Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 138
Filtrar
1.
Int Immunopharmacol ; 90: 107128, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33191180

RESUMEN

Sunitinib is a tyrosine kinase inhibitor for many tumors. Inflammation is one of the most important factors in the development of intestinal tumors. Many inflammation-related factors are regulated by tyrosine kinase receptors. It is reasonable to hypothesize that sunitinib can regulate the development of intestinal tumors by regulating the expression and/or activity of inflammation-related factors. Here, ApcMin/+ male mouse model was used to investigate the effect and mechanism of sunitinib malate against intestinal cancer. Results show that compared to vehicle, after sunitinib malate treatment, overall survival of ApcMin/+ mice was lengthened up to 25 days, with a gain of body weight, reduction of spleen/body weight index, and RBC, WBC and HGC regulated to normal levels of wild type mice, and a number of polyps no less than 1 mm significantly reduced. Meanwhile, in the intestines, the nuclear ß-Catenin protein and c-Myc mRNA were both down-regulated, and Bcl-6 was significantly reduced with Caspase-3 up regulated. Furthermore, inflammation-related factors including IL-6, TNF-α, IL-1α, IL-1ß and IFN-γ were down-regulated at mRNA levels in the intestines. These results suggest that sunitinib malate can significantly improve the survival status and inhibit intestinal tumor development in male ApcMin/+ mice, through inhibiting inflammation-related factors, while suppressing ß-cateinin/c-Myc pathway and re-balancing protein levels of Bcl-6 and Caspase-3.


Asunto(s)
Antiinflamatorios/farmacología , Anticarcinógenos/farmacología , Caspasa 3/metabolismo , Colon/efectos de los fármacos , Citocinas/metabolismo , Mediadores de Inflamación/metabolismo , Neoplasias Intestinales/prevención & control , Pólipos Intestinales/prevención & control , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Sunitinib/farmacología , beta Catenina/metabolismo , Animales , Colon/enzimología , Colon/patología , Citocinas/genética , Regulación de la Expresión Génica , Genes APC , Neoplasias Intestinales/enzimología , Neoplasias Intestinales/genética , Neoplasias Intestinales/patología , Pólipos Intestinales/enzimología , Pólipos Intestinales/genética , Pólipos Intestinales/patología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-myc/genética , Transducción de Señal
2.
Front Immunol ; 11: 581301, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33193386

RESUMEN

Murine and human invariant natural killer T (iNKT) lymphocytes are activated by α-galactosylceramide (α-GalCer) presented on CD1d. α-GalCer was first described as a lipid that had strong anti-metastatic effects in a mouse melanoma model, and it has subsequently been shown to induce efficient iNKT cell dependent tumor immunity in several tumor models. We have shown that α-GalCer treatment leads to a weak reduction of polyp burden in the autochthonous ApcMin/+ mouse model for human colon cancer, however this treatment resulted in upregulation of the inhibitory receptor PD-1 on iNKT cells. While anti-PD-1 treatment can prevent immune-suppression in other cancer types, human colon cancer is generally resistant to this treatment. Here we have used the ApcMin/+ model to investigate whether a combined treatment with α-GalCer and PD-1 blockade results in improved effects on polyp development. We find that PD-1 expression was high on T cells in polyps and lamina propria (LP) of ApcMin/+ mice compared to polyp free Apc+/+ littermates. Anti-PD-1 treatment alone promoted Tbet expression in iNKT cells and CD4 T cells, but did not significantly reduce polyp numbers. However, the combined treatment with anti-PD-1 and α-GalCer had synergistic effects, resulting in highly significant reduction of polyp numbers in the small and large intestine. Addition of PD-1 blockade to α-GalCer treatment prevented loss of iNKT cells that were skewed towards a TH1-like iNKT1 phenotype specifically in polyps. It also resulted in TH1 skewing and increased granzyme B expression of CD4 T cells. Taken together this demonstrates that a combination of immune stimulation targeting iNKT cells and checkpoint blockade may be a promising approach to develop for improved tumor immunotherapy.


Asunto(s)
Neoplasias del Colon/inmunología , Neoplasias del Colon/prevención & control , Galactosilceramidas/administración & dosificación , Células T Asesinas Naturales/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Proteína de la Poliposis Adenomatosa del Colon/deficiencia , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Anticuerpos Bloqueadores/administración & dosificación , Femenino , Humanos , Mucosa Intestinal/inmunología , Pólipos Intestinales/inmunología , Pólipos Intestinales/prevención & control , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/prevención & control , Receptor de Muerte Celular Programada 1/inmunología , Linfocitos T Reguladores/inmunología , Células TH1/inmunología
3.
Pharmacol Res ; 159: 104809, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32502642

RESUMEN

Microbiome therapy has attracted a keen interest from both research and business sectors. Our lab has been applying this "second genome" platform to assess the functionality of herbal medicines with fulfilling results. In this study, we applied this platform to assess the potential cancer-preventive effects of three selected adaptogenic plants. The flower buds from these plants were used to constitute Preparations SL and FSP according to the receipts of two commonly consumed Chinese medicinal decoctions for gastrointestinal discomfort. Preparation SL contains Sophorae japonica and Lonicerae Japonicae, and Preparation FSP contains Sophorae japonica and Gardenia Jasminoides. SL and FSP extracts significantly (p < 0.001) lowered the polyp burden, as well as the expressions of oncogenic signaling molecules, such as MAPK/ERK, PI3K/AKT, and STAT3 in ApcMin/+ mice. The inflamed gut was alleviated by shifting M1 to M2 macrophage phenotypes and the associated immune cytokines. The other remarkable change was on the extracellular tight junction protein complex, where the occludin, ZO-1, ICAM-1, E-cadherin were significantly (p < 0.05) upregulated while the N-cadherin and ß-catenin were downregulated in the treated mice. The above physiological changes in the gut epithelial barrier were companied with the changes in gut microbiome. The 16S Sequencing data revealed a marked decrease in the potential pathogens (especially Helicobacter species and hydrogen sulfide producing-bacteria) and the increase in beneficial bacteria (especially for species from the genera of Akkermansia, Barnesiella, Coprococcus, Lachnoclostridium, and Ruminococcus). The majority of which were the short-chain fatty acids (SCFAs) producers. Meanwhile SCFAs-sensing G protein-coupled receptors (GPCRs), including GPR41, GPR43, and GPR109a were also significantly upregulated. In a recent report, we proved that the bacteria-derived SCFAs plays an essential role to the anti-cancer effects of the mushroom polysaccharides and saponins in ApcMin/+ mice. In this study, we further demonstrated that butyrate treatment could enhance the extracellular tight junction protein complex as effective as the treatments with SL and FSP to the ApcMin/+ mice. Our findings provide strong evidence of the vital role of the SCFA-producers and their metabolites to the cancer-preventive properties of the SL and FSP preparations.


Asunto(s)
Anticarcinógenos/farmacología , Bacterias/efectos de los fármacos , Neoplasias Colorrectales/prevención & control , Ácidos Grasos/metabolismo , Flores/química , Microbioma Gastrointestinal , Mucosa Intestinal/efectos de los fármacos , Pólipos Intestinales/prevención & control , Magnoliopsida/química , Uniones Estrechas/efectos de los fármacos , Animales , Anticarcinógenos/aislamiento & purificación , Bacterias/metabolismo , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/microbiología , Citocinas/metabolismo , Gardenia/química , Genes APC , Interacciones Huésped-Patógeno , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Pólipos Intestinales/inmunología , Pólipos Intestinales/metabolismo , Pólipos Intestinales/microbiología , Lonicera/química , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Sophora/química , Proteínas de Uniones Estrechas/metabolismo , Uniones Estrechas/metabolismo
4.
Sci Rep ; 9(1): 14783, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31616028

RESUMEN

Inulin-rich foods exert a prebiotic effect, as this polysaccharide is able to enhance beneficial colon microbiota populations, giving rise to the in situ production of short-chain fatty acids (SCFAs) such as propionic and butyric acids. These SCFAs are potent preventive agents against colorectal cancer due to their histone deacetylases inhibitory properties, which induce apoptosis in tumor colonocytes. As colorectal cancer is the fourth most common neoplasia in Europe with 28.2 new cases per 100,000 inhabitants, a cost-effective preventive strategy has been tested in this work by redesigning common porcine meat products (chorizo sausages and cooked ham) consumed by a substantial proportion of the population towards potential colorectal cancer preventive functional foods. In order to test the preventive effect of these inulin-rich meat products against colorectal cancer, an animal model (Rattus norvegicus F344) was used, involving two doses of azoxymethane (10 mg/kg) and two treatments with dextran sodium sulfate (DSS) during a 20-week assay period. Control feed, control sausages, functional sausages (15.7% inulin), control cooked ham and functional cooked ham (10% inulin) were used to feed the corresponding animal cohorts. Then, the animals were sacrificed and their digestive tract tissues were analyzed. The results showed a statistically significant 49% reduction in the number of colon polyps in the functional meat products cohorts with respect to the control meat products animals, as well as an increase in the cecum weight (an indicator of a diet rich in prebiotic fiber), a 51.8% increase in colon propionate production, a 39.1% increase in colon butyrate concentrations, and a reduction in the number of hyperplastic Peyer's patches. Metagenomics studies also demonstrated colon microbiota differences, revealing a significant increase in Bacteroidetes populations in the functional meat products (mainly due to an increase in Bacteroidaceae and Prevotellaceae families, which include prominent propionate producers), together with a reduction in Firmicutes (especially due to lower Lachnospiraceae populations). However, functional meat products showed a remarkable increase in the anti-inflammatory and fiber-fermentative Blautia genus, which belongs to this Lachnospiraceae family. The functional meat products cohorts also presented a reduction in important pro-inflammatory bacterial populations, such as those of the genus Desulfovibrio and Bilophila. These results were corroborated in a genetic animal model of CRC (F344/NSlc-Apc1588/kyo) that produced similar results. Therefore, processed meat products can be redesigned towards functional prebiotic foods of interest as a cost-effective dietary strategy for preventing colorectal cancer in human populations.


Asunto(s)
Neoplasias Colorrectales/prevención & control , Alimentos Funcionales , Pólipos Intestinales/prevención & control , Inulina/administración & dosificación , Productos de la Carne , Neoplasias Experimentales/prevención & control , Animales , Azoximetano/toxicidad , Colon/metabolismo , Colon/microbiología , Colon/patología , Neoplasias Colorrectales/inducido químicamente , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/microbiología , Sulfato de Dextran/toxicidad , Fibras de la Dieta/administración & dosificación , Ácidos Grasos Volátiles/biosíntesis , Microbioma Gastrointestinal/genética , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Pólipos Intestinales/inducido químicamente , Pólipos Intestinales/genética , Pólipos Intestinales/microbiología , Masculino , Metagenómica , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/genética , Neoplasias Experimentales/microbiología , Prebióticos/administración & dosificación , Ratas , Porcinos
5.
Oncologist ; 23(4): 399-e33, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29371475

RESUMEN

LESSONS LEARNED: Motivating patients to enroll in chemopreventive studies is challenging.Chemoprevention with toxic drugs is not feasible. BACKGROUND: LKB1 mutations are the underlying genetic abnormality causing Peutz-Jeghers syndrome (PJS) and are a potential target for everolimus. In this phase II study, the efficacy of everolimus on polyp and tumor growth in PJS patients was investigated. METHODS: Adult patients with a proven LKB1 mutation and who were suitable for everolimus treatment were included in two different PJS cohorts: (a) patients with unresectable malignancies and (b) patients with high-risk polyps. Treatment in both groups was oral everolimus, 10 mg daily. Response rates were primary endpoints for both cohorts. RESULTS: Between October 2011 and April 2016, only two patients were enrolled, one in each cohort. A 49-year-old patient with advanced pancreatic cancer in cohort 1 was progressive after 2 months. A 52-year-old male patient in cohort 2 experienced severe toxicity and refused treatment after 4 months, even though endoscopy suggested stabilization of polyps. Adverse events included dental inflammations, mucositis, and rash. In 2016, the trial was aborted for lack of accrual, despite extensive accrual efforts in an area where PJS is highly prevalent and care is highly centralized. CONCLUSION: Due to accrual problems, no conclusions can be drawn about the value of everolimus in PJS treatment, questioning the feasibility of this agent for chemoprevention.


Asunto(s)
Antineoplásicos/uso terapéutico , Everolimus/uso terapéutico , Pólipos Intestinales/prevención & control , Síndrome de Peutz-Jeghers/tratamiento farmacológico , Síndrome de Peutz-Jeghers/prevención & control , Quinasas de la Proteína-Quinasa Activada por el AMP , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Quimioprevención , Everolimus/administración & dosificación , Everolimus/efectos adversos , Humanos , Pólipos Intestinales/tratamiento farmacológico , Masculino , Persona de Mediana Edad , Mutación , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Síndrome de Peutz-Jeghers/genética , Síndrome de Peutz-Jeghers/patología , Proteínas Serina-Treonina Quinasas/genética
6.
Curr Cancer Drug Targets ; 18(9): 905-911, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28786349

RESUMEN

BACKGROUND: Recent epidemiological and experimental studies have shown that obesity is a major risk factor for Colorectal Cancer (CRC). Regular intake of high fat-containing diet can promote obesity and metabolic syndrome by increasing the insulin resistance and inflammatory response which contribute to carcinogenesis. Previously, we have shown that inhibition of polyol pathway enzyme aldose reductase (AR) prevents carcinogens- and inflammatory growth factorsinduced CRC. However, the effect of AR inhibition on a high-fat diet (HFD)-induced formation of intestinal polyps in Apc-deficient Min (multiple intestinal neoplasia; ApcMin/+) mice is not known. METHODS: We examined the effect of AR inhibitor, fidarestat on the HFD-induced formation of preneoplastic intestinal polyps in ApcMin/+ mice which is an excellent model of colon cancer. RESULTS: APCMin/+ mice fed for 12 weeks of HFD caused a significant increase in the formation of polyps in the small and large intestines and fidarestat given along with the HFD prevented the number of intestinal polyps. Fidarestat also decreased the size of the polyps in the intestines of HFDtreated APC Min mice. Further, the expression levels of beta-catenin, PCNA, PKC-ß2, P-AKT, Pp65, COX-2, and iNOS in the small and large intestines of HFD-treated mice significantly increased, and AR inhibitor prevented it. CONCLUSION: Our results thus suggest that fidarestat could be used as a potential chemopreventive drug for intestinal cancers due to APC gene mutations.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/fisiología , Aldehído Reductasa/antagonistas & inhibidores , Neoplasias del Colon/prevención & control , Dieta Alta en Grasa/efectos adversos , Inhibidores Enzimáticos/farmacología , Imidazolidinas/farmacología , Pólipos Intestinales/prevención & control , Animales , Neoplasias del Colon/enzimología , Neoplasias del Colon/etiología , Femenino , Pólipos Intestinales/enzimología , Pólipos Intestinales/etiología , Masculino , Ratones , Ratones Endogámicos C57BL
7.
Nucleic Acids Res ; 45(21): 12256-12269, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-28981717

RESUMEN

We report here a robust, tunable, and reversible transcription control system for endogenous genes. The REMOTE-control system (Reversible Manipulation of Transcription at Endogenous loci) employs enhanced lac repression and tet activation systems. With this approach, we show in mouse embryonic stem cells that endogenous Dnmt1 gene transcription could be up- or downregulated in a tunable, inducible, and reversible manner across nearly two orders of magnitude. Transcriptional repression of Dnmt1 by REMOTE-control was potent enough to cause embryonic lethality in mice, reminiscent of a genetic knockout of Dnmt1 and could substantially suppress intestinal polyp formation when applied to an ApcMin model. Binding by the enhanced lac repressor was sufficiently tight to allow strong attenuation of transcriptional elongation, even at operators located many kilobases downstream of the transcription start site and to produce invariably tight repression of all of the strong viral/mammalian promoters tested. Our approach of targeting tet transcriptional activators to the endogenous Dnmt1 promoter resulted in robust upregulation of this highly expressed housekeeping gene. Our system provides exquisite control of the level, timing, and cell-type specificity of endogenous gene expression, and the potency and versatility of the system will enable high resolution in vivo functional analyses.


Asunto(s)
Regulación de la Expresión Génica , Animales , ADN (Citosina-5-)-Metiltransferasa 1/genética , Células Madre Embrionarias/metabolismo , Ingeniería Genética , Pólipos Intestinales/genética , Pólipos Intestinales/prevención & control , Represoras Lac/metabolismo , Ratones , Células 3T3 NIH , Regiones Promotoras Genéticas , Elongación de la Transcripción Genética
8.
Rev Esp Enferm Dig ; 109(12): 875, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29072082

RESUMEN

Chemoprevention is a practical approach to reduce the risk of various cancers including colorectal cancer (CRC). The goal is to reduce the incidence of pre-neoplastic adenomatous polyps and prevent its progression to CRC. Curcumin and silibinin prevent intestinal polyp formation in mice. Curcumin sensitizes silymarin to exert synergistic anticancer activity in colon cancer cells. Patients presenting with multiple colorectal adenomatous polyps (MCRA) have a high lifetime risk for CRC. We present a 57-year-old man with MCRA, without deleterious germline APC or MYH mutations. Our patient had 54 polyps in the first colonoscopy, most of 3 to 8 mm and one of 20 mm with high grade dysplasia / adenocarcinoma. Four subsequent colonoscopies showed continuous development of adenomatous polyps treated by polypectomy for the most part and some with heat. After the treatment with curcumin for 3 months and a half followed by silibinin for 9 months, we find many less polyps than in the previous colonoscopies, going from the finding of 40 adenomas of 3-6 mm in the pre-treatment colonoscopy to 3 polyps after treatment.


Asunto(s)
Antineoplásicos/uso terapéutico , Quimioprevención/métodos , Neoplasias Colorrectales/prevención & control , Curcumina/uso terapéutico , Pólipos Intestinales/prevención & control , Silimarina/uso terapéutico , Neoplasias Colorrectales/cirugía , Quimioterapia Combinada , Humanos , Pólipos Intestinales/cirugía , Masculino , Persona de Mediana Edad , Recurrencia , Silibina
9.
J Appl Microbiol ; 122(3): 770-784, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28004480

RESUMEN

AIMS: To investigate the in vivo effects of Lactobacillus rhamnosus GG (LGG) on intestinal polyp development and the interaction between this single-organism probiotic and the gut microbiota therein. METHODS AND RESULTS: The ApcMin/+ mouse model was used to study the potential preventive effect of LGG on intestinal polyposis, while shotgun metagenomic sequencing was employed to characterize both taxonomic and functional changes within the gut microbial community. We found that the progression of intestinal polyps in the control group altered the community functional profile remarkably despite small variation in the taxonomic diversity. In comparison, the consumption of LGG helped maintain the overall functional potential and taxonomic profile in the resident microbes, thereby leading to a 25% decrease of total polyp counts. Furthermore, we found that LGG enriched those microbes or microbial activities related to short-chain fatty acid production (e.g. Roseburia and Coprococcus), as well as suppressed the ones that can lead to inflammation (e.g. Bilophila wadsworthia). CONCLUSIONS: Our study using shotgun metagenomics highlights how single probiotic LGG may exert its beneficial effects and decrease polyp formation in mice by maintaining gut microbial functionality. SIGNIFICANCE AND IMPACT OF THE STUDY: This probiotic intervention targeting microbiota may be used in conjugation with other dietary supplements or drugs as part of prevention strategies for early-stage colon cancer, after further clinical validations in human.


Asunto(s)
Pólipos Intestinales/prevención & control , Lacticaseibacillus rhamnosus/crecimiento & desarrollo , Microbiota/efectos de los fármacos , Probióticos/uso terapéutico , Sulindac/uso terapéutico , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Humanos , Metagenómica/métodos , Ratones , Filogenia , Probióticos/farmacología , Organismos Libres de Patógenos Específicos , Sulindac/farmacología
10.
PLoS One ; 11(2): e0147289, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26910063

RESUMEN

Nonsteroidal anti-inflammatory drugs have been shown to reduce the incidence of gastrointestinal cancers, but the propensity of these drugs to cause ulcers and bleeding limits their use. H2S has been shown to be a powerful cytoprotective and anti-inflammatory substance in the digestive system. This study explored the possibility that a H2S-releasing nonsteroidal anti-inflammatory drug (ATB-346) would be effective in a murine model of hereditary intestinal cancer (APCMin+ mouse) and investigated potential mechanisms of action via transcriptomics analysis. Daily treatment with ATB-346 was significantly more effective at preventing intestinal polyp formation than naproxen. Significant beneficial effects were seen with a treatment period of only 3-7 days, and reversal of existing polyps was observed in the colon. ATB-346, but not naproxen, significantly decreased expression of intestinal cancer-associated signaling molecules (cMyc, ß-catenin). Transcriptomic analysis identified 20 genes that were up-regulated in APCMin+ mice, 18 of which were reduced to wild-type levels by one week of treatment with ATB-346. ATB-346 is a novel, gastrointestinal-sparing anti-inflammatory drug that potently and rapidly prevents and reverses the development of pre-cancerous lesions in a mouse model of hereditary intestinal tumorigenesis. These effects may be related to the combined effects of suppression of cyclooxygenase and release of H2S, and correction of most of the APCMin+-associated alterations in the transcriptome. ATB-346 may represent a promising agent for chemoprevention of tumorigenesis in the GI tract and elsewhere.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Carcinogénesis/efectos de los fármacos , Sulfuro de Hidrógeno/química , Neoplasias Intestinales/patología , Neoplasias Intestinales/prevención & control , Naproxeno/análogos & derivados , Animales , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/uso terapéutico , Quimioprevención , Colon/efectos de los fármacos , Colon/metabolismo , Colon/patología , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Pólipos Intestinales/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Naproxeno/química , Naproxeno/uso terapéutico , Proteínas Proto-Oncogénicas c-myc/metabolismo , beta Catenina/metabolismo
11.
Oncotarget ; 7(8): 8640-52, 2016 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-26840084

RESUMEN

This study aimed to identify gastric mucosal protectants that suppress intestinal tumorigenesis in a mouse model. We chose six gastric mucosal protectants (ecabet sodium hydrate, irsogladine maleate, rebamipide, sofalcone, teprenone and troxipide) and examined their effects on the activity of oxidative stress-related transcriptional factors, including AP-1, NF-jB, NRF2, p53 and STAT3, in Caco-2 cells using a luciferase reporter gene assay. Among the six protectants, irsogladine maleate clearly inhibited NF-jB and AP-1 transcriptional activity. Furthermore, the chemopreventive property of irsogladine maleate was examined in a Min mouse model of familial adenomatous polyposis. Treatment with irsogladine maleate at doses of 5 and 50 ppm significantly reduced the number of intestinal polyps to 69% and 66% of the untreated control value, respectively. In these polyps, mRNA levels of the downstream targets of NF-jB, such as IL-1ß and IL-6, were decreased by irsogladine maleate treatment. Moreover, the levels of oxidative stress-related markers, reactive carbonyl species, in the livers of Min mice were clearly decreased following the administration of irsogladine maleate. This study demonstrated that irsogladine maleate suppresses intestinal polyp formation in Min mice partly through the NF-jB signaling pathway, thus reducing oxidative stress.


Asunto(s)
Antiulcerosos/farmacología , Mucosa Gástrica/efectos de los fármacos , Genes APC/fisiología , Pólipos Intestinales/prevención & control , Mutación/genética , Triazinas/farmacología , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patología , Humanos , Técnicas para Inmunoenzimas , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Pólipos Intestinales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos
12.
J Cancer Res Clin Oncol ; 142(5): 913-25, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26702935

RESUMEN

PURPOSE: Naringin is a natural dietary flavonoid compound. We aimed to evaluate the effects of naringin on intestinal tumorigenesis in the adenomatous polyposis coli multiple intestinal neoplasia (Apc (Min/+)) mouse model. METHODS: Apc (Min/+) mice were given either naringin (150 mg/kg) or vehicle by p.o. gavage daily for 12 consecutive weeks. Mice were killed with ether, and blood samples were collected to assess the concentrations of IL-6 and PGE2. Total intestines were removed, and the number of polyps was examined. Tissue samples of intestinal polyps were subjected to the assays of histopathology, immunohistochemical analysis and Western blotting analysis. RESULTS: Apc (Min/+) mice fed with naringin developed less and smaller polyps in total intestines. Naringin prevented intestinal tumorigenesis without adverse effects. Histopathologic analysis revealed the reduction of dysplastic cells and dysplasia in the adenomatous polyps. The treatments' effects might arise from its anti-proliferation, induction of apoptosis and modulation of GSK-3ß and APC/ß-catenin signaling pathways. Naringin also exerted its effects on tumorigenesis through anti-chronic inflammation. CONCLUSION: Naringin prevented intestinal tumorigenesis likely through a collection of activities including anti-proliferation, induction of apoptosis, modulation of GSK-3ß and APC/ß-catenin pathways and anti-inflammation. Naringin is a potential chemopreventive agent for reducing the risk of colonic cancers.


Asunto(s)
Transformación Celular Neoplásica/efectos de los fármacos , Suplementos Dietéticos , Modelos Animales de Enfermedad , Flavanonas/farmacología , Neoplasias Intestinales/prevención & control , Pólipos Intestinales/prevención & control , Proteína de la Poliposis Adenomatosa del Colon/fisiología , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Citocinas/metabolismo , Femenino , Humanos , Técnicas para Inmunoenzimas , Mediadores de Inflamación/metabolismo , Neoplasias Intestinales/metabolismo , Neoplasias Intestinales/patología , Pólipos Intestinales/metabolismo , Pólipos Intestinales/patología , Ratones , Ratones Endogámicos C57BL , Células Tumorales Cultivadas
13.
Nat Med ; 21(8): 906-13, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26107252

RESUMEN

The inflammasome activates caspase-1 and the release of interleukin-1ß (IL-1ß) and IL-18, and several inflammasomes protect against intestinal inflammation and colitis-associated colon cancer (CAC) in animal models. The absent in melanoma 2 (AIM2) inflammasome is activated by double-stranded DNA, and AIM2 expression is reduced in several types of cancer, but the mechanism by which AIM2 restricts tumor growth remains unclear. We found that Aim2-deficient mice had greater tumor load than Asc-deficient mice in the azoxymethane/dextran sodium sulfate (AOM/DSS) model of colorectal cancer. Tumor burden was also higher in Aim2(-/-)/Apc(Min/+) than in APC(Min/+) mice. The effects of AIM2 on CAC were independent of inflammasome activation and IL-1ß and were primarily mediated by a non-bone marrow source of AIM2. In resting cells, AIM2 physically interacted with and limited activation of DNA-dependent protein kinase (DNA-PK), a PI3K-related family member that promotes Akt phosphorylation, whereas loss of AIM2 promoted DNA-PK-mediated Akt activation. AIM2 reduced Akt activation and tumor burden in colorectal cancer models, while an Akt inhibitor reduced tumor load in Aim2(-/-) mice. These findings suggest that Akt inhibitors could be used to treat AIM2-deficient human cancers.


Asunto(s)
Neoplasias del Colon/prevención & control , Proteína Quinasa Activada por ADN/fisiología , Proteínas de Unión al ADN/fisiología , Inflamasomas/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Animales , Colitis/complicaciones , Femenino , Células HCT116 , Humanos , Pólipos Intestinales/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación
14.
Oncol Rep ; 33(4): 1947-55, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25672292

RESUMEN

Non-steroidal anti-inflammatory drugs (NSAIDs) have been suggested as the potential new class of preventive or therapeutic antitumor agents. The aim of the present study was to evaluate the antitumor activity of the novel NSAID, CG100649. CG100649 is a novel NSAID dual inhibitor for COX-2 and carbonic anhydrase (CA)-I/-II. In the present study, we investigated the alternative mechanism by which CG100649 mediated suppression of the colon cancer growth and development. The anchorage­dependent and -independent clonogenic assay showed that CG100649 inhibited the clonogenicity of human colon cancer cells. The flow cytometric analysis showed that CG100649 induced the G2/M cell cycle arrest in colon cancer cells. Animal studies showed that CG100649 inhibited the tumor growth in colon cancer xenograft in nude mice. Furthermore, quantitative PCR and FACS analysis demonstrated that CG100649 upregulated the expression of TNF-related apoptosis-inducing ligand (TRAIL) receptors (DR4 and DR5) but decreased the expression of decoy receptors (DcR1 and DcR2) in colon cancer cells. The results showed that CG100649 treatment sensitized TRAIL­mediated growth suppression and apoptotic cell death. The combination treatment resulted in significant repression of the intestinal polyp formation in APCmin/+ mice. Our data clearly demonstrated that CG100649 contains preventive and therapeutic activity for colon cancer. The present study may be useful for identification of the potential benefit of the NSAID CG100649, for the achievement of a better treatment response in colon cancer.


Asunto(s)
Anticarcinógenos/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias del Colon/tratamiento farmacológico , Furanos/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Sulfonamidas/uso terapéutico , Ligando Inductor de Apoptosis Relacionado con TNF/uso terapéutico , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Anticarcinógenos/farmacología , Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Neoplasias del Colon/patología , Sinergismo Farmacológico , Femenino , Furanos/administración & dosificación , Furanos/farmacología , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Proteínas Ligadas a GPI/biosíntesis , Proteínas Ligadas a GPI/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Pólipos Intestinales/prevención & control , Intestino Delgado/patología , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/biosíntesis , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Miembro 10c de Receptores del Factor de Necrosis Tumoral/biosíntesis , Miembro 10c de Receptores del Factor de Necrosis Tumoral/genética , Organismos Libres de Patógenos Específicos , Sulfonamidas/administración & dosificación , Sulfonamidas/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/administración & dosificación , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Receptores Señuelo del Factor de Necrosis Tumoral/biosíntesis , Receptores Señuelo del Factor de Necrosis Tumoral/genética , Ensayo de Tumor de Célula Madre , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Carcinogenesis ; 35(7): 1613-9, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24632492

RESUMEN

The promotion and progression of carcinogenesis are susceptible to nutritional interventions aimed at counteracting cancer development. Lipid metabolism is essential in the onset and progression of tumors and for cancer cell survival. In this study, we tested the effects of diets enriched with natural compounds, such as olive oil and salmon oil, in mice that spontaneously develop intestinal polyps (Apc(Min/+) mice). For this purpose, we evaluated polyp number and volume, intestinal mucosa proliferation/apoptosis, estrogen receptors (ERs) expression, fatty acid synthase and 3-hydroxy-3-methylglutaryl coenzyme A (HMGCoA) reductase gene expression and enzymatic activity. Compared with the standard diet, the salmon oil-enriched diet, containing a high percentage of omega-3 polyunsaturated fatty acids, and, to a lesser extent, olive oil-enriched diet reduced polyp number and volume through a reduction of proliferation and a marked proapoptotic effect. These biological effects were mediated by an inhibition of fatty acid synthase and HMGCoA reductase gene expression and activity and an increase of ERß/ERα ratio. Our findings suggest that a proper dietary lifestyle could contribute to primary cancer prevention.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/fisiología , Apoptosis , Neoplasias del Colon/prevención & control , Ácidos Grasos Omega-3/uso terapéutico , Pólipos Intestinales/prevención & control , Aceites de Plantas/administración & dosificación , Animales , Western Blotting , Proliferación Celular , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Hidroximetilglutaril-CoA Reductasas/genética , Hidroximetilglutaril-CoA Reductasas/metabolismo , Mucosa Intestinal/metabolismo , Pólipos Intestinales/metabolismo , Pólipos Intestinales/patología , Metabolismo de los Lípidos , Masculino , Ratones , Ratones Endogámicos C57BL , Microsomas/enzimología , Aceite de Oliva , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Receptor fas/genética , Receptor fas/metabolismo
16.
Asian Pac J Cancer Prev ; 14(7): 4435-40, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23992016

RESUMEN

Aloe vera gel supercritical CO2 extract (AVGE) has been shown to contain five phytosterols, reduce visceral fat accumulation, and influence the metabolism of glucose and lipids in animal model experiments. Recent epidemiologic studies have shown that obesity is an established risk factor for several cancers including colorectal cancer. Therefore, we examined the effects of AVGE on intestinal polyp formation in Apc-deficient Min mice fed a high-fat diet. Male Min mice were divided into normal diet (ND), high fat diet (HFD), low dose AVGE (HFD+LAVGE) and high dose AVGE (HFD+HAVGE) groups. The ND group received AIN-93G diet and the latter 3 groups were given modified high-fat AIN-93G diet (HFD) for 7 weeks. AVGE was suspended in 0.5% carboxymethyl cellulose (CMC) and administered orally to mice in HFD+LAVGE and HFD+HAVGE groups every day (except on Sunday) for 7 weeks at a dose of 3.75 and 12.5 mg/kg body weight, respectively. ND and HFD groups received 0.5% CMC alone. Between weeks 4 and 7, body weights in the HFD and HFD+LAVGE groups were reduced more than those in the ND group. However, body weights were not reduced in the HFD+HAVGE group. Mice were sacrificed at the end of the experiment and their intestines were scored for polyps. No significant differences were observed in either the incidence and multiplicity of intestinal polyps (≥0.5 mm in a diameter) among the three groups fed HFD. However, when intestinal polyps were categorized by their size into 0.5-1.4, 1.5-2.4, or ≥2.5 mm, the incidence and multiplicity of large polyps (≥2.5 mm) in the intestine in the HFD+HAVGE group were significantly lower than those in the HFD group. We measured plasma lipid (triglycerides and total cholesterol) and adipocytokine [interleukin-6 and high molecular weight (HMW) adiponectin] levels as possible indicators of mechanisms of inhibition. The results showed that HMW adiponectin levels in the HFD group were significantly lower than those in the ND group. However, the levels in the HFD+HAVGE group were significantly higher than those in the HFD group. These results indicate that HAVGE reduced large-sized intestinal polyps and ameliorated reduction in plasma HMW adiponectin levels in Min mice fed HFD.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/fisiología , Aloe/química , Dieta Alta en Grasa , Pólipos Intestinales/prevención & control , Obesidad/prevención & control , Extractos Vegetales/farmacología , Adiponectina/sangre , Animales , Peso Corporal/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Lípidos/sangre , Masculino , Ratones , Ratones Endogámicos C57BL
17.
World J Gastroenterol ; 19(34): 5671-7, 2013 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-24039360

RESUMEN

AIM: To evaluate if 3 mo oral supplementation with Eviendep® was able to reduce the number of duodenal polyps in familial adenomatous polyposis (FAP) patients with ileal pouch-anal anastomosis (IPAA). METHODS: Eleven FAP patients with IPAA and duodenal polyps were enrolled. They underwent upper gastrointestinal (GI) endoscopy at the baseline and after 3 mo of treatment. Each patient received 5 mg Eviendep twice a day, at breakfast and dinner time, for 3 mo. Two endoscopists evaluated in a blinded manner the number and size of duodenal polyps. Upper GI endoscopies with biopsies were performed at the baseline (T0) with the assessment of the Spigelman score. Polyps > 10 mm were removed during endoscopy and at the end of the procedure a new Spigelman score was determined (T1). The procedure was repeated 3 mo after the baseline (T2). Four photograms were examined for each patient, at T1 and T2. The examined area was divided into 3 segments: duodenal bulb, second and third portion duodenum. Biopsy specimens were taken from all polyps > 10 mm and from all suspicious ones, defined by the presence of a central depression, irregular surface, or irregular vascular pattern. Histology was classified according to the updated Vienna criteria. RESULTS: At baseline the mean number of duodenal detected polyps was 27.7 and mean sizes were 15.8 mm; the mean Spigelman score was 7.1. After polypectomy the mean number of duodenal detected polyps was 25.7 and mean sizes were 7.6 mm; the mean Spigelman score was 6.4. After 3 mo of Eviendep bid, all patients showed a reduction of number and size of duodenal polyps. The mean number of duodenal polyps was 8 (P = 0.021) and mean size was 4.4 mm; the mean Spigelman score was 6.6. Interrater agreement was measured. Lesions > 1 cm found a very good degree of concordance (kappa 0.851) and a good concordance was as well encountered for smaller lesions (kappa 0.641). CONCLUSION: Our study demonstrated that short-term (90 d) supplementation with Eviendep® in FAP patients with IPAA and with recurrent adenomas in the duodenal mucosa, resulted effective in reducing polyps number of 32% and size of 51%.


Asunto(s)
Poliposis Adenomatosa del Colon/tratamiento farmacológico , Enfermedades Duodenales/prevención & control , Pólipos Intestinales/prevención & control , Fitoestrógenos/uso terapéutico , Fitoterapia , Poliposis Adenomatosa del Colon/dietoterapia , Poliposis Adenomatosa del Colon/cirugía , Adolescente , Adulto , Anastomosis Quirúrgica , Reservorios Cólicos , Fibras de la Dieta/uso terapéutico , Femenino , Humanos , Masculino , Extractos Vegetales/uso terapéutico , Adulto Joven
18.
Cancer Prev Res (Phila) ; 6(9): 908-16, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23852423

RESUMEN

The occurrence of intestinal polyps in people at high risk for developing colorectal cancer provides an opportunity to test the efficacy of chemoprevention agents. In this situation of treating otherwise healthy people, the potential for toxicity must be minimal. The small-molecule flexible heteroarotinoid (Flex-Het), called SHetA2, has chemoprevention activity in organotypic cultures in vitro and lack of toxicity at doses capable of inhibiting xenograft tumor growth in vivo. The objective of this study was to evaluate SHetA2 chemoprevention activity and toxicity in the APC(min/+) murine model. Oral administration of SHetA2 at 30 and 60 mg/kg five days per week for 12 weeks significantly reduced development of intestinal polyps by 40% to 60% depending on the dose and sex of the treatment group. Immunohistochemical and Western blot analysis of polyps showed reduced levels of cyclin D1 and proliferating cell nuclear antigen in both SHetA2 treatment groups. Western blot analysis also showed SHetA2 induction of E-cadherin, Bax, and caspase-3 cleavage along with reduction in Bcl-2, COX-2, and VEGF, consistent with SHetA2 regulation of apoptosis, inflammation, and angiogenesis. Neither dose caused weight loss nor gross toxicity in APC(min/+) or wild-type littermates. Magnetic resonance imaging (MRI) of cardiac function showed no evidence of SHetA2 toxicity. SHetA2 did not alter left ventricular wall thickness. In summary, SHetA2 exerts chemoprevention activity without overt or cardiac toxicity in the APC(min/+) model. SHetA2 modulation of biomarkers in colon polyps identifies potential pharmacodynamic endpoints for SHetA2 clinical trials.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Transformación Celular Neoplásica/efectos de los fármacos , Cromanos/uso terapéutico , Neoplasias del Colon/prevención & control , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/prevención & control , Neoplasias Intestinales/prevención & control , Intestino Delgado/efectos de los fármacos , Tionas/uso terapéutico , Administración Oral , Animales , Biomarcadores/análisis , Transformación Celular Neoplásica/patología , Neoplasias del Colon/etiología , Neoplasias del Colon/patología , Femenino , Cardiopatías/etiología , Cardiopatías/patología , Cardiopatías/prevención & control , Inflamación/etiología , Inflamación/patología , Inflamación/prevención & control , Neoplasias Intestinales/etiología , Neoplasias Intestinales/patología , Pólipos Intestinales/etiología , Pólipos Intestinales/patología , Pólipos Intestinales/prevención & control , Intestino Delgado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/prevención & control
19.
Am J Surg ; 205(5): 618-22; discussion 622, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23592173

RESUMEN

BACKGROUND: National guidelines put forth by the American Cancer Society, the US Multi-Society Task Force on Colorectal Cancer, and the American College of Gastroenterology provide recommendations regarding colorectal cancer screening and follow-up surveillance. Practice patterns may differ from these guidelines. This study analyzes the concordance between a tertiary equal access system and national guidelines for colorectal cancer and polyp surveillance. METHODS: We performed a retrospective database review of all patients at a single institution undergoing screening colonoscopy from 2010 to 2011. Patient demographics, indication for colonoscopy, pathologic findings, and follow-up recommendations documented by the provider were analyzed. Multivariate analysis was performed in an attempt to identify predictors of discordant recommendations. RESULTS: One thousand four hundred twenty patients were identified (mean age, 54.3 ± 7.7 years, 48.6% women). The gastroenterology service performed the majority of colonoscopies (87.2%) compared with the surgery service (11.6%). The major indications were routine screening (84.4%) and a strong family history of colorectal cancer (12.2%). The adenoma detection rate for the entire cohort was 27.4%. Other pathologic conditions identified included hyperplastic polyps (16%), lymphoid aggregates (3.5%), and invasive adenocarcinoma (0.1%). Overall, follow-up recommendations correlated with established guidelines in 97% of cases. By multivariate analysis, only the final pathologic finding of lymphoid aggregates was associated with discordant recommendations (odds ratio [OR], 4.62; 95% confidence interval [CI], 1.64 to 12.99; P = .004). When comparing discordant recommendations between specialties, there was a statistically significant difference between gastroenterology (1.6%) and surgery (7.6%) (P < .0001) providers; surgeons trended toward recommending earlier follow-up examinations (P = .37). CONCLUSIONS: Overall, surveillance recommendations correlated well with current national guidelines. Concordance rates were higher with gastroenterologists in this cohort. Alterations based on final pathologic examination and individual cases remain clinically important.


Asunto(s)
Colonoscopía/normas , Neoplasias Colorrectales/diagnóstico , Detección Precoz del Cáncer/normas , Adhesión a Directriz/estadística & datos numéricos , Pautas de la Práctica en Medicina/estadística & datos numéricos , Adenocarcinoma/diagnóstico , Adenocarcinoma/prevención & control , Adenoma/diagnóstico , Adenoma/prevención & control , Adulto , Anciano , Anciano de 80 o más Años , Colonoscopía/estadística & datos numéricos , Neoplasias Colorrectales/prevención & control , Cirugía Colorrectal/normas , Cirugía Colorrectal/estadística & datos numéricos , Detección Precoz del Cáncer/estadística & datos numéricos , Femenino , Estudios de Seguimiento , Gastroenterología/normas , Gastroenterología/estadística & datos numéricos , Humanos , Pólipos Intestinales/diagnóstico , Pólipos Intestinales/prevención & control , Masculino , Persona de Mediana Edad , Análisis Multivariante , Guías de Práctica Clínica como Asunto , Estudios Retrospectivos , Washingtón
20.
PLoS One ; 8(3): e59392, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23527181

RESUMEN

Chemoprevention is a pragmatic approach to reduce the risk of colorectal cancer, one of the leading causes of cancer-related death in western countries. In this regard, maslinic acid (MA), a pentacyclic triterpene extracted from wax-like coatings of olives, is known to inhibit proliferation and induce apoptosis in colon cancer cell lines without affecting normal intestinal cells. The present study evaluated the chemopreventive efficacy and associated mechanisms of maslinic acid treatment on spontaneous intestinal tumorigenesis in Apc(Min/+) mice. Twenty-two mice were randomized into 2 groups: control group and MA group, fed with a maslinic acid-supplemented diet for six weeks. MA treatment reduced total intestinal polyp formation by 45% (P<0.01). Putative molecular mechanisms associated with suppressing intestinal polyposis in Apc(Min/+) mice were investigated by comparing microarray expression profiles of MA-treated and control mice and by analyzing the serum metabolic profile using NMR techniques. The different expression phenotype induced by MA suggested that it exerts its chemopreventive action mainly by inhibiting cell-survival signaling and inflammation. These changes eventually induce G1-phase cell cycle arrest and apoptosis. Moreover, the metabolic changes induced by MA treatment were associated with a protective profile against intestinal tumorigenesis. These results show the efficacy and underlying mechanisms of MA against intestinal tumor development in the Apc(Min/+) mice model, suggesting its chemopreventive potential against colorectal cancer.


Asunto(s)
Transformación Celular Neoplásica/efectos de los fármacos , Pólipos Intestinales/prevención & control , Triterpenos/farmacología , Animales , Suplementos Dietéticos , Perfilación de la Expresión Génica , Genes APC , Masculino , Ratones , Ratones Endogámicos , Análisis por Micromatrices , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...