Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 272
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Food Funct ; 12(21): 10632-10643, 2021 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-34585698

RESUMEN

Safflower yellow (SY) is the main active ingredient isolated from the traditional Chinese medicine Carthamus tinctorius, which is a valuable natural edible pigment that is widely used to treat cerebrovascular and cardiovascular diseases. However, the effect of SY on hepatocellular carcinoma (HCC) remains unclear. In this study, we showed that SY decreased the degree of injury and inhibited the release of inflammatory factors in the liver of a diethylnitrosamine (DEN)-induced HCC mouse model. Flow cytometry and immunoblotting showed that SY increased the infiltration of CD8+ T cells and Gr-1+ macrophages to improve the immune microenvironment by affecting the expression of collagen fibers. Further cellular experiments showed that SY degraded the collagens in the liver cells through the TGF-ß/Smad signalling pathway. SY also regulated the gut microbiota which may contribute to the immune microenvironment. In conclusion, SY exhibited a potent effect on the development of HCC by enhancing liver immune infiltration by promoting collagen degradation and modulating the gut microbiota. This study provides novel insights into the mechanism of SY as a candidate for the treatment of HCC in the future.


Asunto(s)
Carcinoma Hepatocelular/inducido químicamente , Chalcona/análogos & derivados , Dietilnitrosamina/toxicidad , Microbioma Gastrointestinal/efectos de los fármacos , Neoplasias Hepáticas/inducido químicamente , Hígado/efectos de los fármacos , Animales , Carcinoma Hepatocelular/prevención & control , Línea Celular Tumoral , Chalcona/farmacología , Colágeno/metabolismo , Humanos , Hígado/inmunología , Hígado/metabolismo , Neoplasias Hepáticas/prevención & control , Macrófagos Peritoneales/efectos de los fármacos , Masculino , Ratones , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/prevención & control , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
2.
World J Gastroenterol ; 26(17): 2064-2081, 2020 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-32536775

RESUMEN

BACKGROUND: Epigallocatechin gallate (EGCG) is a polyhydroxy phenolic compound extracted from tea and its antitumor effect has received widespread attention. We explored the inhibitory effect of EGCG on dimethylhydrazine (DMH)-induced colorectal cancer (CRC) using a rat model, predicted the interaction between EGCG and CRC target genes using a database, and explained the EGCG associated target pathways and mechanisms in CRC. AIM: To understand the inhibitory mechanisms of EGCG on CRC cell proliferation and identify its pharmacological targets by network pharmacology analysis. METHODS: DMH (40 mg/kg, s.c., twice weekly for eight weeks) was used to induce CRC in rats. After model establishment, the rats were administered with EGCG (50, 100, or 200 mg/kg, p.o., once daily for eight weeks) and killed 12 and 20 wk after the start of the experiment. Formation of aberrant crypt foci and tumor was studied by histological analysis. Using network pharmacology analysis, candidate and collective targets of EGCG and CRC were identified, and Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes analyses were used to predict the pathways altered by EGCG. RESULTS: At week 12, high-dose EGCG treatment significantly reduced the tumor formation rate, total number of tumors, cancerous and non-cancerous tumors, tumor volume, ascites formation, and aberrant crypt foci count. At week 20, all three doses of EGCG were effective. Seventy-eight collective targets of EGCG and CRC were identified, of which 28 genes were dysregulated in CRC. Kyoto Encyclopedia of Genes and Genomes and GO analyses showed that the dysregulated genes were enriched in hsa05210 (CRC), hsa04115 (p53 signaling pathway), and hsa04151 (PI3K-Akt signaling pathway), GO:0043124 (negative regulation of I-kappaB kinase/NF-kappaB signaling pathway), GO:0043409 (negative regulation of mitogen-activated protein kinase cascade), and GO:2001244 (positive regulation of intrinsic apoptotic signaling pathway) respectively. CONCLUSION: EGCG inhibits the formation of DMH-induced CRC by regulating key pathways involved in tumorigenesis.


Asunto(s)
Focos de Criptas Aberrantes/prevención & control , Anticarcinógenos/farmacología , Catequina/análogos & derivados , Neoplasias Colorrectales/prevención & control , Neoplasias Experimentales/prevención & control , Focos de Criptas Aberrantes/inducido químicamente , Focos de Criptas Aberrantes/genética , Focos de Criptas Aberrantes/patología , Animales , Anticarcinógenos/uso terapéutico , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Catequina/farmacología , Catequina/uso terapéutico , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Colon/efectos de los fármacos , Colon/patología , Neoplasias Colorrectales/inducido químicamente , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Dimetilhidrazinas/toxicidad , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Masculino , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Mapas de Interacción de Proteínas/efectos de los fármacos , Mapas de Interacción de Proteínas/genética , Ratas , Recto/efectos de los fármacos , Recto/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
3.
Cancer Med ; 9(11): 3964-3973, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32243710

RESUMEN

Naturally occurring tumor in animals receiving high minerals from deep oceans (DOM: hardness 600 mg/L) from 6 months of age until natural death was firstly assessed in 200 Sprague Dawley rats, randomized into four groups: Control (C), DOM (D), Fructose (F), and Fructose + DOM (FD). Fructose drink contained 11% fructose. Tumor incidence (necropsy at death) in the D group was ~40% lower than that in the C group (P < .05), together with lower body mass gain and greater locomotive activity during their initial 18 months (P < .05) but not during later life. X-ray image analysis on abnormal solid tissue among survivors at 18 and 24 months of age confirms a similar trend, exhibiting ~50% and ~65% lower tumor incidence than the C and F groups, respectively. Reduced-to-oxidized glutathione ratio (GSH/GSSG) declined with age for the first three quarters of life on all groups (P < .05), followed by a resurgence during end-life among survivors at 24 months. This resurgence is markedly associated with lower tumor expansion but unrelated with DOM supplementation. Our results demonstrate valuable application of minerals and trace elements from deep oceans, as a vastly available natural source, on tumor suppression during normal aging.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Fructosa/toxicidad , Minerales/farmacología , Neoplasias Experimentales/prevención & control , Edulcorantes/toxicidad , Animales , Carcinogénesis/patología , Femenino , Esperanza de Vida , Masculino , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/patología , Océanos y Mares , Ratas , Ratas Sprague-Dawley
4.
Proc Natl Acad Sci U S A ; 117(11): 6075-6085, 2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-32123074

RESUMEN

MicroRNA-31 (miR-31) is overexpressed in esophageal squamous cell carcinoma (ESCC), a deadly disease associated with dietary Zn deficiency and inflammation. In a Zn deficiency-promoted rat ESCC model with miR-31 up-regulation, cancer-associated inflammation, and a high ESCC burden following N-nitrosomethylbenzylamine (NMBA) exposure, systemic antimiR-31 delivery reduced ESCC incidence from 85 to 45% (P = 0.038) and miR-31 gene knockout abrogated development of ESCC (P = 1 × 10-6). Transcriptomics, genome sequencing, and metabolomics analyses in these Zn-deficient rats revealed the molecular basis of ESCC abrogation by miR-31 knockout. Our identification of EGLN3, a known negative regulator of nuclear factor κB (NF-κB), as a direct target of miR-31 establishes a functional link between oncomiR-31, tumor suppressor target EGLN3, and up-regulated NF-κB-controlled inflammation signaling. Interaction among oncogenic miR-31, EGLN3 down-regulation, and inflammation was also documented in human ESCCs. miR-31 deletion resulted in suppression of miR-31-associated EGLN3/NF-κB-controlled inflammatory pathways. ESCC-free, Zn-deficient miR-31-/- rat esophagus displayed no genome instability and limited metabolic activity changes vs. the pronounced mutational burden and ESCC-associated metabolic changes of Zn-deficient wild-type rats. These results provide conclusive evidence that miR-31 expression is necessary for ESCC development.


Asunto(s)
Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas de Esófago/genética , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , MicroARNs/metabolismo , Neoplasias Experimentales/genética , Animales , Carcinógenos/toxicidad , Línea Celular Tumoral , Suplementos Dietéticos , Neoplasias Esofágicas/inducido químicamente , Neoplasias Esofágicas/patología , Neoplasias Esofágicas/prevención & control , Carcinoma de Células Escamosas de Esófago/inducido químicamente , Carcinoma de Células Escamosas de Esófago/patología , Carcinoma de Células Escamosas de Esófago/prevención & control , Esófago/patología , Regulación Neoplásica de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Masculino , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , FN-kappa B/metabolismo , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/patología , Neoplasias Experimentales/prevención & control , Nitrosaminas/toxicidad , Ratas , Ratas Transgénicas , Transducción de Señal/genética , Zinc/administración & dosificación , Zinc/deficiencia
5.
Eur J Cancer Prev ; 28(5): 383-389, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30234553

RESUMEN

Multicomponent therapy has gained interest for its potential to synergize and subsequently lower the effective dose of each constituent required to reduce colon cancer risk. We have previously showed that rapidly cycling Lgr5 stem cells are exquisitely sensitive to extrinsic dietary factors that modulate colon cancer risk. In the present study, we quantified the dose-dependent synergistic properties of dietary n-3 polyunsaturated fatty acids (PUFA) and curcumin (Cur) to promote targeted apoptotic deletion of damaged colonic Lgr5 stem cells. For this purpose, both heterogeneous bulk colonocytes and Lgr5 stem cells were isolated from Lgr5-EGFP-IRES-CreER knock-in mice injected with azoxymethane (AOM). Isolated cells were analyzed for DNA damage (γH2AX), apoptosis (cleaved caspase-3), and targeted apoptosis (both γH2AX and cleaved caspase-3) at 12 h post-AOM injection. Comparison of the percentage of targeted apoptosis in Lgr5 stem cells (GFP) across a broad bioactive dose-range revealed an ED50 of 16.0 mg/day n-3 PUFA + 15.9 mg/day Cur. This corresponded to a human equivalent dose of 3.0 g n-3 PUFA + 3.0 g Cur. In summary, our results provide evidence that a low dose (n-3 PUFA + Cur) combination diet reduces AOM-induced DNA damage in Lgr5 stem cells and enhances targeted apoptosis of DNA-damaged cells, implying that a lower human equivalent dose can be utilized in future human clinical trials.


Asunto(s)
Neoplasias del Colon/prevención & control , Curcumina/administración & dosificación , Ácidos Grasos Omega-3/administración & dosificación , Células Madre Neoplásicas/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , Animales , Apoptosis/efectos de los fármacos , Azoximetano/toxicidad , Carcinógenos/toxicidad , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/patología , Colon/citología , Colon/efectos de los fármacos , Colon/patología , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/patología , Suplementos Dietéticos , Relación Dosis-Respuesta a Droga , Femenino , Técnicas de Sustitución del Gen , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Transgénicos , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/patología , Neoplasias Experimentales/prevención & control , Células Madre Neoplásicas/patología , Receptores Acoplados a Proteínas G/genética
6.
BMC Cancer ; 18(1): 1111, 2018 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-30424722

RESUMEN

BACKGROUND: Colorectal cancer has been found to be attenuated either with prophylactic manipulation of gut microbiome with probiotics or celecoxib, a non-steroidal anti-inflammatory drug mainly by suppressing early pro-carcinogenic markers in various experimental studies. Therefore, the present study was designed to assess the prophylactic potential of combinatorial administration of probiotics (Lactobacillus rhamnosus GG, Lactobacillus acidophilus) and celecoxib in experimental colon carcinogenesis. METHODS: Six groups of Spraugue Dawely rats received probiotics L.rhamnosus GG or/and L.acidophilus in combination with celecoxib one week prior to the inducement of tumor by 1,2-dimethylhydrazine (DMH) and the treatment continued for 18 weeks. Prophylactic potentials of probiotics and celecoxib were determined by employing various methods such as tumor incidence, tumor burden, tumor multiplicity, apoptosis, caspase activity, expression of proto-oncogene K-ras and tumor suppressor p53 gene in colonic tumors. RESULTS: Interestingly, it was found that one week prior supplementation of both probiotics and celecoxib reduced tumor burden, tumor multiplicity, down-regulated the expression of anti-apoptotic Bcl-2, proto-oncogene K-ras and up-regulated pro-apoptotic Bax as well as tumor suppressor p53 in L.rhamnosus GG + celecoxib+DMH animals compared with counter controls and DMH-treated. CONCLUSIONS: It can be concluded that such combinatorial approach may be useful in reducing the burden and severity of disease in highly susceptible individuals but needs to be validated clinically.


Asunto(s)
Celecoxib/farmacología , Neoplasias Colorrectales/dietoterapia , Lacticaseibacillus rhamnosus , Lactobacillus acidophilus , Probióticos/administración & dosificación , 1,2-Dimetilhidrazina/toxicidad , Animales , Apoptosis/efectos de los fármacos , Carcinogénesis/efectos de los fármacos , Carcinogénesis/patología , Carcinógenos/toxicidad , Celecoxib/uso terapéutico , Colon/efectos de los fármacos , Colon/patología , Neoplasias Colorrectales/inducido químicamente , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/prevención & control , Terapia Combinada/métodos , Regulación hacia Abajo/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/dietoterapia , Neoplasias Experimentales/prevención & control , Proto-Oncogenes Mas , Ratas , Ratas Sprague-Dawley , Resultado del Tratamiento , Regulación hacia Arriba/efectos de los fármacos , Proteína X Asociada a bcl-2/metabolismo
7.
Nutrients ; 10(9)2018 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-30213082

RESUMEN

Pancreatic cancer remains a daunting foe despite a vast number of accumulating molecular analyses regarding the mutation and expression status of a variety of genes. Indeed, most pancreatic cancer cases uniformly present with a mutation in the KRAS allele leading to enhanced RAS activation. Yet our understanding of the many epigenetic/environmental factors contributing to disease incidence and progression is waning. Epidemiologic data suggest that diet may be a key factor in pancreatic cancer development and potentially a means of chemoprevention at earlier stages. While diets high in ω3 fatty acids are typically associated with tumor suppression, diets high in ω6 fatty acids have been linked to increased tumor development. Thus, to better understand the contribution of these polyunsaturated fatty acids to pancreatic carcinogenesis, we modeled early stage disease by targeting mutant KRAS to the exocrine pancreas and administered diets rich in these fatty acids to assess tumor formation and altered cell-signaling pathways. We discovered that, consistent with previous reports, the ω3-enriched diet led to reduced lesion penetrance via repression of proliferation associated with reduced phosphorylated AKT (pAKT), whereas the ω6-enriched diet accelerated tumor formation. These data provide a plausible mechanism underlying previously observed effects of fatty acids and suggest that administration of ω3 fatty acids can reduce the pro-survival, pro-growth functions of pAKT. Indeed, counseling subjects at risk to increase their intake of foods containing higher amounts of ω3 fatty acids could aid in the prevention of pancreatic cancer.


Asunto(s)
Anticarcinógenos/administración & dosificación , Transformación Celular Neoplásica/metabolismo , Dieta , Ácidos Grasos Omega-3/administración & dosificación , Neoplasias Experimentales/prevención & control , Conductos Pancreáticos/enzimología , Neoplasias Pancreáticas/prevención & control , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Animales , Apoptosis , Línea Celular , Proliferación Celular , Supervivencia Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Dieta/efectos adversos , Regulación hacia Abajo , Humanos , Ratones Transgénicos , Mutación , Neoplasias Experimentales/enzimología , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Conductos Pancreáticos/patología , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Fosforilación , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo
8.
Cancer Prev Res (Phila) ; 11(5): 265-278, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29437671

RESUMEN

An urgent need exists for the development of more efficacious molecular strategies targeting nonmelanoma skin cancer (NMSC), the most common malignancy worldwide. Inflammatory signaling downstream of Toll-like receptor 4 (TLR4) has been implicated in several forms of tumorigenesis, yet its role in solar UV-induced skin carcinogenesis remains undefined. We have previously shown in keratinocyte cell culture and SKH-1 mouse epidermis that topical application of the specific TLR4 antagonist resatorvid (TAK-242) blocks acute UV-induced AP-1 and NF-κB signaling, associated with downregulation of inflammatory mediators and MAP kinase phosphorylation. We therefore explored TLR4 as a novel target for chemoprevention of UV-induced NMSC. We selected the clinical TLR4 antagonist resatorvid based upon target specificity, potency, and physicochemical properties. Here, we confirm using ex vivo permeability assays that topical resatorvid can be effectively delivered to skin, and using in vivo studies that topical resatorvid can block UV-induced AP-1 activation in mouse epidermis. We also report that in a UV-induced skin tumorigenesis model, topical resatorvid displays potent photochemopreventive activity, significantly suppressing tumor area and multiplicity. Tumors harvested from resatorvid-treated mice display reduced activity of UV-associated signaling pathways and a corresponding increase in apoptosis compared with tumors from control animals. Further mechanistic insight on resatorvid-based photochemoprevention was obtained from unsupervised hierarchical clustering analysis of protein readouts via reverse-phase protein microarray revealing a significant attenuation of key UV-induced proteomic changes by resatorvid in chronically treated high-risk SKH-1 skin prior to tumorigenesis. Taken together, our data identify TLR4 as a novel molecular target for topical photochemoprevention of NMSC. Cancer Prev Res; 11(5); 265-78. ©2018 AACRSee related editorial by Sfanos, p. 251.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Neoplasias Cutáneas/prevención & control , Sulfonamidas/farmacología , Receptor Toll-Like 4/antagonistas & inhibidores , Rayos Ultravioleta/efectos adversos , Administración Cutánea , Animales , Carcinogénesis/efectos de la radiación , Evaluación Preclínica de Medicamentos , Epidermis/efectos de los fármacos , Epidermis/metabolismo , Epidermis/efectos de la radiación , Femenino , Humanos , Ratones , Ratones Pelados , Ratones Transgénicos , FN-kappa B/metabolismo , Neoplasias Experimentales/etiología , Neoplasias Experimentales/prevención & control , Permeabilidad , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Neoplasias Cutáneas/etiología , Sulfonamidas/uso terapéutico , Receptor Toll-Like 4/metabolismo , Factor de Transcripción AP-1/metabolismo
9.
Eur J Cancer Prev ; 27(2): 110-117, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-27926538

RESUMEN

This study focused on the chemopreventive effects of Spirogyra neglecta extract (SNE) and dried S. neglecta mixed diet on the early stages of 1,2-dimethylhydrazine (DMH)-induced colon carcinogenesis in rats. Male Wistar rats were injected with DMH to initiate aberrant crypt foci (ACF) formation. In the initiation stage, SNE significantly decreased the number of ACF in the colon of DMH-treated rats. Rats that received a low dose of SNE showed enhanced activity of several detoxifying and antioxidant enzymes. In the postinitiation stage, a low dose of SNE significantly decreased the number of ACF in the colon of DMH-treated rats. It significantly reduced the number of proliferating cell nuclear antigen-positive cells and increased the number of apoptotic cells in colonic crypts. S. neglecta thus inhibited the development of the early stages of DMH-induced colon carcinogenesis in rats by modulation of xenobiotic metabolizing enzymes and inhibition of cell proliferation as well as induction of apoptosis.


Asunto(s)
Focos de Criptas Aberrantes/prevención & control , Anticarcinógenos/uso terapéutico , Neoplasias del Colon/prevención & control , Extractos Vegetales/uso terapéutico , Spirogyra/química , 1,2-Dimetilhidrazina/toxicidad , Focos de Criptas Aberrantes/inducido químicamente , Focos de Criptas Aberrantes/patología , Animales , Anticarcinógenos/farmacología , Apoptosis/efectos de los fármacos , Hidrocarburo de Aril Hidroxilasas/metabolismo , Carcinógenos/toxicidad , Proliferación Celular/efectos de los fármacos , Colon/efectos de los fármacos , Colon/patología , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/patología , Humanos , Masculino , Estadificación de Neoplasias , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/prevención & control , Extractos Vegetales/farmacología , Ratas , Ratas Wistar , Resultado del Tratamiento
10.
Sci Rep ; 7(1): 14513, 2017 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-29109519

RESUMEN

Network pharmacology has become a powerful means of understanding the mechanisms underlying the action of Chinese herbs in cancer treatment. This study aims to validate the preventive effects and molecular mechanisms of a clinical prescription XIAOPI formula against breast cancer. In vivo breast cancer xenograft data showed that XIAOPI delayed breast cancer development and efficiently inhibited lung metastasis, accompanied by prolonged survival benefits and decreased cancer stem cell subpopulations. However, similar phenomenon were not observed in a cell model. The herb-ingredient-target network analysis further identified a total of 81 genes closely correlated with the breast cancer chemoprevention effects of XIAOPI. Cytokine array analysis further validated CXCL-1 as the key target of XIAOPI both in vitro and in vivo. Evaluation of the mechanism demonstrated that CXCL-1 administration significantly abrogated the metastatic inhibition effects of XIAOPI on breast cancer migration, invasion, stem cells subpopulations, epithelial-mesenchymal transition(EMT), or mammosphere formation abilities. Overall, our study provides experimental evidence and molecular mechanisms that may facilitate the safe and effective use of herbal medicine for the prevention of breast cancer growth or metastasis, and may lead to CXCL-1-based therapeutic strategies for mammary malignancies.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/prevención & control , Quimiocina CXCL1/metabolismo , Medicamentos Herbarios Chinos/farmacología , Metástasis de la Neoplasia/prevención & control , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/fisiología , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Ratones Transgénicos , Invasividad Neoplásica/patología , Invasividad Neoplásica/fisiopatología , Invasividad Neoplásica/prevención & control , Metástasis de la Neoplasia/patología , Metástasis de la Neoplasia/fisiopatología , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Neoplasias Experimentales/prevención & control , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología
11.
Cancer Prev Res (Phila) ; 10(11): 671-679, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28851689

RESUMEN

Prostacyclin (prostaglandin I2, PGI2) overproduction in FVB/N mice prevents the formation of carcinogen and tobacco smoke-induced adenomas, and administration of the oral prostacyclin analogue iloprost to wild-type mice also prevented carcinogen-induced mouse lung adenoma formation. Former smokers taking oral iloprost showed improved bronchial dysplasia histology compared with placebo. Next-generation oral prostacyclin analogues, like treprostinil, were developed for the treatment of pulmonary arterial hypertension (PAH). On the basis of our prior studies with iloprost, we performed preclinical studies examining the ability of treprostinil to chemoprevent urethane-induced murine lung adenocarcinoma. We determined the MTD in chow (prior studies had delivered treprostinil by gavage), and this dose produced serum levels in the experimental animals similar to those found in PAH patients treated with treprostinil. We then examined the chemopreventive efficacy of treprostinil exposure initiated both before (1 week) and after (6 weeks) urethane exposure to better model chemoprevention studies conducted in former smokers. Neither of these dosing strategies prevented murine lung cancer; however, we did detect changes in pulmonary inflammatory cell infiltrate and expression of CXCR4 (a chemokine receptor previously shown to increase in response to treprostinil exposure) in tumor-bearing, treprostinil-treated animals, indicating that the drug was bioavailable. One potential explanation stems from iloprost and treprostinil differentially activating cell surface prostaglandin receptors and intracellular peroxisome proliferator-activated receptors. When murine lung tumor cells were treated with treprostinil, their proliferation rate increased; in contrast, iloprost had no effect on proliferation. Future investigations comparing these two agents will provide insight into iloprost's chemopreventive mechanisms. Cancer Prev Res; 10(11); 671-9. ©2017 AACR.


Asunto(s)
Adenocarcinoma/prevención & control , Antihipertensivos/uso terapéutico , Epoprostenol/análogos & derivados , Neoplasias Pulmonares/prevención & control , Neoplasias Experimentales/prevención & control , Adenocarcinoma/inducido químicamente , Adenocarcinoma del Pulmón , Animales , Disponibilidad Biológica , Carcinógenos/toxicidad , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Epoprostenol/farmacología , Epoprostenol/uso terapéutico , Femenino , Humanos , Pulmón/citología , Pulmón/efectos de los fármacos , Pulmón/patología , Neoplasias Pulmonares/inducido químicamente , Ratones , Ratones Transgénicos , Neoplasias Experimentales/inducido químicamente , Receptores CXCR4/metabolismo , Receptores de Prostaglandina/metabolismo , Resultado del Tratamiento , Uretano/toxicidad
12.
Digestion ; 95(4): 275-280, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28494451

RESUMEN

BACKGROUND: Colorectal cancer is the main leading cause of cancer-related deaths worldwide. Present data suggest that plant-derived anthocyanins have anti-inflammatory and chemopreventive properties. This study was aimed at evaluating the effect of an anthocyanin-rich extract from bilberries on colorectal tumour development and growth in the administration of azoxymethan (AOM)/dextran sodium sulfate (DSS) mouse model. METHODS: Colonic carcinogenesis was induced by AOM and DSS 3 or 5%, respectively, in 50 female Balb/c mice. Mice received either normal food (controls) or a diet containing either 10 or 1% anthocyanin-rich bilberry extract. Colonoscopy took place at week 4 and 9 after initiation of carcinogenesis. After termination at week 9, colon samples were analysed macroscopically and microscopically. RESULTS: Mice receiving 10% anthocyanins showed significantly (p < 0.004) less reduced colon length (12.1 cm [8.5-14.4 cm]) as compared to controls (11.2 cm [9.8-12.3]) indicating less inflammation. Mice fed with 10% anthocyanin-rich extract revealed significantly less mean tumour numbers (n = 1.2) compared to control (n = 14) and anthocyanin 1% treated mice (n = 10.6, p < 0.001). CONCLUSION: Anthocyanins prevented the formation and growth of colorectal cancer in AOM/DSS-treated Balb/c mice. Further studies should investigate the mechanisms of how anthocyanins influence the development of colorectal cancer.


Asunto(s)
Antocianinas/uso terapéutico , Carcinoma in Situ/prevención & control , Neoplasias del Colon/prevención & control , Animales , Azoximetano , Carcinoma in Situ/inducido químicamente , Carcinoma in Situ/diagnóstico , Carcinoma in Situ/patología , Colon/patología , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/diagnóstico , Neoplasias del Colon/patología , Colonoscopía , Sulfato de Dextran , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Ratones Endogámicos BALB C , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/diagnóstico , Neoplasias Experimentales/patología , Neoplasias Experimentales/prevención & control , Fitoterapia
13.
Cancer Biol Ther ; 18(4): 257-267, 2017 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-28045589

RESUMEN

Selenium (Se) is an essential dietary micronutrient that has been examined for protection against different types of cancers including colon cancer. Despite an established inverse association between Se and chronic inflammation induced colon cancer (CICC), the mechanistic understanding of Se's protective effects requires additional in-vivo studies using preclinical animal models of CICC. Adiponectin (APN) is an adipocytokine that is protective against CICC as well. However, its role in the anti-mutagenic effects of the Se-diet remains unknown. To address this knowledge gap, here we examine the ability of dietary Se in reducing CICC in APN knockout mice (KO) and its wild-type C57BL/6. CICC was induced with the colon cancer agent 1,2 dimethyl hydrazine (DMH) along with dextran sodium sulfate (DSS). Se-enhanced diet increased selenoproteins, Gpx-1 and Gpx-2, in the colon tissues, thereby reducing oxidative stress. Se-mediated reduction of CICC was evident from the histopathological studies in both mouse models. In both mice, reduction in inflammation and tumorigenesis associated well with reduced p65 phosphorylation and elevated 53 phosphorylation. Finally, we show that in both models Se-administration promotes goblet cell differentiation with a concomitant increase in the levels of associated proteins, Muc-2 and Math-1. Our findings suggest that Se's protection against CICC involves both colonic epithelial protection and anti-tumor effects that are independent of APN.


Asunto(s)
Adiponectina/genética , Colitis Ulcerosa/complicaciones , Neoplasias del Colon/prevención & control , Micronutrientes/metabolismo , Selenio/metabolismo , 1,2-Dimetilhidrazina/toxicidad , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Carcinogénesis/patología , Diferenciación Celular , Transformación Celular Neoplásica/patología , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/patología , Colon/patología , Neoplasias del Colon/etiología , Neoplasias del Colon/patología , Sulfato de Dextran/toxicidad , Glutatión Peroxidasa/metabolismo , Células Caliciformes/patología , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mucina 2/metabolismo , Mutagénesis , Neoplasias Experimentales/etiología , Neoplasias Experimentales/patología , Neoplasias Experimentales/prevención & control , Fosforilación , Factor de Transcripción ReIA/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Glutatión Peroxidasa GPX1
14.
Oncotarget ; 7(47): 76944-76954, 2016 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-27769066

RESUMEN

Although epidemiological and preclinical studies have shown the preventative effect of n-3 polyunsaturated fatty acids (PUFAs) on colorectal cancer (CRC), the underlying molecular mechanisms are not clear. In this study, we revealed that elevation of n-3/n-6 PUFAs ratio suppress the mechanistic target of rapamycin complex 1 (mTORC1) and prevent colorectal tumorigenesis. The transgenic expression of fat-1, a desaturase that catalyzes the conversion of n-6 to n-3 PUFAs and produces n-3 PUFAs endogenously, repressed colorectal tumor cell growth and remarkably reduced tumor burden, and alleviated anemia as well as hyperlipidemia in APCMin/+ (adenomatous polyposis coli) mice, a classic CRC model that best simulates most clinical cases. In contrast to arachidonic acid (AA, C20:4 n-6), either Docosahexaenoic acid (DHA, C22:6 n-3), eicosapentaenoic acid (EPA, C20:5 n-3), or a combination of DHA and AA, efficiently inhibited the proliferation of CRC cell lines and promoted apoptosis in these cells. The ectopic expression of fat-1 had similar effects in colon epithelial cells with APC depletion. Mechanistically, elevation of n-3/n-6 ratio suppressed mTORC1 activity in tumors of APCMin/+ mice, CRC cell lines with APC mutation, and in normal colon epithelial cells with APC depletion. In addition, elevation of n-3/n-6 ratio repressed mTORC1 activity and inhibited adipogenic differentiation in preadipocytes with APC knockdown, as well as alleviated hyperlipidemia in APCMin/+ mice. Taken together, our findings have provided novel insights into the potential mechanism by which increase in n-3/n-6 PUFAs ratio represses CRC development, and also a new rationale for utilizing n-3 PUFAs in CRC prevention and treatment.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Neoplasias Colorrectales/prevención & control , Ácido Graso Desaturasas/genética , Ácidos Grasos Omega-3/administración & dosificación , Serina-Treonina Quinasas TOR/genética , Células 3T3-L1 , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Neoplasias Colorrectales/genética , Ácidos Grasos Omega-3/farmacología , Ácidos Grasos Omega-6/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HCT116 , Humanos , Ratones , Ratones Transgénicos , Mutación , Neoplasias Experimentales/genética , Neoplasias Experimentales/prevención & control
15.
Oncotarget ; 7(38): 61643-61655, 2016 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-27557503

RESUMEN

Individuals with ulcerative colitis (UC) are at a high risk for developing colorectal cancer (CRC). Huangqin Decoction (HQD), a traditional Chinese medicinal formula chronicled in the Shang Han Lun, is commonly used to treat gastrointestinal symptoms. However, experimental evidence for supporting the clinical practice is lacking. This study used modern biomedical approaches to investigate the protective/preventive effects of HQD in dextran sulfate sodium (DSS)-induced acute/chronic UC and azoxymethane (AOM)/DSS-induced CRC in mice. HQDs were prepared in 4 different ways: HQD-1 and HQD-2 were prepared in boiling water, whereas HQD-3 and HQD-4 were prepared in heated ethanol (70%). For HQD-1 and HQD-3, the 4 constituent herbs were processed together, whereas for HQD-2 and HQD4, these herbs were processed individually and then combined. The mice were administered 9.1 g/kg HQD via oral gavage daily. HQD-1 significantly inhibited DSS-induced acute UC, whereas HQD-3 and HQD-4 exhibited mild ameliorative effects; but HQD-2 had no protective effect and resulted in a higher mortality rate. This higher mortality rate may be due to the greater abundance of baicalein and wogonin in HQD-2 than HQD-1. Furthermore, HQD-1 protected against DSS-induced chronic UC and significantly inhibited AOM/DSS-induced CRC in mice. HQD-1 also inhibited the production of inflammatory cytokines and increased antioxidant capacity both in chronic DSS and AOM/DSS treated mice. Overall, HQD-1 inhibits the development of acute/chronic colitis and prevents colitis-associated CRC, possibly by inhibiting inflammation and preventing oxidative stress induced cellular damage.


Asunto(s)
Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/prevención & control , Neoplasias Colorrectales/prevención & control , Extractos Vegetales/farmacología , Scutellaria baicalensis/química , Animales , Antioxidantes/química , Azoximetano/química , Cromatografía Líquida de Alta Presión , Neoplasias Colorrectales/inducido químicamente , Citocinas/metabolismo , Sulfato de Dextran , Modelos Animales de Enfermedad , Medicamentos Herbarios Chinos/farmacología , Flavanonas/química , Inflamación , Masculino , Medicina Tradicional China , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/prevención & control , Estrés Oxidativo
16.
Cancer Immunol Res ; 4(5): 441-51, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26896171

RESUMEN

TNF is a potent promoter of carcinogenesis and potentially important target for cancer prevention. TNF is produced as functionally distinct transmembrane and soluble molecules (tmTNF and sTNF, respectively), but their individual roles in carcinogenesis are unexplored. We investigated the participation of tmTNF and sTNF in chemically induced carcinogenesis in mice. We found that injection of XPro1595, a dominant-negative TNF biologic (DN-TNF) and specific antagonist of sTNF, decreased tumor incidence and growth, and prolonged survival of 3-methylcholanthrene (MCA)-injected mice. Similar results were obtained following the exclusion of both TNF forms by either TNF-receptor 2-Fc fusion protein (TNFR2-Fc) treatment or TNF gene deletion. In addition, gene deletion of TNFR1, which is preferentially triggered by sTNF, was temporarily blocked, whereas gene deletion of TNFR2, which is preferentially triggered by tmTNF, enhanced MCA-induced carcinogenesis. Concomitantly with carcinogenesis induction, MCA increased circulating IL1α, accumulation of myeloid-derived suppressor cells (MDSC), STAT3 phosphorylation, and immunosuppression in the spleen. In sharp contrast, DN-TNF treatment dramatically decreased IL1α and increased the essential immunoregulatory cytokines IL1ß, IL12p70, and IL17 in the peripheral blood of MCA-injected mice. In addition, MDSC accumulation, STAT3 phosphorylation, and immunosuppression in MCA-injected mice were prevented by DN-TNF treatment, TNFR2-Fc treatment, and/or gene deletion of TNF or TNFR1, but not deletion of TNFR2. These findings reveal that sTNF is both an essential promoter of carcinogenesis and a pivotal regulator of MDSCs, and indicate that sTNF could be a significant target for cancer prevention and therapy. Cancer Immunol Res; 4(5); 441-51. ©2016 AACR.


Asunto(s)
Neoplasias Experimentales/prevención & control , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Carcinogénesis/efectos de los fármacos , Citocinas/biosíntesis , Células Dendríticas/inmunología , Evaluación Preclínica de Medicamentos/métodos , Femenino , Eliminación de Gen , Tolerancia Inmunológica , Células Asesinas Naturales/inmunología , Metilcolantreno , Ratones Endogámicos C57BL , Ratones SCID , Terapia Molecular Dirigida/métodos , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Factor de Transcripción STAT3/metabolismo , Solubilidad , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/farmacología , Factor de Necrosis Tumoral alfa/uso terapéutico
17.
Asian Pac J Cancer Prev ; 16(18): 8119-26, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26745048

RESUMEN

Cancer is a leading cause of death worldwide. Due to the toxic side effects of the commonly used chemotherapeutic drug cyclophosphamide (CTX), the use of herbal medicines with fewer side effects but having potential use as inducing anti-cancer outcomes in situ has become increasingly popular. The present study sought to investigate the effects of a methanolic extract of Bauhinia tomentosa against Dalton's ascites lymphoma (DAL) induced ascites as well as solid tumors in BALB/c mice. Specifically, B. tomentosa extract was administered intraperitonealy (IP) at 10 mg/kg. BW body weight starting just after tumor cell implantation and thereafter for 10 consecutive days. In the ascites tumor model hosts, administration of extract resulted in a 52% increase in the life span. In solid tumor models, co-administration of extract and CTX significantly reduced tumor volume (relative to in untreated hosts) by 73% compared to just by 52% when the extract alone was provided. Co-administration of the extract also mitigated CTX-induced toxicity, including decreases in WBC count, and in bone marrow cellularity and α-esterase activity. Extract treatment also attenuated any increases in serum levels of TNFα, iNOS, IL-1ß, IL-6, GM-CSF, and VEGF seen in tumor-bearing hosts. This study confirmed that, the potent antitumor activity of B.tomentosa extract may be associated with immune modulatory effects by regulating anti-oxidants and cytokine levels.


Asunto(s)
Bauhinia/química , Mediadores de Inflamación/sangre , Péptidos y Proteínas de Señalización Intercelular/sangre , Linfoma/prevención & control , Neoplasias Experimentales/prevención & control , Fitoterapia , Extractos Vegetales/farmacología , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Humanos , Linfoma/metabolismo , Linfoma/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Células Tumorales Cultivadas
18.
Nutr Cancer ; 66(6): 1059-69, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24945996

RESUMEN

Pu-erh tea is a functional tea production in China. The functional effects should be proved. The oral cancer preventive and antimetastatic effects of Pu-erh tea in vitro and in vivo have been studied respectively. Pu-erh tea showed an inhibitory effect on human tongue carcinoma TCA8113 cells proliferation tested by 3-(4,5-Dimethyl-2-Thiazolyl)-2,5-Diphenyltetrazolium Bromide assay and induced TCA8113 apoptosis shown anticancer effect. The antimetastatic effect of Pu-erh tea in TCA8113 cells was proved by the decreasing of matrix metalloproteinases (MMPs) and increasing of tissue inhibitors of metalloproteinases (TIMPs) mRNA transcription. In the animal experiments, the tumor volumes and lymph node metastasis rates of Pu-erh tea-treated mice were smaller than control mice. Pu-erh tea reduced the levels of the serum proinflammatory cytokines interleukin (IL)-6, IL-12, tumor necrosis factor-α, and interferon-γ to a greater extent compared with the control mice, and the levels of 200 µg/mL treatment was more close to the normal mice than 100 µg/mL treated mice. Pu-erh tea also significantly induced apoptosis in tissues of mice (P < 0.05) by upregulating Bax and downregulating Bcl-2. These results demonstrate Pu-erh tea has cancer preventive and anti-metastatic effects on buccal mucosa cancer, the higher concentration get better efficiency.


Asunto(s)
Antineoplásicos/farmacología , Mucosa Bucal/efectos de los fármacos , Neoplasias Experimentales/prevención & control , Té/química , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , China , Regulación hacia Abajo , Femenino , Humanos , Interferón gamma/sangre , Interleucina-12/sangre , Interleucina-6/sangre , Metaloproteinasas de la Matriz/metabolismo , Ratones , Mucosa Bucal/citología , Mucosa Bucal/patología , Neoplasias de la Boca/prevención & control , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Inhibidores Tisulares de Metaloproteinasas/metabolismo , Factor de Necrosis Tumoral alfa/sangre , Regulación hacia Arriba , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
19.
Mutagenesis ; 29(3): 177-87, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24531571

RESUMEN

A complex dietary supplement designed to impact multiple mechanisms associated with aging and cancer reduced overall tumorigenesis in cancer-prone heterozygous Trp53+/- mice by ~30% (P < 0.018). Carcinomas were reduced by 67% (P < 0.006). Remarkably, metastasis (a leading cause of cancer mortality) was undetectable in treated animals (P < 0.004), and the occurrence of multiple primary tumours was reduced by 74% (P < 0.012). Reduction of pulmonary adenocarcinoma by 62% (P < 0.021) was of particular note given that lung cancer is the second leading cause of death in humans. Tumours showed pronounced age-related expression in untreated animals older than 600 days. Benefits of treatment only emerged in these later ages, suggesting that the supplement acted on mechanisms common to aging and cancer. The supplement was administered daily on bagel bits that were usually eaten within minutes by the mice. Although longevity was not statistically different between treatments, longevity was strongly related to the compliance of mice in eating the supplement. Linear regression revealed a strong positive relationship between the proportion of supplement eaten and the longevity of mice within the treatment group (P < 0.0001).


Asunto(s)
Suplementos Dietéticos , Neoplasias Experimentales/prevención & control , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética , Adenocarcinoma/genética , Adenocarcinoma/prevención & control , Adenocarcinoma del Pulmón , Animales , Suplementos Dietéticos/análisis , Genes p53 , Humanos , Modelos Lineales , Longevidad , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/prevención & control , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/prevención & control , Neoplasias Experimentales/genética , Neoplasias Experimentales/secundario , Neoplasias Primarias Múltiples/genética , Neoplasias Primarias Múltiples/prevención & control , Estrés Oxidativo
20.
Cancer Prev Res (Phila) ; 7(2): 246-54, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24346344

RESUMEN

Urinary bladder cancer prevention studies were performed with the nonsteroidal anti-inflammatory drugs (NSAID) naproxen (a standard NSAID with a good cardiovascular profile), sulindac, and their nitric oxide (NO) derivatives. In addition, the effects of the ornithine decarboxylase inhibitor, difluoromethylornithine (DFMO), alone or combined with a suboptimal dose of naproxen or sulindac was examined. Agents were evaluated at their human equivalent doses (HED), as well as at lower doses. In the hydroxybutyl(butyl)nitrosamine (OH-BBN) model of urinary bladder cancer, naproxen (400 or 75 ppm) and sulindac (400 ppm) reduced the incidence of large bladder cancers by 82%, 68%, and 44%, respectively, when the agents were initially given 3 months after the final dose of the carcinogen; microscopic cancers already existed. NO-naproxen was highly effective, whereas NO-sulindac was inactive. To further compare naproxen and NO-naproxen, we examined their effects on gene expression in rat livers following a 7-day exposure. Limited, but similar, gene expression changes in the liver were induced by both agents, implying that the primary effects of both are mediated by the parent NSAID. When agents were initiated 2 weeks after the last administration of OH-BBN, DFMO at 1,000 ppm had limited activity, a low dose of naproxen (75 ppm) and sulindac (150 ppm) were highly and marginally effective. Combining DFMO with suboptimal doses of naproxen had minimal effects, whereas the combination of DMFO and sulindac was more active than either agent alone. Thus, naproxen and NO-naproxen were highly effective, whereas sulindac was moderately effective in the OH-BBN model at their HEDs.


Asunto(s)
Antiinflamatorios no Esteroideos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Eflornitina/administración & dosificación , Neoplasias Experimentales/prevención & control , Óxido Nítrico/administración & dosificación , Neoplasias de la Vejiga Urinaria/prevención & control , Animales , Quimioprevención/métodos , Evaluación Preclínica de Medicamentos , Femenino , Naproxeno/administración & dosificación , Naproxeno/análogos & derivados , Neoplasias Experimentales/inducido químicamente , Ratas , Ratas Endogámicas F344 , Sulindac/administración & dosificación , Neoplasias de la Vejiga Urinaria/inducido químicamente
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA