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











Intervalo de año de publicación
1.
Cancer Res ; 77(21): 5741-5754, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28923859

RESUMEN

The trans-sulfuration enzyme cystathionine-ß-synthase (CBS) and its product hydrogen sulfide (H2S) are aberrantly upregulated in colorectal cancers, where they contribute to tumor growth and progression by both autocrine and paracrine mechanisms. However, it is unknown whether the CBS/H2S axis plays a role in colorectal carcinogenesis. Here, we report upregulation of CBS in human biopsies of precancerous adenomatous polyps and show that forced upregulation of CBS in an adenoma-like colonic epithelial cell line is sufficient to induce metabolic and gene expression profiles characteristic of colorectal cancer cells. Differentially expressed metabolites (65 increased and 20 decreased) clustered into the glycolytic pathway, nucleotide sugars, intermediates of the pentose phosphate pathway, and lipogenesis, including primarily phospholipids, sphingolipids, and bile acids. CBS upregulation induced broad changes in the NCM356 cell transcriptome with over 350 differentially expressed genes. These genes overlapped significantly with gene sets related to glycolysis, hypoxia, and a colon cancer cell phenotype, including genes regulated by NF-κB, KRAS, p53, and Wnt signaling, genes downregulated after E-cadherin knockdown, and genes related to increased extracellular matrix, cell adhesion, and epithelial-to-mesenchymal transition. The CBS-induced switch to an anabolic metabolism was associated with increased NCM356 cell bioenergetics, proliferation, invasion through Matrigel, resistance to anoikis, and CBS-dependent tumorigenesis in immunocompromised mice. Genetic ablation of CBS in CBS heterozygous mice (CBS+/- ) reduced the number of mutagen-induced aberrant colonic crypt foci. Taken together, these results establish that activation of the CBS/H2S axis promotes colon carcinogenesis. Cancer Res; 77(21); 5741-54. ©2017 AACR.


Asunto(s)
Pólipos Adenomatosos/genética , Colon/metabolismo , Cistationina betasintasa/genética , Mucosa Intestinal/metabolismo , Regulación hacia Arriba , Pólipos Adenomatosos/metabolismo , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Línea Celular , Movimiento Celular/genética , Colon/patología , Cistationina betasintasa/metabolismo , Femenino , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Humanos , Sulfuro de Hidrógeno/metabolismo , Mucosa Intestinal/patología , Masculino , Metabolómica/métodos , Ratones Noqueados , Ratones Desnudos , Trasplante Heterólogo
2.
Oncotarget ; 8(33): 55332-55352, 2017 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-28903423

RESUMEN

Tumor cells undergo a critical remodeling of intracellular Ca2+ homeostasis that contribute to important cancer hallmarks. Store-operated Ca2+ entry (SOCE), a Ca2+ entry pathway modulated by mitochondria, is dramatically enhanced in colon cancer cells. In addition, most cancer cells display the Warburg effect, a metabolic switch from mitochondrial metabolism to glycolysis that provides survival advantages. Accordingly, we investigated mitochondria control of store-operated currents (SOCs) in two cell lines previously selected for representing human normal colonic cells and colon cancer cells. We found that, in normal cells, mitochondria are important for SOCs activity but they are unable to prevent current inactivation. In contrast, in colon cancer cells, mitochondria are dispensable for SOCs activation but are able to prevent the slow, Ca2+-dependent inactivation of SOCs. This effect is associated to increased ability of tumor cell mitochondria to take up Ca2+ due to increased mitochondrial potential (ΔΨ) linked to the Warburg effect. Consistently with this view, selected non-steroidal anti-inflammatory drugs (NSAIDs) depolarize mitochondria, inhibit mitochondrial Ca2+ uptake and promote SOC inactivation, leading to inhibition of both SOCE and cancer cell proliferation. Thus, mitochondria sustain store-operated currents in colon cancer cells but not in normal colonic cells and this effect is counteracted by selected NSAIDs providing a mechanism for cancer chemoprevention.

3.
Int J Mol Sci ; 18(5)2017 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-28448473

RESUMEN

Colorectal cancer (CRC) cells undergo the remodeling of intracellular Ca2+ homeostasis, which contributes to cancer hallmarks such as enhanced proliferation, invasion and survival. Ca2+ remodeling includes critical changes in store-operated Ca2+ entry (SOCE) and Ca2+ store content. Some changes have been investigated at the molecular level. However, since nearly 100 genes are involved in intracellular Ca2+ transport, a comprehensive view of Ca2+ remodeling in CRC is lacking. We have used Next Generation Sequencing (NGS) to investigate differences in expression of 77 selected gene transcripts involved in intracellular Ca2+ transport in CRC. To this end, mRNA from normal human colonic NCM460 cells and human colon cancer HT29 cells was isolated and used as a template for transcriptomic sequencing and expression analysis using Ion Torrent technology. After data transformation and filtering, exploratory analysis revealed that both cell types were well segregated. In addition, differential gene expression using R and bioconductor packages show significant differences in expression of selected voltage-operated Ca2+ channels and store-operated Ca2+ entry players, transient receptor potential (TRP) channels, Ca2+ release channels, Ca2+ pumps, Na⁺/Ca2+ exchanger isoforms and genes involved in mitochondrial Ca2+ transport. These data provide the first comprehensive transcriptomic analysis of Ca2+ remodeling in CRC.


Asunto(s)
Canales de Calcio/genética , Calcio/metabolismo , Perfilación de la Expresión Génica , Canales de Calcio/metabolismo , Línea Celular Tumoral , Análisis por Conglomerados , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Regulación de la Expresión Génica , Células HT29 , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Análisis de Componente Principal , Análisis de Secuencia de ARN , Intercambiador de Sodio-Calcio/genética , Intercambiador de Sodio-Calcio/metabolismo , Canales de Potencial de Receptor Transitorio/genética , Canales de Potencial de Receptor Transitorio/metabolismo
4.
Oncotarget ; 6(29): 27403-15, 2015 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-26299804

RESUMEN

Previous studies suggest the anti-inflammatory drug, sulindac inhibits tumorigenesis by a COX independent mechanism involving cGMP PDE inhibition. Here we report that the cGMP PDE isozymes, PDE5 and 10, are elevated in colon tumor cells compared with normal colonocytes, and that inhibitors and siRNAs can selectively suppress colon tumor cell growth. Combined treatment with inhibitors or dual knockdown suppresses tumor cell growth to a greater extent than inhibition from either isozyme alone. A novel sulindac derivative, ADT-094 was designed to lack COX-1/-2 inhibitory activity but have improved potency to inhibit PDE5 and 10. ADT-094 displayed >500 fold higher potency to inhibit colon tumor cell growth compared with sulindac by activating cGMP/PKG signaling to suppress proliferation and induce apoptosis. Combined inhibition of PDE5 and 10 by treatment with ADT-094, PDE isozyme-selective inhibitors, or by siRNA knockdown also suppresses ß-catenin, TCF transcriptional activity, and the levels of downstream targets, cyclin D1 and survivin. These results suggest that dual inhibition of PDE5 and 10 represents novel strategy for developing potent and selective anticancer drugs.


Asunto(s)
Acetamidas/química , Neoplasias del Colon/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Indenos/química , Inhibidores de Fosfodiesterasa/química , Hidrolasas Diéster Fosfóricas/metabolismo , beta Catenina/metabolismo , Apoptosis , Células CACO-2 , Línea Celular Tumoral , Proliferación Celular , Simulación por Computador , Ciclina D1/metabolismo , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Células HT29 , Humanos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Concentración 50 Inhibidora , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Sulindac/química , Survivin , Transcripción Genética , beta Catenina/antagonistas & inhibidores
5.
Cancer Lett ; 365(1): 68-78, 2015 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-26021766

RESUMEN

Cancer cells rely mostly on glycolysis to meet their energetic demands, producing large amounts of lactate that are extruded to the tumour microenvironment by monocarboxylate transporters (MCTs). The role of MCTs in the survival of colorectal cancer (CRC) cells is scarce and poorly understood. In this study, we aimed to better understand this issue and exploit these transporters as novel therapeutic targets alone or in combination with the CRC classical chemotherapeutic drug 5-Fluorouracil. For that purpose, we characterized the effects of MCT activity inhibition in normal and CRC derived cell lines and assessed the effect of MCT inhibition in combination with 5-FU. Here, we demonstrated that MCT inhibition using CHC (α-cyano-4-hydroxycinnamic acid), DIDS (4,4'-diisothiocyanatostilbene-2,2'-disulphonic acid) and quercetin decreased cell viability, disrupted the glycolytic phenotype, inhibited proliferation and enhanced cell death in CRC cells. These results were confirmed by specific inhibition of MCT1/4 by RNA interference. Notably, we showed that 5-FU cytotoxicity was potentiated by lactate transport inhibition in CRC cells, either by activity inhibition or expression silencing. These findings provide novel evidence for the pivotal role of MCTs in CRC maintenance and survival, as well as for the use of these transporters as potential new therapeutic targets in combination with CRC conventional therapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Colorrectales/metabolismo , Ácido Láctico/metabolismo , Transportadores de Ácidos Monocarboxílicos/antagonistas & inhibidores , Antimetabolitos Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Relación Dosis-Respuesta a Droga , Fluorouracilo/farmacología , Glucólisis/efectos de los fármacos , Humanos , Moduladores del Transporte de Membrana/farmacología , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas Musculares/antagonistas & inhibidores , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Interferencia de ARN , Simportadores/antagonistas & inhibidores , Simportadores/genética , Simportadores/metabolismo , Factores de Tiempo , Transfección
6.
Am J Pathol ; 185(4): 1135-44, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25794709

RESUMEN

Methionine adenosyltransferase 2B (MAT2B) encodes for variant proteins V1 and V2 that interact with GIT1 to increase ERK activity and growth in human liver and colon cancer cells. MAT2B or GIT1 overexpression activates MEK. This study explores the mechanism for MEK activation. We examined protein-protein interactions by co-immunoprecipitation and verified by confocal microscopy and pull-down assay using recombinant or in vitro translated proteins. Results were confirmed in an orthotopic liver cancer model. We found that MAT2B and GIT1-mediated MEK1/2 activation was not mediated by PAK1 or Src in HepG2 or RKO cells. Instead, MAT2B and GIT1 interact with B-Raf and c-Raf and enhance recruitment of Raf proteins to MEK1/2. MAT2B-GIT1 activates c-Raf, which is the key mediator for MEK/12 activation, because this still occurred in RKO cells that express constitutively active B-Raf mutant. The mechanism lies with the ability of MAT2B-GIT1 to activate Ras and promote B-Raf/c-Raf heterodimerization. Interestingly, MAT2B but not GIT1 can directly interact with Ras, which increases protein stability. Finally, increased Ras-Raf-MEK signaling occurred in phenotypically more aggressive liver cancers overexpressing MAT2B variants and GIT1. In conclusion, interaction between MAT2B and GIT1 serves as a scaffold and facilitates signaling in multiple steps of the Ras/Raf/MEK/ERK pathway, further emphasizing the importance of MAT2B/GIT1 interaction in cancer growth.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular/metabolismo , Neoplasias del Colon/metabolismo , Neoplasias Hepáticas/metabolismo , Metionina Adenosiltransferasa/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas ras/metabolismo , Línea Celular Tumoral , Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Activación Enzimática , Humanos , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/patología , Unión Proteica , Multimerización de Proteína , Proteínas Proto-Oncogénicas c-raf/metabolismo , Regulación hacia Arriba , Quinasas p21 Activadas/metabolismo , Familia-src Quinasas/metabolismo
7.
Proc Natl Acad Sci U S A ; 111(46): 16520-5, 2014 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-25368155

RESUMEN

Colorectal tumorigenesis is driven by genetic alterations in the adenomatous polyposis coli (APC) tumor suppressor pathway and effectively inhibited by nonsteroidal antiinflammatory drugs (NSAIDs). However, how NSAIDs prevent colorectal tumorigenesis has remained obscure. We found that the extrinsic apoptotic pathway and the BH3 interacting-domain death agonist (BID) are activated in adenomas from NSAID-treated patients. Loss of BID abolishes NSAID-mediated tumor suppression, survival benefit, and apoptosis in tumor-initiating stem cells in APC(Min/+) mice. BID-mediated cross-talk between the extrinsic and intrinsic apoptotic pathways is responsible for selective killing of neoplastic cells by NSAIDs. We further demonstrate that NSAIDs induce death receptor signaling in both cancer and normal cells, but only activate BID in cells with APC deficiency and ensuing c-Myc activation. Our results suggest that NSAIDs suppress intestinal tumorigenesis through BID-mediated synthetic lethality triggered by death receptor signaling and gatekeeper mutations, and provide a rationale for developing more effective cancer prevention strategies and agents.


Asunto(s)
Poliposis Adenomatosa del Colon/prevención & control , Antiinflamatorios no Esteroideos/farmacología , Apoptosis , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/fisiología , Genes APC , Poliposis Adenomatosa del Colon/patología , Animales , Proteínas Reguladoras de la Apoptosis/fisiología , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/antagonistas & inhibidores , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/deficiencia , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/genética , Caspasas/fisiología , Línea Celular Tumoral , Colon/patología , Regulación Neoplásica de la Expresión Génica , Genes myc , Humanos , Indometacina/farmacología , Intestino Delgado/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mitocondrias/metabolismo , Especificidad de Órganos , Pirazoles/farmacología , ARN Interferente Pequeño/farmacología , Receptores de Muerte Celular/fisiología , Células Madre/metabolismo , Células Madre/patología , Sulfonamidas/farmacología , Sulindac/farmacología
8.
J Biol Chem ; 289(42): 28765-82, 2014 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-25143380

RESUMEN

We have investigated the molecular basis of intracellular Ca(2+) handling in human colon carcinoma cells (HT29) versus normal human mucosa cells (NCM460) and its contribution to cancer features. We found that Ca(2+) stores in colon carcinoma cells are partially depleted relative to normal cells. However, resting Ca(2+) levels, agonist-induced Ca(2+) increases, store-operated Ca(2+) entry (SOCE), and store-operated currents (ISOC) are largely enhanced in tumor cells. Enhanced SOCE and depleted Ca(2+) stores correlate with increased cell proliferation, invasion, and survival characteristic of tumor cells. Normal mucosa cells displayed small, inward Ca(2+) release-activated Ca(2+) currents (ICRAC) mediated by ORAI1. In contrast, colon carcinoma cells showed mixed currents composed of enhanced ICRAC plus a nonselective ISOC mediated by TRPC1. Tumor cells display increased expression of TRPC1, ORAI1, ORAI2, ORAI3, and STIM1. In contrast, STIM2 protein was nearly depleted in tumor cells. Silencing data suggest that enhanced ORAI1 and TRPC1 contribute to enhanced SOCE and differential store-operated currents in tumor cells, whereas ORAI2 and -3 are seemingly less important. In addition, STIM2 knockdown decreases SOCE and Ca(2+) store content in normal cells while promoting apoptosis resistance. These data suggest that loss of STIM2 may underlie Ca(2+) store depletion and apoptosis resistance in tumor cells. We conclude that a reciprocal shift in TRPC1 and STIM2 contributes to Ca(2+) remodeling and tumor features in colon cancer.


Asunto(s)
Calcio/metabolismo , Moléculas de Adhesión Celular/metabolismo , Neoplasias Colorrectales/metabolismo , Regulación Neoplásica de la Expresión Génica , Canales Catiónicos TRPC/metabolismo , Apoptosis , Carcinogénesis , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Colon/metabolismo , Fenómenos Electrofisiológicos , Perfilación de la Expresión Génica , Silenciador del Gen , Humanos , Inositol 1,4,5-Trifosfato/química , Mucosa Intestinal/patología , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Molécula de Interacción Estromal 1 , Molécula de Interacción Estromal 2
9.
Cancer Cell ; 25(4): 469-83, 2014 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-24735923

RESUMEN

MicroRNA deregulation is frequent in human colorectal cancers (CRCs), but little is known as to whether it represents a bystander event or actually drives tumor progression in vivo. We show that miR-135b overexpression is triggered in mice and humans by APC loss, PTEN/PI3K pathway deregulation, and SRC overexpression and promotes tumor transformation and progression. We show that miR-135b upregulation is common in sporadic and inflammatory bowel disease-associated human CRCs and correlates with tumor stage and poor clinical outcome. Inhibition of miR-135b in CRC mouse models reduces tumor growth by controlling genes involved in proliferation, invasion, and apoptosis. We identify miR-135b as a key downsteam effector of oncogenic pathways and a potential target for CRC treatment.


Asunto(s)
Neoplasias del Colon/genética , MicroARNs/genética , Animales , Procesos de Crecimiento Celular/genética , Línea Celular Tumoral , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Xenoinjertos , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , MicroARNs/metabolismo , Transfección
10.
Mol Cancer Ther ; 12(9): 1848-59, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23804703

RESUMEN

Nonsteroidal anti-inflammatory drugs (NSAID) display promising antineoplastic activity for colorectal and other cancers, but toxicity from COX inhibition limits their long-term use for chemoprevention. Previous studies have concluded that the basis for their tumor cell growth inhibitory activity does not require COX inhibition, although the underlying mechanism is poorly understood. Here, we report that the NSAID sulindac sulfide inhibits cyclic guanosine 3',5'-monophosphate phosphodiesterase (cGMP PDE) activity to increase intracellular cGMP levels and activate cGMP-dependent protein kinase (PKG) at concentrations that inhibit proliferation and induce apoptosis of colon tumor cells. Sulindac sulfide did not activate the cGMP/PKG pathway, nor affect proliferation or apoptosis in normal colonocytes. Knockdown of the cGMP-specific PDE5 isozyme by siRNA and PDE5-specific inhibitors tadalafil and sildenafil also selectively inhibited the growth of colon tumor cells that expressed high levels of PDE5 compared with colonocytes. The mechanism by which sulindac sulfide and the cGMP/PKG pathway inhibits colon tumor cell growth involves the transcriptional suppression of ß-catenin to inhibit Wnt/ß-catenin T-cell factor transcriptional activity, leading to downregulation of cyclin D1 and survivin. These observations suggest that safer and more efficacious sulindac derivatives can be developed for colorectal cancer chemoprevention by targeting PDE5 and possibly other cGMP-degrading isozymes.


Asunto(s)
Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/patología , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Sulindac/análogos & derivados , Vía de Señalización Wnt/efectos de los fármacos , Antineoplásicos/análisis , Apoptosis/efectos de los fármacos , Células CACO-2 , Carbolinas/farmacología , Línea Celular , Neoplasias del Colon/metabolismo , GMP Cíclico/genética , Ciclina D1/metabolismo , Células HCT116 , Células HT29 , Humanos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Inhibidores de Fosfodiesterasa 5/farmacología , Piperazinas/farmacología , Purinas/farmacología , Citrato de Sildenafil , Sulfonas/farmacología , Sulindac/análisis , Sulindac/farmacología , Survivin , Tadalafilo
11.
Am J Physiol Gastrointest Liver Physiol ; 303(11): G1270-8, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22982339

RESUMEN

Subepithelial myofibroblasts are involved in the initiation and coordination of intestinal epithelial repair, but the molecular signaling pathways are largely unknown. The cellular adaptations that occur during repair range from dedifferentiation and migration to proliferation and redifferentiation, in a way that is strongly reminiscent of normal crypt-to-villus epithelial maturation. We therefore hypothesized that Wnt/ß-catenin signaling may have a pivotal role in intestinal epithelial wound repair. We used the established scratch wound method in Caco-2 cells and in nontransformed NCM460 cells to monitor the effects of IL-1ß-stimulated colonic myofibroblasts (CCD-18co) on intestinal epithelial repair, with immunoblotting and immunodepletion to examine the conditioned media. Conditioned media from IL-1ß-stimulated, but not -untreated, myofibroblasts increased Caco-2 wound closure twofold over 24 h. IL-1ß-stimulated myofibroblasts downregulated the differentiation marker sucrase-isomaltase in the Caco-2 cells, whereas the proliferation marker c-myc was upregulated. Array expression profiling identified Wnt-5a as the Wnt-related gene that was most upregulated (28-fold) by IL-1ß stimulation of CCDs. Recombinant Wnt-5a enhanced proliferation of Caco-2 and NCM460 cells. In scratch assays, it increased migration of the leading edge in both cell lines. Wnt-5a immunodepletion of the IL-1ß-CCD conditioned media abrogated the ability to enhance the repair. Wnt-5a often acts through a noncanonical signal transduction pathway. Further experiments supported this pathway in epithelial wound healing: IL-1ß-CCD-mediated repair was not affected by the addition of the canonical Wnt antagonist Dickkopf-1. Furthermore, media from stimulated myofibroblasts (but not Wnt-5a-depleted media) increased c-jun in Caco-2 cell nuclear extracts. Myofibroblast-mediated noncanonical Wnt-5a signaling is therefore important in the dedifferentiation and migration stages of epithelial wound repair.


Asunto(s)
Interleucina-1beta/farmacología , Miofibroblastos/efectos de los fármacos , Proteínas Proto-Oncogénicas/fisiología , Proteínas Wnt/fisiología , Cicatrización de Heridas/efectos de los fármacos , Células CACO-2 , Desdiferenciación Celular , Línea Celular , Movimiento Celular , Medios de Cultivo Condicionados/farmacología , Regulación hacia Abajo , Humanos , Miofibroblastos/fisiología , Proteínas Proto-Oncogénicas/biosíntesis , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba , Proteínas Wnt/biosíntesis , Proteína Wnt-5a , Cicatrización de Heridas/fisiología , beta Catenina/metabolismo
12.
Cancer Lett ; 324(1): 98-108, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22579651

RESUMEN

We recently demonstrated that p38α is required to maintain colorectal cancer (CRC) metabolism, as its inhibition leads to FoxO3A activation, autophagy, cell death, and tumor growth reduction both in vitro and in vivo. Here we show that inhibition of p38α is followed by TRAIL-mediated activation of caspase-8 and FoxO3A-dependent HER3 upregulation with consequent overactivation of the MEK-ERK1/2 survival pathway. p38α and MEK combined inhibition specifically induces apoptosis by enabling TRAIL signaling propagation through t-Bid and caspase-3, and fosters cell death in CRC cells and preclinical mouse models. Current MEK1-directed pharmacological strategies could thus be exploited, in combination with p38α inhibition, to develop new approaches for CRC treatment.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Benzamidas/farmacología , Caspasa 8/metabolismo , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Línea Celular Tumoral , Neoplasias Colorrectales/tratamiento farmacológico , Difenilamina/análogos & derivados , Difenilamina/farmacología , Inhibidores Enzimáticos/farmacología , Femenino , Células HT29 , Humanos , Imidazoles/farmacología , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 1/metabolismo , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Proteína Quinasa 14 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Fosforilación , Piridinas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
13.
J Nat Prod ; 75(1): 26-33, 2012 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-22216935

RESUMEN

Hamamelis virginiana (witch hazel) bark is a rich source of condensed and hydrolyzable tannins reported to exert a protective action against colon cancer. The present study characterizes different witch hazel tannins as selective cytotoxic agents against colon cancer. To cover the structural diversity of the tannins that occur in H. virginiana bark, the hydrolyzable tannins, hamamelitannin and pentagalloylglucose, together with a proanthocyanidin-rich fraction (F800H4) were selected for the study. Treatment with these compounds reduced tumor viability and induced apoptosis, necrosis, and S-phase arrest in the cell cycle of HT29 cells, with hamamelitannin being the most efficient. Owing to polyphenol-mediated H(2)O(2) formation in the incubation media, the antiproliferative effect was determined in the presence and absence of catalase to rule out any such interference. The presence of catalase significantly changed the IC(50) only for F800H4. Furthermore, at concentrations that inhibit the growth of HT29 cells by 50%, hamamelitannin had no harmful effects on NCM460 normal colonocytes, whereas pentagalloylglucose inhibited both cancerous and normal cell growth. Using the TNPTM assay, we identified a highly reactive phenolic position in hamamelitannin, which may explain its efficacy at inhibiting colon cancer growth.


Asunto(s)
Antineoplásicos Fitogénicos/aislamiento & purificación , Antineoplásicos Fitogénicos/farmacología , Ácido Gálico/análogos & derivados , Hamamelis/química , Hexosas/aislamiento & purificación , Hexosas/farmacología , Taninos Hidrolizables/aislamiento & purificación , Taninos Hidrolizables/farmacología , Antineoplásicos Fitogénicos/química , Apoptosis/efectos de los fármacos , Neoplasias del Colon , Ensayos de Selección de Medicamentos Antitumorales , Ácido Gálico/química , Ácido Gálico/aislamiento & purificación , Ácido Gálico/farmacología , Hexosas/química , Humanos , Peróxido de Hidrógeno/análisis , Taninos Hidrolizables/química , Estructura Molecular , Corteza de la Planta/química
14.
Nutr Cancer ; 64(1): 128-35, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22171558

RESUMEN

Methylselenol is hypothesized to be a critical selenium metabolite for anticancer action, and differential chemopreventive effects of methylselenol on cancerous and noncancerous cells may play an important role. In this study, the submicromolar concentrations of methylselenol were generated by incubating methionase with seleno-L methionine, and colon-cancer-derived HCT-116 cells and noncancerous colon NCM460 cells were exposed to methylselenol. Methylselenol exposure inhibited cell growth and led to an increase in G1 and G2 fractions with a concomitant drop in S-phase and an induction of apoptosis in HCT116, but to a much lesser extent in NCM460 colon cells. Similarly, the examination of mitogen-activated protein kinase (MAPK) and cellular myelocytomatosis oncogene (c-Myc) signaling status revealed that methylselenol inhibited the phosphorylation of extracellular-regulated kinase1/2 and p38 mitogen-activated protein kinase and the expression of c-Myc in HCT116 cells, but also to a lesser extent in NCM460 cells. The other finding is that methylselenol inhibits sarcoma kinase phosphorylation in HCT116 cells. In contrast, methylselenol upregulated the phosphorylation of sarcoma and focal adhesion kinase survival signals in the noncancerous NCM460 cells. Collectively, methylselenol's stronger potential of inhibiting cell proliferation/survival signals in the cancerous HCT116 cells when compared with that in noncancerous NCM460 cells may partly explain the potential of methylselenol's anticancer action.


Asunto(s)
Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Metanol/análogos & derivados , Compuestos de Organoselenio/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Colon/citología , Colon/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Relación Dosis-Respuesta a Droga , Activación Enzimática , Quinasa 1 de Adhesión Focal/metabolismo , Fase G1/efectos de los fármacos , Humanos , Metanol/farmacología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Fase S/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
15.
Nutr Cancer ; 63(2): 248-55, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21271458

RESUMEN

Sulforaphane (SFN) is a naturally occurring chemopreventive agent; the induction of cell cycle arrest and apoptosis is a key mechanism by which SFN exerts its colon cancer prevention. However, little is known about the differential effects of SFN on colon cancer and normal cells. In this study, we demonstrated that SFN (15 µmol/L) exposure (72 h) inhibited cell proliferation by up to 95% in colon cancer cells (HCT116) and by 52% in normal colon mucosa-derived (NCM460) cells. Our data also showed that SFN exposure (5 and 10 µmol/L) led to the reduction of G1 phase cell distribution and an induction of apoptosis in HCT116 cells, but to a much lesser extent in NCM460 cells. Furthermore, the examination of mitogen-activated protein kinase (MAPK) signaling status revealed that SFN upregulated the phosphorylation of extracellular-regulated kinase 1/2 (ERK1/2) in NCM460 cells but not in HCT116 cells. In contrast, SFN enhanced the phosphorylation of stress-activated protein kinase (SAPK) and decreased cellular myelocytomatosis oncogene (c-Myc) expression in HCT116 cells but not NCM460 cells. Taken together, the activation of survival signaling in NCM460 cells and apoptotic signaling in HCT116 cells may play a critical role in SFN's stronger potential of inhibiting cell proliferation in colon cancer cells than in normal colon cells.


Asunto(s)
Anticarcinógenos/farmacología , Apoptosis , Transducción de Señal , Tiocianatos/farmacología , Ciclo Celular , Línea Celular , Proliferación Celular , Colon/citología , Colon/metabolismo , Neoplasias del Colon/metabolismo , Fase G1 , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Humanos , Isotiocianatos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Sulfóxidos
16.
Anticancer Res ; 30(6): 1881-5, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20651330

RESUMEN

BACKGROUND/AIM: Methionine inhibits proliferation of breast and prostate cancer cells. This study aimed to determine cell cycle effects of methionine and selectivity for cancer cells. MATERIALS AND METHODS: MCF-7 (breast), LNCaP (prostate), and LS-174 (colon) cancer cells (wild-type p53), DU-145 (prostate) and SW480 (colon) cancer cells (mutated p53), and immortalized, non-tumorigenic MCF-10A (breast), BPH-1 (prostate), and NCM-460 (colon) epithelial cells were used. Cell cycle effects were assessed by flow cytometry and cell cycle-related gene expression by microarray analysis and QRT-PCR. RESULTS: L-Methionine at 5 mg/ml for 72 hours (non-apoptotic) arrested cell cycle in LNCaP, DU145, and MCF-7 cells, but not in untransformed cells, nor in LS-174 cells. LNCaP and MCF-7 cells were arrested at G(1), but DU-145 at S. Methionine up-regulated CDKIs and down-regulated CDKs. CONCLUSION: L-Methionine selectively inhibits proliferation of breast and prostate cancer cells, but not non-tumorigenic cells, and may thus have therapeutic benefits. p53 status appeared to determine the cell cycle stage at which methionine acts.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Metionina/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias de la Mama/patología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Neoplasias Pancreáticas/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína p53 Supresora de Tumor/análisis
17.
J Cell Physiol ; 225(1): 73-83, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20648625

RESUMEN

The extracellular Ca(2+)-sensing receptor (CaR) is increasingly implicated in the regulation of multiple cellular functions in the gastrointestinal tract, including secretion, proliferation and differentiation of intestinal epithelial cells. However, the signaling mechanisms involved remain poorly defined. Here we examined signaling pathways activated by the CaR, including Ca(2+) oscillations, in individual human colon epithelial cells. Single cell imaging of colon-derived cells expressing the CaR, including SW-480, HT-29, and NCM-460 cells, shows that stimulation of this receptor by addition of aromatic amino acids or by an elevation of the extracellular Ca(2+) concentration promoted striking intracellular Ca(2+) oscillations. The intracellular calcium oscillations in response to extracellular Ca(2+) were of sinusoidal pattern and mediated by the phospholipase C/diacylglycerol/inositol 1,4,5-trisphosphate pathway as revealed by a biosensor that detects the accumulation of diacylglycerol in the plasma membrane. The intracellular calcium oscillations in response to aromatic amino acids were of transient type, that is, Ca(2+) spikes that returned to baseline levels, and required an intact actin cytoskeleton, a functional Rho, Filamin A and the ion channel TRPC1. Further analysis showed that re-expression and stimulation of the CaR in human epithelial cells derived from normal colon and from colorectal adenocarcinoma inhibits their proliferation. This inhibition was associated with the activation of the signaling pathway that mediates the generation of sinusoidal, but not transient, intracellular Ca(2+) oscillations. Thus, these results indicate that the CaR can function in two signaling modes in human colonic epithelial cells offering a potential link between gastrointestinal responses and food/nutrients uptake and metabolism.


Asunto(s)
Señalización del Calcio/fisiología , Calcio/metabolismo , Proliferación Celular , Colon/citología , Células Epiteliales/fisiología , Receptores Sensibles al Calcio/metabolismo , Actinas/metabolismo , Línea Celular , Proteínas Contráctiles/metabolismo , Citoesqueleto/metabolismo , Células Epiteliales/citología , Filaminas , Humanos , Proteínas de Microfilamentos/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Sensibles al Calcio/genética , Canales Catiónicos TRPC/metabolismo , Proteínas de Unión al GTP rho/metabolismo
18.
Nature ; 464(7291): 1058-61, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20348907

RESUMEN

Cancer chemoprevention uses natural, synthetic, or biological substances to reverse, suppress, or prevent either the initial phase of carcinogenesis or the progression of neoplastic cells to cancer. It holds promise for overcoming problems associated with the treatment of late-stage cancers. However, the broad application of chemoprevention is compromised at present by limited effectiveness and potential toxicity. To overcome these challenges, here we developed a new chemoprevention approach that specifically targets premalignant tumour cells for apoptosis. We show that a deficiency in the adenomatous polyposis coli (APC) gene and subsequent activation of beta-catenin lead to the repression of cellular caspase-8 inhibitor c-FLIP (also known as CFLAR) expression through activation of c-Myc, and that all-trans-retinyl acetate (RAc) independently upregulates tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) death receptors and suppresses decoy receptors. Thus, the combination of TRAIL and RAc induces apoptosis in APC-deficient premalignant cells without affecting normal cells in vitro. In addition, we show that short-term and non-continuous TRAIL and RAc treatment induce apoptosis specifically in intestinal polyps, strongly inhibit tumour growth, and prolong survival in multiple intestinal neoplasms C57BL/6J-Apc(Min)/J (Apc(Min)) mice. With our approach, we further demonstrate that TRAIL and RAc induce significant cell death in human colon polyps, providing a potentially selective approach for colorectal cancer chemoprevention by targeting APC-deficient cells for apoptosis.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/deficiencia , Apoptosis/efectos de los fármacos , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/prevención & control , Vitamina A/análogos & derivados , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Diterpenos , Regulación de la Expresión Génica/efectos de los fármacos , Genes APC , Humanos , Pólipos Intestinales/tratamiento farmacológico , Pólipos Intestinales/patología , Ratones , Ratones Endogámicos C57BL , Lesiones Precancerosas/tratamiento farmacológico , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ésteres de Retinilo , Transducción de Señal/efectos de los fármacos , Tasa de Supervivencia , Ligando Inductor de Apoptosis Relacionado con TNF/administración & dosificación , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/uso terapéutico , Factores de Tiempo , Vitamina A/administración & dosificación , Vitamina A/farmacología , Vitamina A/uso terapéutico , beta Catenina/metabolismo
19.
Int J Cancer ; 126(4): 864-75, 2010 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19697327

RESUMEN

Expression of gastrin and cholecystokinin 2 (CCK(2)) receptor splice variants (CCK(2)R and CCK(2i4sv)R) are upregulated in human colonic adenomas where they are thought to contribute to tumor growth and progression. To determine the effects of ectopic CCK(2) receptor variant expression on colonic epithelial cell growth in vitro and in vivo, we employed the non-tumorigenic colonic epithelial cell line, NCM356. Receptor expression was induced using a retroviral expression vector containing cDNAs for either CCK(2i4sv)R or CCK(2)R. RT-PCR and intracellular Ca(2+) ([Ca(2+)](i)) imaging of RIE/CCK(2)R cells treated with conditioned media (CM) from NCM356 revealed that NCM356 cells express gastrin mRNA and secrete endogenous, biologically active peptide. NCM356 cells expressing either CCK(2)R or CCK(2i4sv)R (71 and 81 fmol/mg, respectively) grew faster in vitro, and exhibited an increase in basal levels of phosphorylated ERK (pERK), compared with vector. CCK(2) receptor selective antagonist, YM022, partially inhibited the growth of both receptor-expressing NCM356 cells, but not the control cells. Inhibitors of mitogen activated protein kinase pathway (MEK/ERK) or protein kinase C (PKC) isozymes partially inhibited the elevated levels of basal pERK and in vitro growth of receptor-expressing cells. Vector-NCM356 cells did not form tumors in nude mice, whereas, either CCK(2) receptor-expressing cells formed large tumors. Autocrine activation CCK(2) receptor variants are sufficient to increase in vitro growth and tumorigenicity of non-transformed NCM356 colon epithelial cells through a pathway involving PKC and the MEK/ERK axis. These findings support the hypothesis that expression of gastrin and its receptors in human colonic adenomas contributes to tumor growth and progression.


Asunto(s)
Colon/fisiología , Neoplasias Colorrectales/patología , Mucosa Intestinal/fisiología , Receptor de Colecistoquinina B/genética , Adenoma/patología , Animales , Calcio/metabolismo , Carcinoma/genética , Técnicas de Cultivo de Célula/métodos , División Celular/genética , Colon/metabolismo , Colon/patología , Neoplasias Colorrectales/genética , Cartilla de ADN , Progresión de la Enfermedad , Gastrinas/genética , Gastrinas/metabolismo , Variación Genética , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Ratones , Mutación , Estadificación de Neoplasias , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Free Radic Biol Med ; 47(8): 1190-8, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19647792

RESUMEN

Although oxidative stress induces mitochondrial DNA (mtDNA) damage, a role for redox in modulating mtDNA oxidation and repair is relatively unexplored. This study examines the contribution of cellular glutathione (GSH) redox status to menadione (MQ)-induced mtDNA damage and postoxidant mtDNA recovery in a nontransformed NCM460 colonic cell line. We show that MQ caused dose-dependent increases in mtDNA damage that were blunted by N-acetylcysteine, a thiol antioxidant. Damage to mtDNA paralleled mitochondrial protein disulfide formation and glutathione disulfide increases in the cytosol and mitochondria and was exacerbated by inhibition of GSH synthesis in accordance with decreased cytosolic and mitochondrial GSH. Blockade of mitochondrial GSH (mtGSH) transport potentiated mtDNA damage, which was prevented by overexpression of the oxoglutarate mtGSH carrier, underscoring a link between mtGSH and mtDNA responsiveness to oxidative stress. The removal of MQ posttreatment elicited mtDNA recovery to basal levels by 4 h, indicating complete repair. Notably, mtDNA recovery was preceded by restored cytosolic and mtGSH levels at 2 h, suggesting a connection between the maintenance of cell GSH and effective mtDNA repair. The MQ-induced dose-dependent increase in mtDNA damage was attenuated by overexpressing mitochondrial 8-oxoguanine DNA glycosylase (Ogg1), consistent with 7,8-dihydro-8-oxoguanine being a major oxidative mtDNA lesion. Collectively, the results show that oxidative mtDNA damage in colonic cells is highly responsive to the mtGSH status and that postoxidant mtDNA recovery may also be GSH sensitive.


Asunto(s)
Colon/efectos de los fármacos , Colon/metabolismo , Daño del ADN , ADN Mitocondrial/genética , Glutatión/metabolismo , Oxidantes/farmacología , Células Cultivadas , ADN Glicosilasas/metabolismo , Células Epiteliales/metabolismo , Disulfuro de Glutatión/metabolismo , Guanina/análogos & derivados , Guanina/metabolismo , Humanos , Oxidación-Reducción , Vitamina K 3/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA