Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Biomark Cancer ; 7(Suppl 1): 29-37, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26568685

RESUMEN

Colon cancer development and malignant progression are driven by genetic and epigenetic alterations in tumor cells and by factors from the tumor microenvironment. Cancer cells become reliant on the activity of specific oncogenes and on prosurvival and proliferative signals they receive from the abnormal environment they create and reside in. Accordingly, the response to anticancer therapy is determined by genetic and epigenetic changes that are intrinsic to tumor cells and by the factors present in the tumor microenvironment. Recent advances in the understanding of the involvement of the tumor microenvironment in tumor progression and therapeutic response are optimizing the application of prognostic and predictive factors in colon cancer. Moreover, new targets in the tumor microenvironment that are amenable to therapeutic intervention have been identified. Because stromal cells are with rare exceptions genetically stable, the tumor microenvironment has emerged as a preferred target for therapeutic drugs. In this review, we discuss the role of stromal fibroblasts and macrophages in colon cancer progression and in the response of colon cancer patients to therapy.

2.
Indian J Biochem Biophys ; 40(3): 180-5, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22900308

RESUMEN

The effect of chronic cold stress on the composition and function of rat intestinal brush border membrane (BBM) was studied. Various lipid fractions from intestinal BBM viz. cholesterol (p < 0.01), phospholipids (p < 0.01), triglycerides (p < 0.05) and gangliosides (p < 0.05) were significantly reduced in cold stressed animals, as compared to controls. Analysis of membrane saccharide content revealed a significant increase in sialic acid (25%) and hexosamine (36%) contents and a reduction in fucose (19%) content in cold stressed rats. Determination of various enzyme activities in BBM showed significantly enhanced activities of alkaline phosphatase ( p < 0.01), lactase ( p < 0.001) and leucine aminopeptidase ( p < 0.001), whereas sucrase activity was reduced ( p < 0.05) under these conditions. The magnitude and site of these alterations across the crypt-villus axis varied from enzyme to enzyme. These findings suggest that chronic cold stress results in profound alterations in intestinal BBM. Altered structure and function of intestinal BBM may play a role in stress-induced derangements in gastrointestinal tract.


Asunto(s)
Respuesta al Choque por Frío , Intestinos/enzimología , Intestinos/ultraestructura , Animales , Metabolismo de los Hidratos de Carbono , Mucosa Intestinal/metabolismo , Intestinos/citología , Metabolismo de los Lípidos , Masculino , Microvellosidades/enzimología , Microvellosidades/metabolismo , Ratas , Factores de Tiempo
3.
Front Oncol ; 4: 88, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24818101

RESUMEN

Signaling between tumor cells and the associated stroma has an important impact on cancer initiation and progression. The tumor microenvironment has a paradoxical role in tumor progression and fibroblasts, a major component of the tumor stroma, have been shown to either inhibit or promote cancer development. In this study, we established that normal intestinal fibroblasts activate STAT1 signaling in colon cancer cells and, in contrast to cancer-associated fibroblasts, inhibit growth of tumor cells. Treatment of 18Co fibroblasts with the proinflammatory cytokine TNFα interfered with their ability to trigger STAT1 signaling in cancer cells. Accordingly, intestinal myofibroblasts isolated from patients with ulcerative colitis or Crohn's disease, which are activated and produce high levels of TNFα, failed to stimulate STAT1 signaling in tumor cells, demonstrating that activated myofibroblasts lose the ability to trigger growth-inhibitory STAT1 signaling in tumor cells. Finally, we confirmed that silencing of STAT1 in tumor cells alters the crosstalk between tumor cells and fibroblasts, suggesting STAT1 as a novel link between intestinal inflammation and colon cancer. We demonstrated that normal fibroblasts restrain the growth of carcinoma cells, at least in part, through the induction of STAT1 signaling in cancer cells and showed that changes in the microenvironment, as they occur in inflammatory bowel disease, alter the crosstalk between carcinoma cells and fibroblasts, perturb the homeostasis of intestinal tissue, and thereby contribute to tumor progression.

4.
PLoS One ; 7(9): e45462, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23029025

RESUMEN

BACKGROUND: Tumor cells become addicted to both activated oncogenes and to proliferative and pro-survival signals provided by the abnormal tumor microenvironment. Although numerous soluble factors have been identified that shape the crosstalk between tumor cells and stroma, it has not been established how oncogenic mutations in the tumor cells alter their interaction with normal cells in the tumor microenvironment. PRINCIPAL FINDINGS: We showed that the isogenic HCT116 and Hke-3 cells, which differ only by the presence of the mutant kRas allele, both stimulate macrophages to produce IL1ß. In turn, macrophages enhanced Wnt signaling, proliferation and survival in both HCT116 and Hke-3 cells, demonstrating that signaling by oncogenic kRas in tumor cells does not impact their interaction with macrophages. HCT116 cells are heterozygous for ß-catenin (HCT116(WT/MT)), harboring one wild type (WT) and one mutant (MT) allele, but isogenic lines that carry only the WT (HCT116(WT)) or MT ß-catenin allele (HCT116(MT)) have been generated. We showed that macrophages promoted Wnt signaling in cells that carry the MT ß-catenin allele, but not in HCT116(WT) cells. Consistent with this observation, macrophages and IL1ß failed to stabilize Snail in HCT116(WT) cells, and to protect these cells from TRAIL-induced apoptosis. Finally, we demonstrated that HCT116 cells expressing dominant negative TCF4 (dnTCF4) or HCT116 cells with silenced Snail failed to stimulate IL1ß production in macrophages, demonstrating that tumor cells activate macrophages via a Wnt-dependent factor. SIGNIFICANCE: Our data demonstrate that oncogenic ß-catenin mutations in tumor cells, and subsequent activation of Wnt signaling, not only trigger cell-intrinsic alterations, but also have a significant impact on the crosstalk of tumor cells with the tumor associated macrophages.


Asunto(s)
Comunicación Celular/genética , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Macrófagos/metabolismo , Mutación , Factores de Transcripción/metabolismo , beta Catenina/genética , Alelos , Apoptosis , Línea Celular Tumoral , Neoplasias del Colon/inmunología , Genes ras , Células HCT116 , Humanos , Macrófagos/inmunología , Modelos Biológicos , Monocitos/metabolismo , Transducción de Señal , Factores de Transcripción de la Familia Snail , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Proteínas Wnt/metabolismo
5.
PLoS One ; 5(7): e11700, 2010 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-20661477

RESUMEN

BACKGROUND: We recently reported that colon tumor cells stimulate macrophages to release IL-1beta, which in turn inactivates GSK3beta and enhances Wnt signaling in colon cancer cells, generating a self-amplifying loop that promotes the growth of tumor cells. PRINCIPAL FINDINGS: Here we describe that macrophages protect HCT116 and Hke-3 colon cancer cells from TRAIL-induced apoptosis. Inactivation of IL-1beta by neutralizing IL-1beta antibody, or silencing of IL-1beta in macrophages inhibited their ability to counter TRAIL-induced apoptosis. Accordingly, IL-1beta was sufficient to inhibit TRAIL-induced apoptosis. TRAIL-induced collapse of the mitochondrial membrane potential (Delta psi) and activation of caspases were prevented by macrophages or by recombinant IL-1beta. Pharmacological inhibition of IL-1beta release from macrophages by vitamin D(3), a potent chemopreventive agent for colorectal cancer, restored the ability of TRAIL to induce apoptosis of tumor cells cultured with macrophages. Macrophages and IL-1beta failed to inhibit TRAIL-induced apoptosis in HCT116 cells expressing dnIkappaB, dnAKT or dnTCF4, confirming that they oppose TRAIL-induced cell death through induction of Wnt signaling in tumor cells. We showed that macrophages and IL-1beta stabilized Snail in tumor cells in an NF-kappaB/Wnt dependent manner and that Snail deficient tumor cells were not protected from TRAIL-induced apoptosis by macrophages or by IL-1beta, demonstrating a crucial role of Snail in the resistance of tumor cells to TRAIL. SIGNIFICANCE: We have identified a positive feedback loop between tumor cells and macrophages that propagates the growth and promotes the survival of colon cancer cells: tumor cells stimulate macrophages to secrete IL-1beta, which in turn, promotes Wnt signaling and stabilizes Snail in tumor cells, conferring resistance to TRAIL. Vitamin D(3) halts this amplifying loop by interfering with the release of IL-1beta from macrophages. Accordingly, vitamin D(3) sensitizes tumor cells to TRAIL-induced apoptosis, suggesting that the therapeutic efficacy of TRAIL could be augmented by this readily available chemopreventive agent.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Interleucina-1beta/metabolismo , Macrófagos/fisiología , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Factores de Transcripción/metabolismo , Western Blotting , Línea Celular , Línea Celular Tumoral , Colecalciferol/farmacología , Neoplasias del Colon/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Proteínas Wnt/metabolismo
6.
Cancer Microenviron ; 2(1): 69-80, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19779850

RESUMEN

Progression of colon cancer from microadenoma to macroscopic tumors is coupled to augmentation of canonical Wnt signaling. We recently reported that tumor associated macrophages, through interleukin 1ß (IL-1ß) dependent phosphorylation of GSK3ß, promote Wnt signaling in colon cancer cells, demonstrating that proinflammatory cytokines can enhance TCF4/ß-catenin transcriptional activity in tumor cells. Here we investigated the pathway whereby IL-1ß inactivates GSK3ß and promotes Wnt signaling in colon cancer cells. We showed that normal human monocytes, THP1 macrophages and IL-1 failed to induce Wnt signaling in tumor cells expressing dominant negative IκB (dnIκB), demonstrating that macrophages and IL-1 activate Wnt signaling in a NF-κB-dependent manner. NF-κB activity was required for macrophages and IL-1 to activate PDK1 and AKT in tumor cells and thereby inhibit GSK3ß activity. Consistently, dominant negative AKT (dnAKT), or pharmacological inhibition of AKT in tumor cells, prevented macrophage/IL-1 mediated phosphorylation of GSK3ß, activation of Wnt signaling, and induction of c-jun and c-myc, confirming that macrophages and IL-1 promote Wnt signaling in an AKT dependent manner. Finally, we showed IL-1 and macrophages failed to promote growth of colon cancer cells with impaired NF-κB or AKT signaling, confirming the requirement for NF-κB and AKT activation for the protumorigenic activity of tumor associated macrophages. Thus, we showed that IL-1 and tumor associated macrophages activate NF-κB-dependent PDK1/AKT signaling in tumor cells, and thereby inactivate GSK3ß, enhance Wnt signaling and promote growth of colon cancer cells, establishing a novel molecular link between inflammation and tumor growth.

7.
Exp Cell Res ; 314(7): 1507-18, 2008 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-18314102

RESUMEN

HDAC inhibitors exert potent anti-tumorigenic and anti-inflammatory activity. Their effects are selective for transformed cells, and we recently demonstrated that transformation of epithelial cells with k-Ras sensitizes cells to HDACi induced apoptosis. The aim of this study was to determine whether the ability of HDACi to modulate signaling by a major pro-inflammatory cytokine, TNFalpha, is also restricted to cells that harbor mutant k-Ras. We used the system of two isogenic cell lines that differ by the presence of mutant k-Ras, HCT116 and Hke3 cells. Treatment of cells with TNFalpha alone did not induce apoptosis; however HDACi potentiated TNFalpha-induced apoptosis in both HCT116 and Hke3 cells. Thus, the ability of HDACi to sensitize cells to TNFalpha-induced apoptosis appears to be k-Ras independent. We demonstrated that HDACi inhibited TNFalpha-induced NF-kappaB transcriptional and DNA binding activity in both cell lines, underlying the increased apoptosis in cells treated with both agents. We showed that overexpression of HDAC2 enhanced TNFalpha-induced NF-kappaB activity and that silencing of HDAC2 decreased NF-kappaB activity. Finally, silencing of HDAC2 expression was sufficient to sensitize colon cancer cells to TNFalpha-induced apoptosis. The ability of HDACi to interfere with NF-kappaB activity is likely to contribute to their potent anti-tumorigenic and anti-inflammatory activity.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Histona Desacetilasas/deficiencia , FN-kappa B/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/farmacología , Caspasa 9/metabolismo , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , ADN/metabolismo , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Silenciador del Gen/efectos de los fármacos , Células HCT116 , Histona Desacetilasa 2 , Inhibidores de Histona Desacetilasas , Humanos , FN-kappa B/genética , Unión Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteínas Represoras/antagonistas & inhibidores , Factor de Transcripción ReIA/metabolismo , Proteína bcl-X/genética , Proteínas ras/metabolismo
8.
Biochem Pharmacol ; 76(2): 198-207, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18561895

RESUMEN

Cucurbitacins have been shown to inhibit proliferation in a variety of cancer cell lines. The aim of this study was to determine their biological activity in colon cancer cell lines that do not harbor activated STAT3, the key target of cucurbitacin. In order to establish the role of activated kRas in the responsiveness of cells to cucurbitacins, we performed experiments in isogenic colon cancer cell lines, HCT116 and Hke-3, which differ only by the presence of an activated kRas allele. We compared the activity of 23, 24-dihydrocucurbitacin B (DHCB) and cucurbitacin R (CCR), two cucurbitacins that we recently isolated, with cucurbitacin I (CCI), a cucurbitacin with established antitumorigenic activity. We showed that cucurbitacins induced dramatic changes in the cytoskeleton (collapse of actin and bundling of tubulin microfilaments), inhibited proliferation and finally induced apoptosis of both HCT116 and Hke-3 cells. However, the presence of oncogenic kRas significantly decreased the sensitivity of cells to the three cucurbitacins tested, CCR, DHCB and CCI. We confirmed that mutational activation of kRas protects cells from cucurbitacin-induced apoptosis using nontransformed intestinal epithelial cells with inducible expression of kRasV12. Cucurbitacins induced the expression of p53 and p21 predominantly in HCT116 cells that harbor mutant Ras. Using HCT116 cells with targeted deletion of p53 or p21 we confirmed that p53 and p21 protect cells from apoptosis induced by cucurbitacins. These results demonstrated that sensitivity of human colon cancer cell lines to cucurbitacins depends on the kRas and p53/p21 status, and established that cucurbitacins can exert antitumorigenic activity in the absence of activated STAT3.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias del Colon/tratamiento farmacológico , Proteínas Proto-Oncogénicas/metabolismo , Triterpenos/farmacología , Proteínas ras/metabolismo , Apoptosis , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Humanos , Proteínas Proto-Oncogénicas p21(ras) , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda