapeutic drugs with a broad spectrum of activity. However, resistance to TopoII poisons limits their effectiveness. We show that MDM2 SNP309 rendered a panel of cancer cell lines that are homozygous for SNP309 selectively resistant (approximately 10-fold) to certain TopoII-targeting chemotherapeutic drugs (etoposide, mitoxantrone, amsacrine, and ellipticine). The mechanism underlying this observation was Mdm2-mediated down-regulation of TopoII; on drug exposure, MDM2 bound to TopoII and resulted in decreased cellular enzyme content. Knockdown of MDM2 by RNA interference stabilized TopoIIalpha and decreased resistance to TopoII-targeting drugs. Thus, MDM2 SNP309 (T-->G) may represent a relatively common, previously unappreciated determinant of drug sensitivity. Given the frequency of SNP309 in the general population (40% in heterozygous T/G and 12% in homozygous G/G condition), our observation may have important implications for the individualization of cancer chemotherapy.


Asunto(s)
Antineoplásicos/farmacología , ADN-Topoisomerasas de Tipo II/metabolismo , Resistencia a Antineoplásicos/genética , Polimorfismo de Nucleótido Simple , Proteínas Proto-Oncogénicas c-mdm2/genética , Amsacrina/farmacología , Animales , Línea Celular Tumoral , ADN-Topoisomerasas de Tipo II/efectos de los fármacos , Elipticinas/farmacología , Etopósido/farmacología , Humanos , Immunoblotting , Ratones , Microscopía Confocal , Mitoxantrona/farmacología , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-mdm2/efectos de los fármacos , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
4.
Cancer Res ; 66(6): 3015-23, 2006 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-16540650

RESUMEN

Elongation factor-2 kinase (eEF-2 kinase), also known as Ca(2+)/calmodulin-dependent kinase III, regulates protein synthesis by controlling the rate of peptide chain elongation. The activity of eEF-2 kinase is increased in glioblastoma and other malignancies, yet its role in neoplasia is uncertain. Recent evidence suggests that autophagy plays an important role in oncogenesis and that this can be regulated by mammalian target of rapamycin (mTOR). Because eEF-2 kinase lies downstream of mTOR, we studied the role of eEF-2 kinase in autophagy using human glioblastoma cell lines. Knockdown of eEF-2 kinase by RNA interference inhibited autophagy in glioblastoma cell lines, as measured by light chain 3 (LC3)-II formation, acidic vesicular organelle staining, and electron microscopy. In contrast, overexpression of eEF-2 kinase increased autophagy. Furthermore, inhibition of autophagy markedly decreased the viability of glioblastoma cells grown under conditions of nutrient depletion. Nutrient deprivation increased eEF-2 kinase activity and decreased the activity of S6 kinase, suggesting an involvement of mTOR pathway in the eEF-2 kinase regulation of autophagy. These results suggest that eEF-2 kinase plays a regulatory role in the autophagic process in tumor cells; and eEF-2 kinase is a downstream member of the mTOR signaling; eEF-2 kinase may promote cancer cell survival under conditions of nutrient deprivation through regulating autophagy. Therefore, eEF-2 kinase may be a part of a survival mechanism in glioblastoma and targeting this kinase may represent a novel approach to cancer treatment.


Asunto(s)
Autofagia/fisiología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/fisiología , Glioblastoma/enzimología , Glioblastoma/patología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/antagonistas & inhibidores , Proteínas Quinasas Dependientes de Calcio-Calmodulina/biosíntesis , Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Línea Celular Tumoral , Quinasa del Factor 2 de Elongación , Glioblastoma/genética , Humanos , ARN Interferente Pequeño/genética , Transfección
5.
Cancer Prev Res (Phila) ; 11(12): 735-778, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30530635

RESUMEN

The recent pace, extent, and impact of paradigm-changing cancer prevention science has been remarkable. The American Association for Cancer Research (AACR) convened a 3-day summit, aligned with five research priorities: (i) Precancer Atlas (PCA). (ii) Cancer interception. (iii) Obesity-cancer linkage, a global epidemic of chronic low-grade inflammation. (iv) Implementation science. (v) Cancer disparities. Aligned with these priorities, AACR co-led the Lancet Commission to formally endorse and accelerate the NCI Cancer Moonshot program, facilitating new global collaborative efforts in cancer control. The expanding scope of creative impact is perhaps most startling-from NCI-funded built environments to AACR Team Science Awarded studies of Asian cancer genomes informing global primary prevention policies; cell-free epigenetic marks identifying incipient neoplastic site; practice-changing genomic subclasses in myeloproliferative neoplasia (including germline variant tightly linked to JAK2 V617F haplotype); universal germline genetic testing for pancreatic cancer; and repurposing drugs targeting immune- and stem-cell signals (e.g., IL-1ß, PD-1, RANK-L) to cancer interception. Microbiota-driven IL-17 can induce stemness and transformation in pancreatic precursors (identifying another repurposing opportunity). Notable progress also includes hosting an obesity special conference (connecting epidemiologic and molecular perspectives to inform cancer research and prevention strategies), co-leading concerted national implementation efforts in HPV vaccination, and charting the future elimination of cancer disparities by integrating new science tools, discoveries and perspectives into community-engaged research, including targeted counter attacks on e-cigarette ad exploitation of children, Hispanics and Blacks. Following this summit, two unprecedented funding initiatives were catalyzed to drive cancer prevention research: the NCI Cancer Moonshot (e.g., PCA and disparities); and the AACR-Stand Up To Cancer bold "Cancer Interception" initiative.


Asunto(s)
Investigación Biomédica/tendencias , Neoplasias/prevención & control , Obesidad/epidemiología , Prevención Primaria/organización & administración , Investigación Biomédica/organización & administración , Congresos como Asunto , Implementación de Plan de Salud , Disparidades en el Estado de Salud , Humanos , Neoplasias/etnología , Neoplasias/etiología , Obesidad/complicaciones , Prevención Primaria/métodos , Prevención Primaria/tendencias , Salud Pública/estadística & datos numéricos , Salud Pública/tendencias , Sociedades Médicas/organización & administración , Sociedades Médicas/tendencias , Sociedades Científicas/organización & administración , Sociedades Científicas/tendencias , Estados Unidos/epidemiología
6.
Biochem Pharmacol ; 74(6): 851-9, 2007 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-17662696

RESUMEN

Drug resistance caused by overexpression of P-glycoprotein (P-gp), the MDR1 (ABCB1) gene product, limits the therapeutic outcome. Expression of MDR1 can be induced by divergent stimuli, and involves a number of transcriptional factors. We found that the expression of CtBP1 (C-terminal-binding protein 1), a transcriptional co-regulator, was increased (approximately 4-fold) in human multidrug resistant (MDR) cancer cell lines, NCI/ADR-RES and A2780/DX, as compared to their sensitive counterparts. Silencing of CtBP1 expression by RNAi decreased the MDR1 mRNA and P-gp. Knockdown of CtBP1 also enhanced the sensitivity of MDR cells to chemotherapeutic drugs that are transported by P-gp and increased intracellular drug accumulation. In a reporter gene assay, co-transfection of MDR1 promoter constructs with a CtBP1 expression vector resulted in a approximately 2-4-fold induction of MDR1 promoter activity. CtBP1 appeared to contribute to the activation of MDR1 transcription through directly interacting with the MDR1 promoter, as evidenced by its physical binding to the promoter region of the MDR1 gene in chromatin immunoprecipitation and electromobility shift assays. Histone modifications at the MDR1 promoter, such as mono-methylation, di-methylation, and acetylation of histone H3, were not found to be affected by silencing of CtBP1 expression. Our results reveal a novel role for CtBP1 as an activator of MDR1 gene transcription, and suggest that CtBP1 might be one of the key transcription factors involved in the induction of MDR1 gene. Therefore, CtBP1 may represent a potentially new target for inhibiting drug resistance mediated by overexpression of the MDR1 gene.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Oxidorreductasas de Alcohol/genética , Proteínas de Unión al ADN/genética , Resistencia a Múltiples Medicamentos/genética , Regulación Neoplásica de la Expresión Génica/genética , Genes MDR/genética , Oxidorreductasas de Alcohol/fisiología , Proteínas de Unión al ADN/fisiología , Resistencia a Antineoplásicos/genética , Histonas , Humanos , Regiones Promotoras Genéticas , Activación Transcripcional
7.
Cancer Res ; 65(9): 3806-10, 2005 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15867377

RESUMEN

Eukaryotic elongation factor-2 kinase (eEF-2 kinase) is a highly conserved calcium/calmodulin-dependent enzyme involved in the regulation of protein translation and cell proliferation. Rapid changes in the activity and abundance of eEF-2 kinase have been observed on growth stimulation, and increased enzyme activity is characteristic of malignant cell growth. Yet the mechanism for controlling the turnover of this kinase is unknown. The ubiquitin-proteasome pathway regulates the degradation of many cellular proteins, including transcription factors, cell cycle regulators, and signal transduction proteins. Therefore, we determined whether the ubiquitin-proteasome pathway regulates the turnover of eEF-2 kinase. We found that eEF-2 kinase was a relatively short-lived protein with a half-life of less than 6 hours. eEF-2 kinase was ubiquitinated in vivo as determined by coimmunoprecipitation and polyubiquitin affinity matrix. Incubation of purified eEF-2 kinase with a source of ubiquitination enzymes (rabbit reticulocyte lysate), purified ubiquitin, and ATP revealed the presence of increasing molecular weight species of ubiquitinated eEF-2 kinase. Treatment of cells with MG132, a proteasome inhibitor, inhibited eEF-2 kinase degradation and induced the accumulation of polyubiquitinated forms of the enzyme, resulting in an increase in its half-life. These results suggest involvement of the proteasome in the turnover of the ubiquitinated kinase. Because eEF-2 kinase is chaperoned by heat shock protein 90 (Hsp90), we next determined if disruption of the Hsp90-eEF-2 kinase complex promoted degradation of the kinase. Treatment of cells with geldanamycin, an Hsp90 inhibitor, enhanced ubiquitination of eEF-2 kinase and decreased the half-life of the kinase to less than 2 hours. These results indicate that cellular levels of eEF-2 kinase are maintained by a balance between association with Hsp90 and degradation by the ubiquitin-proteasome pathway. In conclusion, these data show that the turnover of eEF-2 kinase is regulated by the ubiquitin-proteasome pathway and, therefore, modulating the ubiquitination of eEF-2 kinase might control the abundance of this enzyme and have implications in the treatment of certain forms of cancer.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Línea Celular Tumoral , Quinasa del Factor 2 de Elongación , Estabilidad de Enzimas , Femenino , Glioblastoma/enzimología , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Humanos , Neoplasias Ováricas/enzimología
8.
Cancer Res ; 65(15): 6660-7, 2005 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-16061646

RESUMEN

Invasion and metastases of cancer cells and the development of resistance to anticancer therapies are the main causes of morbidity and mortality from cancer. For more than two decades, these two important but not clearly related aspects in the biology of cancer have been extensively studied. Specifically, P-glycoprotein and CD44 have been characterized and are known to be determinants of multidrug resistance (MDR) and metastases. Despite this body of knowledge, few reports have linked the two phenotypes and only recently have there been reasons to suspect a direct connection. In this report, we show that a novel physical and genetic interaction between CD44s and P-glycoprotein is in part responsible for the correlation between MDR and invasive potential in cancer cells. P-glycoprotein-specific substrates that interfere with its function reduced in vitro invasion, migration, and the physical colocalization of CD44s and P-glycoprotein. CD44 expression in sensitive cells promoted the expression of P-glycoprotein and the MDR phenotype. RNA interference of MDR1 inhibited the rate of cell migration. These data indicate that there is a close interaction between CD44 and P-glycoprotein that results in the concurrent expression and modulation of two malignant phenotypes, invasion and MDR.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Movimiento Celular/fisiología , Receptores de Hialuranos/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Línea Celular Tumoral , Membrana Celular/metabolismo , Movimiento Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Femenino , Flupentixol/farmacología , Humanos , Receptores de Hialuranos/biosíntesis , Receptores de Hialuranos/genética , Inmunoprecipitación , Invasividad Neoplásica , Neoplasias/tratamiento farmacológico , Interferencia de ARN , Transfección
9.
Clin Cancer Res ; 11(12): 4275-7, 2005 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15958606

RESUMEN

Translational research is difficult to define but recognizable to all who engage in it. Academic medical centers struggle to participate effectively, in contrast to the biotechnology or pharmaceutical industry, which are designed for nothing else. The process of translational research can be viewed as a cycle with defined phases and identifiable checkpoints. From the original hypothesis, through early scientific testing, investigators with different skill sets are required to move a fundamental observation through preclinical tests of clinical relevance then ultimately into the clinic. The various investigators must be able to organize effective research teams, whose compositions will differ as one moves closer to treating patients. Each phase, from discovery through application, has a set of barriers that can be summarized as culture, human resources/education, infrastructure, and regulatory. At a retreat of the Clinical Translational Research Committee of the American Association for Cancer Research, many of the issues facing academic centers were discussed and several recommendations are summarized here.


Asunto(s)
Investigación Biomédica/métodos , Neoplasias/terapia , Transferencia de Tecnología , Humanos
10.
Trans Am Clin Climatol Assoc ; 117: 85-101; discussion 101, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-18528466

RESUMEN

Our laboratory discovered that p53 can regulate the sensitivity to cancer therapies by affecting three critical aspects of cancer pharmacology: 1). The expression of drug targets; 2). the access of drugs to intracellular targets; and the response to DNA damage. We review the effects of p53 on antimicrotubule drugs through transcriptional regulation of MAP4 and stathmin (Oncoprotein 18). These two p53-regulated proteins control microtubule dynamics, regulate the sensitivity to taxanes and vinca alkaloids by changing the polymerization dynamics of tubulin and affecting the binding of drugs to microtubules. We found that overexpression of MAP4 increased microtubule polymerization and increased taxane binding and sensitivity. Overexpression of stathmin, a microtubule destabilizer, virtually abolished cellular taxane binding and increased resistance by over 1000-fold. Yet, despite an increased binding of vinca alkaloids to stathmin transfectants, we did not observe increased drug sensitivity. This was explained, at least in part, by a delay in G2/M transit. We also discovered that p53 could regulate the expression of multidrug resistance protein-1 (MRP1), a member of the ABC family of transporters that mediates the sensitivity to vinca alkaloids and anthracyclines. We found that as prostate cancer progressed from low stage/low grade to high stage/high grade there was an increased expression of both MRP1 and staining for p53, a surrogate for p53 mutations. We went on to show that p53 regulated the expression of MRP1 and that this produced resistance to doxorubicin and vinblastine. We further demonstrated that MRP1 overexpression blocked the accumulation of flutamide and hydroxy-flutamide (the active metabolite) without affecting transport of dihydrotesterone, thereby blocking access of the anti-androgen but not the androgen to intracellular androgen receptors. Finally, we reviewed the effects of DNA damage on p53 expression and MAP4 repression as a means to increase the effectiveness of breast cancer treatment. These data demonstrated the possibility of individualizing treatment based on p53 status.


Asunto(s)
Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Antineoplásicos/uso terapéutico , Daño del ADN , ADN de Neoplasias/genética , ADN de Neoplasias/metabolismo , Resistencia a Antineoplásicos , Expresión Génica , Genes p53 , Humanos , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Modelos Biológicos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Mutación , Neoplasias/genética , Estatmina/genética , Estatmina/metabolismo
11.
Cancer Res ; 63(7): 1515-9, 2003 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-12670898

RESUMEN

Overexpression of P-glycoprotein (P-gp), the MDR1 gene product, confers multidrug resistance (MDR) to cancer cells. Clinically, MDR is one of the major causes for chemotherapeutic treatment failure in cancer patients. To explore a new approach to circumventing MDR, we adopted RNA interference to target MDR1 gene expression. RNA interference is a conserved biological response to double-stranded RNA, which results in sequence-specific gene silencing [G. J. Hannon, Nature (Lond.), 418: 244-251, 2002]. We report that introduction of an MDR1-targeted small interfering RNA duplex into drug-resistant cancer cells markedly inhibited the expression of MDR1 mRNA and P-gp, as determined by reverse transcription-PCR and Western blot. Inhibition of P-gp expression by small interfering RNA enhanced the intracellular accumulation of and selectively restored sensitivity to drugs transported by P-gp. These studies indicate that RNA interference can modulate MDR in preclinical models.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Neoplasias de la Mama/tratamiento farmacológico , Resistencia a Múltiples Medicamentos/genética , Genes MDR/genética , ARN Interferente Pequeño/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Doxorrubicina/farmacocinética , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Paclitaxel/farmacocinética , Paclitaxel/farmacología , ARN Interferente Pequeño/administración & dosificación , Transfección , Células Tumorales Cultivadas , Vinblastina/farmacocinética , Vinblastina/farmacología
12.
Cancer Res ; 63(10): 2492-8, 2003 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-12750271

RESUMEN

Prostate cancer is the most common noncutaneous malignancy of American men. Although it can be initially treated with androgen deprivation therapy, tumors that relapse become resistant to future hormonal manipulation. We previously found that the multidrug resistance protein (MRP), MRP1, is overexpressed in advanced stage and grade human prostate cancer and is negatively regulated by p53. In this study, we sought to determine whether the cellular accumulation of the antiandrogen flutamide, a drug commonly used in the treatment of prostate cancer, is affected by MRP1 expression. There were significant differences between the wild-type and MRP1-overexpressing cells in efflux and accumulation of flutamide and hydroxyflutamide, its active metabolite. In contrast, transport of dihydrotestosterone was not affected by MRP1. Treating the cells with leukotriene D4, a known MRP1 substrate, or VX-710, an MRP1 modulator, restored flutamide and hydroxyflutamide accumulation. Finally, intracellular glutathione depletion with buthionine sulfoximine or energy depletion using 2-deoxy-D-glucose/sodium azide restored flutamide accumulation to that of parental cells while incubating the cells at 4 degrees C abolished MRP1-mediated transport. In summary, these studies indicate that flutamide and hydroxyflutamide but not dihydrotestosterone are transported by MRP1 and that these findings may contribute to our understanding of resistance to hormone refractory prostate cancer.


Asunto(s)
Antagonistas de Andrógenos/farmacocinética , Flutamida/farmacocinética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Transporte Biológico , Humanos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/biosíntesis , Células Tumorales Cultivadas
13.
Cancer Res ; 62(23): 6864-9, 2002 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-12460900

RESUMEN

Stathmin is a p53-regulated protein known to influence microtubule dynamics. Because several chemotherapeutic agents used to treat breast cancer alter the dynamic equilibrium of tubulin polymerization, stathmin may play an important role in determining the sensitivity to these drugs. Therefore, we evaluated the effect of stathmin expression on the action of taxanes and Vinca alkaloids using a panel of human breast cancer cell lines. Cell lines harboring mutant p53 expressed high levels of stathmin. Two cell lines with different levels of endogenous stathmin expression and isogenic-paired cell lines transfected to overexpress stathmin were used to determine whether or not stathmin modulated the sensitivity to drugs. Overexpression of stathmin decreased polymerization of microtubules, markedly decreased binding of paclitaxel, and increased binding of vinblastine. Stathmin overexpression decreased sensitivity to paclitaxel and, to a lesser extent, to vinblastine. In contrast, stathmin content had no significant effect on the sensitivity to chemotherapeutic drugs that do not target microtubules. Cell lines overexpressing stathmin were more likely to enter G(2) but less likely to enter mitosis as determined by fluorescence-activated cell sorting and mitotic index. This effect was magnified when stathmin-overexpressing cells were treated with vinblastine as measured by the detection of proteins phosphorylated in early mitosis. These data suggest that the action of antimicrotubule drugs can be affected by stathmin in at least two ways: (a) altered drug binding; and (b) growth arrest at the G(2) to M boundary. Mutant p53 breast cancers exhibiting high levels of stathmin may be resistant to antimicrotubule agents.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Proteínas de Microtúbulos , Paclitaxel/farmacología , Fosfoproteínas/fisiología , Vinblastina/farmacología , Antineoplásicos Fitogénicos/metabolismo , Western Blotting , Neoplasias de la Mama/genética , Ciclo Celular/fisiología , Humanos , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Mutación , Paclitaxel/metabolismo , Fosfoproteínas/biosíntesis , Fosfoproteínas/genética , Estatmina , Transfección , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética , Vinblastina/metabolismo
14.
Cancer Res ; 63(20): 6894-9, 2003 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-14583488

RESUMEN

Recent evidence suggests that the machinery of protein synthesis may provide novel targets for anticancer drugs. For example, aberrations in protein synthesis are commonly encountered in established cancers, and disruption by mutation or overexpression of translation factors can cause cellular transformation. We previously demonstrated that the activity of eukaryotic elongation factor 2 (eEF-2) kinase was markedly increased in several forms of malignancy and that nonspecific inhibitors of this enzyme promoted cell death. On the basis of the predicted amino acid sequence of eEF-2 kinase deduced from the cloned cDNA, we hypothesized that inhibitors of prokaryotic histidine kinases might also inhibit the activity of eEF-2 kinase. We describe herein the screening of a series of imidazolium histidine kinase inhibitors and the identification of an active lead compound, NH125. NH125 inhibited eEF-2 kinase activity (IC(50) = 60 nM) in vitro, blocked the phosphorylation of eEF-2 in intact cells, and showed relative selectivity over other protein kinases: protein kinase C (IC(50) = 7.5 microM), protein kinase A (IC(50) = 80 microM), and calmodulin-dependent kinase II (IC(50) > 100 microM). NH125 decreased the viability of 10 cancer cell lines with IC(50)s ranging from 0.7 to 4.7 microM. Forced overexpression of eEF-2 kinase in a glioma cell line produced 10-fold resistance to NH125. In conclusion, these results suggest that identification of potent inhibitors of eEF-2 kinase may lead to the development of new types of anticancer drugs.


Asunto(s)
Antineoplásicos/farmacología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Imidazoles/farmacología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Quinasa del Factor 2 de Elongación , Humanos , Fosforilación/efectos de los fármacos
15.
Cancer Prev Res (Phila) ; 9(8): 635-7, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27138792

RESUMEN

Malignant diseases develop slowly over time and are often preceded by identifiable premalignancies. As malignancy progresses, so does genomic complexity and the ability of cancers to evade most therapeutic interventions. Accordingly, with some notable exceptions, a relatively low percentage of advanced cancers are effectively treated and even fewer are cured. Despite this appreciation, much less attention has been paid to intercepting the disease process compared with that of treating well-established and refractory disease. One frequently cited reason is that the pharmaceutical industry is not interested in these pursuits. In this commentary, we attempt to define the true hurdles, the degree of difficulty inherent in each, and some important approaches to be considered. Cancer Prev Res; 9(8); 635-7. ©2016 AACR.


Asunto(s)
Neoplasias/prevención & control , Prevención Primaria , Diseño de Fármacos , Industria Farmacéutica/economía , Humanos , Enfermedades no Transmisibles/prevención & control , Prevención Primaria/economía , Prevención Primaria/tendencias
16.
Nat Commun ; 12(1): 5778, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34599183
17.
Cancer Prev Res (Phila) ; 9(1): 2-10, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26744449

RESUMEN

We have entered a transformative period in cancer prevention (including early detection). Remarkable progress in precision medicine and immune-oncology, driven by extraordinary recent advances in genome-wide sequencing, big-data analytics, blood-based technologies, and deep understanding of the tumor immune microenvironment (TME), has provided unprecedented possibilities to study the biology of premalignancy. The pace of research and discovery in precision medicine and immunoprevention has been astonishing and includes the following clinical firsts reported in 2015: driver mutations detected in circulating cell-free DNA in patients with premalignant lesions (lung); clonal hematopoiesis shown to be a premalignant state; molecular selection in chemoprevention randomized controlled trial (RCT; oral); striking efficacy in RCT of combination chemoprevention targeting signaling pathway alterations mechanistically linked to germline mutation (duodenum); molecular markers for early detection validated for lung cancer and showing promise for pancreatic, liver, and ovarian cancer. Identification of HPV as the essential cause of a major global cancer burden, including HPV16 as the single driver of an epidemic of oropharyngeal cancer in men, provides unique opportunities for the dissemination and implementation of public health interventions. Important to immunoprevention beyond viral vaccines, genetic drivers of premalignant progression were associated with increasing immunosuppressive TME; and Kras vaccine efficacy in pancreas genetically engineered mouse (GEM) model required an inhibitory adjuvant (Treg depletion). In addition to developing new (e.g., epigenetic) TME regulators, recent mechanistic studies of repurposed drugs (aspirin, metformin, and tamoxifen) have identified potent immune activity. Just as precision medicine and immune-oncology are revolutionizing cancer therapy, these approaches are transforming cancer prevention. Here, we set out a brief agenda for the immediate future of cancer prevention research (including a "Pre-Cancer Genome Atlas" or "PCGA"), which will involve the inter-related fields of precision medicine and immunoprevention - pivotal elements of a broader domain of personalized public health.


Asunto(s)
Neoplasias/inmunología , Neoplasias/prevención & control , Medicina de Precisión/métodos , Animales , Biomarcadores de Tumor/metabolismo , Biopsia , Vacunas contra el Cáncer , Progresión de la Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal , Humanos , Sistema Inmunológico , Inmunosupresores/uso terapéutico , Masculino , Ratones , Lesiones Precancerosas/patología , Ensayos Clínicos Controlados Aleatorios como Asunto , Análisis de Secuencia de ADN , Microambiente Tumoral
18.
Mol Cancer Res ; 1(6): 420-7, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12692261

RESUMEN

Multidrug resistant (MDR) cancer cells overexpressing P-glycoprotein (P-gp) display variations in invasive and metastatic behavior. We previously reported that these properties of MDR cancer cell lines overexpressing P-gp could be altered by chemotherapeutic drugs or MDR modulators (R. S. Kerbel et al., Cancer Surv., 7: 597-629, 1988). To attempt to clarify the mechanism(s) underlying these observations, we studied the expression of extracellular matrix metalloproteinase inducer (EMMPRIN), a glycoprotein enriched on the surface of tumor cells that can stimulate the production of matrix metalloproteinases (MMPs), in sensitive and MDR cancer cells. Using immunofluorescence staining and fluorescence-activated cell sorting analysis, we found that EMMPRIN expression was increased in MDR carcinoma cell lines, MCF-7/AdrR, KBV-1, and A2780Dx5, as compared to their parental counterparts. The MDR cell lines produced more matrix metalloproteinase-1 (MMP-1), matrix metalloproteinase-2 (MMP-2), and matrix metalloproteinase-9 (MMP-9), as determined by zymography, Western blot, and reverse transcription-PCR. Treatment of MDR cells with an anti-EMMPRIN antibody inhibited the activity of MMP-1, MMP-2, and MMP-9. In MDR cell line MCF-7/AdrR, an increased in vitro invasive ability was observed as compared with the sensitive line MCF-7, and EMMPRIN antibody could inhibit the in vitro invasion in drug-resistant cells. In addition, the expression and activity of MMP-1, MMP-2, and MMP-9 in MDR cells were decreased by treatment with U-0126, an inhibitor of mitogen-activated protein kinase/extracellular signal regulated kinase (MAPK/Erk). Our results suggest that during the development of MDR, the expression of EMMPRIN is responsible for the increased activity of MMP in MDR cell lines.


Asunto(s)
Antígenos CD , Antígenos de Neoplasias , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Glicoproteínas de Membrana/metabolismo , Basigina , División Celular , Línea Celular Tumoral , Humanos , Metaloproteinasas de la Matriz/genética , Metaloproteinasas de la Matriz/metabolismo , Glicoproteínas de Membrana/antagonistas & inhibidores , Invasividad Neoplásica
19.
Semin Oncol ; 32(6 Suppl 7): S16-21, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16360718

RESUMEN

The multidrug resistance (MDR) phenotype is often associated with recurrent breast cancer. Many cytotoxic agents used to treat breast cancer, such as anthracyclines and taxanes, are susceptible to MDR-mediated loss of sensitivity to these agents. Overexpression of mdr-1/P-glycoprotein (P-gp) is one of the main mechanisms underlying the development of the MDR phenotype. Also involved in the development of the MDR phenotype are other proteins from the ATP-binding cassette family of transporters (eg, MRP, BCRP), as well as alterations of tumor targets and their downstream effector molecules. Additionally, P-gp expression in other anatomic locations (such as the brush border of the gastrointestinal epithelium and blood-brain barrier) may further compromise the success of treatment for patients with breast cancer. Several strategies have been developed to overcome or circumvent MDR, mostly through inhibition or modulation of P-gp. Despite successful proof of concept in the laboratory, to date none of these agents has had a major impact in the clinic.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/fisiología , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/terapia , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Recurrencia Local de Neoplasia/tratamiento farmacológico , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/química , Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Basigina/fisiología , Humanos , Metaloproteinasas de la Matriz/metabolismo
20.
Clin Cancer Res ; 9(17): 6419-25, 2003 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-14695143

RESUMEN

PURPOSE: Recent studies suggest that activating point mutations in B-RAF may commonly occur in melanoma. We devised a method to detect point mutations in heterogeneous tissues containing both wild-type and mutant B-RAF and N-RAS genes by using site-directed mutagenesis to introduce new restrictions sites in the cDNA sequence when the specific point mutations are present. We used this technique to determine the incidence of mitogen-activated protein kinase (MAPK) mutations in human melanoma. EXPERIMENTAL DESIGN: We screened 85 melanoma samples for the most common B-RAF and N-RAS mutations found in melanoma using a site-directed mutagenesis-based detection technique. Western blotting was used to evaluate downstream up-regulation of the mitogen-activated protein kinase pathway in these tissues. RESULTS: Thirty-three samples (7 of 25 primaries, 15 of 25 regional metastases, 5 of 25 nodal metastases, and 6 of 10 distant metastases) harbored the V599E B-RAF mutation (39%), 12 contained a Q61R N-RAS mutation and 5 a Q61K N-RAS mutation. Western blotting with antiphosphorylated extracellular signal-regulated kinase 1/2 antibodies demonstrated up-regulation of the MAPK pathway in samples containing activating B-RAF or N-RAS mutations compared with wild-type samples. This method of detection was sensitive and specific with no false positives. CONCLUSIONS: Activating mutations of the MAPK pathway were present in approximately 60% of samples tested and caused activation of this cellular pathway that appears to be important in the pathogenesis of melanoma.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Melanoma/genética , Melanoma/patología , Mutación , Secuencia de Bases , Western Blotting , Codón , Cartilla de ADN/farmacología , ADN Complementario/metabolismo , Humanos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación Puntual , Sensibilidad y Especificidad , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA