Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 22
1.
Oncol Rep ; 43(4): 1053-1066, 2020 04.
Article En | MEDLINE | ID: mdl-32323786

Colorectal cancer (CRC) is one of the most common digestive tract tumors worldwide. Catalpol exerts inhibitory effects on the progression of several cancer types by regulating microRNAs (miRs). However, the precise role and carcinostatic mechanism of catalpol on CRC cells are poorly understood which limits the application of catalpol treatment. In the present study, miR­34a and sirtuin 1 (SIRT1) expression levels were detected in CRC tissues and CRC cell lines by RT­qPCR. Computational software analysis, luciferase assays and western blotting were used to demonstrate the downstream target of miR­34a in CRC cells. Effects of catalpol on cell viability, apoptosis, autophagic flux and the miR­34a/SIRT1 axis in the CRC cells were assessed by CCK­8 assay, flow cytometry, electron microscopy and western blotting, respectively. Whether the miR­34a/SIRT1 axis participated in catalpol­mediated autophagy and apoptosis was investigated. The effects of catalpol on the miR­34a/SIRT1 axis and malignant behavior were evaluated in a rat model of azoxymethane (AOM)­induced CRC. It was revealed that miR­34a expression levels were significantly decreased while SIRT1 was overexpressed in most of the CRC tissues and all the CRC cell lines. Clinically, a low level of miR­34a was correlated with poor clinicopathological characteristics in CRC patients. Catalpol reduced cell viability, suppressed autophagy, promoted apoptosis, and regulated the expression of SIRT1 by inducing miR­34a in vitro and in vivo. The autophagy­inhibiting effect of catalpol may be a mechanism to promote apoptosis of CRC cells. miR­34a mimic transfection resulted in autophagy­suppressive activity similar to that of catalpol, while the miR­34a inhibitor attenuated the antiautophagic effects of catalpol. In conclusion, miR­34a is involved in regulating catalpol­mediated autophagy and malignant behavior by directly inhibiting SIRT1 in CRC.


Autophagy , Colonic Neoplasms/drug therapy , Colorectal Neoplasms/drug therapy , Iridoid Glucosides/pharmacology , MicroRNAs/genetics , Rehmannia/chemistry , Sirtuin 1/metabolism , Aged , Animals , Apoptosis , Azoxymethane/chemistry , Carcinogens/chemistry , Cell Line, Tumor , Cell Proliferation , Cell Survival , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Disease Models, Animal , Drugs, Chinese Herbal/pharmacology , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Rats , Rats, Wistar , Up-Regulation
2.
Proc Natl Acad Sci U S A ; 115(5): 1057-1062, 2018 01 30.
Article En | MEDLINE | ID: mdl-29339485

d-2-hydroxyglutarate (D2HG) is produced in the tricarboxylic acid cycle and is quickly converted to α-ketoglutarate by d-2-hydroxyglutarate dehydrogenase (D2HGDH). In a mouse model of colitis-associated colon cancer (CAC), urine level of D2HG during colitis correlates positively with subsequent polyp counts and severity of dysplasia. The i.p. injection of D2HG results in delayed recovery from colitis and severe tumorigenesis. The colonic expression of D2HGDH is decreased in ulcerative colitis (UC) patients at baseline who progress to cancer. Hypoxia-inducible factor (Hif)-1α is a key regulator of D2HGDH transcription. Our study identifies urine D2HG and tissue D2HGDH expression as biomarkers to identify patients at risk for progressing from colitis to cancer. The D2HG/D2HGDH pathway provides potential therapeutic targets for the treatment of CAC.


Colitis/metabolism , Colitis/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Glutarates/metabolism , Animals , Apoptosis , Azoxymethane/chemistry , Biomarkers, Tumor/metabolism , Biopsy , Caco-2 Cells , Cell Movement , Cell Proliferation , Cell Survival , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Dextran Sulfate/chemistry , Disease Progression , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation , Intestinal Mucosa/pathology , Mice , Risk
3.
Biomed Res Int ; 2017: 9517287, 2017.
Article En | MEDLINE | ID: mdl-28116312

Chemoprevention has become an important area in cancer research due to low success rate of current therapeutic modalities. Diet plays a vital role in the etiology of cancer. This research was carried out to study the chemopreventive properties of germinated rough rice (GRR) crude extract in Sprague-Dawley rats induced with azoxymethane. Germination of rough rice causes significant changes in several chemical compositions of presently bioactive compounds. These compounds may prevent or postpone the inception of cancer. Fifty male Sprague-Dawley rats (6 weeks of age) were randomly divided into 5 groups which were (G1) induced with azoxymethane (AOM) and not given GRR (positive control), (G2) induced with AOM and given 2000 mg/kg GRR, (G3) induced with AOM and given 1000 mg/kg GRR, (G4) induced with AOM and given 500 mg/kg GRR, and (G5) not induced with AOM and not given GRR crude extract (negative control). To induce colon cancer, rats received two IP injections of AOM in saline (15 mg/kg) for two subsequent weeks. Organs were removed and weighed. Aberrant crypt foci (ACF) were evaluated histopathologically. ß-Catenin expressions were determined by Western blot. Treatment with 2000 mg/kg GRR crude extract not only resulted in the greatest reduction in the size and number of ACF but also displayed the highest percentage of nondysplastic ACF. Treatment with 2000 mg/kg GRR also gave the lowest level of expression in ß-catenin. Thus, GRR could be a promising dietary supplement for prevention of CRC.


Aberrant Crypt Foci/chemically induced , Anticarcinogenic Agents/therapeutic use , Azoxymethane/chemistry , Oryza/chemistry , Plant Extracts/chemistry , Animals , Carcinogens , Colon/drug effects , Colonic Neoplasms/chemically induced , Colonic Neoplasms/drug therapy , Diet , Dietary Supplements , Inhibitory Concentration 50 , Male , Mutation , Powders , Rats , Rats, Sprague-Dawley , beta Catenin/metabolism
4.
Phytother Res ; 31(1): 90-99, 2017 Jan.
Article En | MEDLINE | ID: mdl-27730672

Chemopreventative properties of traditional medicines and underlying mechanisms of action are incompletely investigated. This study demonstrates that dietary daikenchuto (TU-100), comprised of ginger, ginseng, and Japanese pepper effectively suppresses intestinal tumor development and progression in the azoxymethane (AOM) and APCmin/+ mouse models. For the AOM model, TU-100 was provided after the first of six biweekly AOM injections. Mice were sacrificed at 30 weeks. APCmin/+ mice were fed diet without or with TU-100 starting at 6 weeks, and sacrificed at 24 weeks. In both models, dietary TU-100 decreased tumor size. In APC min/+ mice, the number of small intestinal tumors was significantly decreased. In the AOM model, both TU-100 and Japanese ginseng decreased colon tumor numbers. Decreased Ki-67 and ß-catenin immunostaining and activation of numerous transduction pathways involved in tumor initiation and progression were observed. EGF receptor expression and stimulation/phosphorylation in vitro were investigated in C2BBe1 cells. TU-100, ginger, and 6-gingerol suppressed EGF receptor induced Akt activation. TU-100 and ginseng and to a lesser extent ginger or 6-gingerol inhibited EGF ERK1/2 activation. TU-100 and some of its components and metabolites of these components inhibit tumor progression in two mouse models of colon cancer by blocking downstream pathways of EGF receptor activation. Copyright © 2016 John Wiley & Sons, Ltd.


Azoxymethane/chemistry , Colonic Neoplasms/drug therapy , Plant Extracts/therapeutic use , Animals , Azoxymethane/pharmacology , Colonic Neoplasms/pathology , Disease Models, Animal , Male , Medicine, Traditional , Mice , Panax , Plant Extracts/administration & dosage , Zanthoxylum , Zingiberaceae
5.
Oncotarget ; 7(38): 61643-61655, 2016 Sep 20.
Article En | MEDLINE | ID: mdl-27557503

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.


Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/prevention & control , Colorectal Neoplasms/prevention & control , Plant Extracts/pharmacology , Scutellaria baicalensis/chemistry , Animals , Antioxidants/chemistry , Azoxymethane/chemistry , Chromatography, High Pressure Liquid , Colorectal Neoplasms/chemically induced , Cytokines/metabolism , Dextran Sulfate , Disease Models, Animal , Drugs, Chinese Herbal/pharmacology , Flavanones/chemistry , Inflammation , Male , Medicine, Chinese Traditional , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/prevention & control , Oxidative Stress
6.
Oncotarget ; 7(36): 58315-58330, 2016 Sep 06.
Article En | MEDLINE | ID: mdl-27506947

Colorectal cancer (CRC) is one of the most common cancers leading to high mortality. However, long-term administration of anti-tumor therapy for CRC is not feasible due to the side effects. Omega-3 polyunsaturated fatty acids (ω-3 PUFAs), particularly DHA and EPA, exert protection against CRC, but the mechanisms are unclear. Here, we show that ω-3 PUFAs inhibit proliferation and induce apoptosis of CRC cells in vitro and alleviate AOM/DSS-induced mice colorectal cancer in vivo. Moreover, ω-3 PUFAs promote phosphorylation and cytoplasmic retention of YAP and this effect was mediated by MST1/2 and LATS1, suggesting that the canonical Hippo Pathway is involved in ω-3 PUFAs function. We further confirmed that increase of pYAP by ω-3 PUFAs was mediated by GPRs, including GPR40 and GPR120, which subsequently activate PKA via Gαs, thus inducing the Hippo pathway activation. These data provide a novel DHA/EPA-GPR40/120-Gαs-PKA-MST1/2-LATS1-YAP signaling pathway which is linked to ω-3 PUFAs-induced inhibition of cell proliferation and promotion of apoptosis in CRC cells, indicating a mechanism that could explain the anti-cancer action of ω-3 PUFAs.


Colorectal Neoplasms/metabolism , Fatty Acids, Omega-3/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, G-Protein-Coupled/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Azoxymethane/chemistry , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/therapy , Cytoplasm/metabolism , Dextran Sulfate/chemistry , HT29 Cells , Hippo Signaling Pathway , Humans , Mice , Mice, Inbred BALB C , Phosphoproteins/metabolism , Phosphorylation , Protein Transport , YAP-Signaling Proteins
7.
J. coloproctol. (Rio J., Impr.) ; 36(1): 53-57, Jan.-Mar. 2016. ilus
Article En | LILACS | ID: lil-780055

Colorectal cancer is the leading cause of malignancy of the gastrointestinal tract. A better understanding of the molecular and cellular changes that lead to the disease is necessary to develop early diagnosis and optimal treatment modalities. Rodent models are rapid, reproducible and exhibit an adenoma-carcinoma sequence similar to that found in humans. The objective of this manuscript is to review the most common chemical carcinogens used to induce experimental tumors and the usual methods of evaluation.


O câncer colorretal é a principal neoplasia maligna do trato gastrointestinal. Um melhor entendimento dos processos moleculares e celulares é necessário para o desenvolvimento de estratégias que permitam um diagnóstico precoce e um tratamento mais eficaz. Modelos que utilizam roedores são rápidos, reprodutíveis e permitem o estudo da sequencia adenoma-carcinoma de forma similar a encontrada em humanos. O objetivo desse manuscrito é revisar os principais modelos de carcinogênese química e os métodos mais usuais para avaliação dos resultados.


Animals , Rats , Colorectal Neoplasms/diagnosis , Models, Animal , Azoxymethane/chemistry , Acids, Heterocyclic , Immunohistochemistry , Colorectal Neoplasms/genetics , 1,2-Dimethylhydrazine , Environmental Biomarkers , Amino Acids, Aromatic , Disease Models, Animal , Alkylation , Endoscopy , Carcinogenesis/chemistry
8.
J Nutr Biochem ; 26(12): 1641-9, 2015 Dec.
Article En | MEDLINE | ID: mdl-26383537

Cancer stem cells (CSCs) are shown to be responsible for initiation and progression of tumors in a variety of cancers. We previously showed that anthocyanin-containing baked purple-fleshed potato (PP) extracts (PA) suppressed early and advanced human colon cancer cell proliferation and induced apoptosis, but their effect on colon CSCs is not known. Considering the evidence of bioactive compounds, such as anthocyanins, against cancers, there is a critical need to study anticancer activity of PP, a global food crop, against colon CSCs. Thus, isolated colon CSCs (positive for CD44, CD133 and ALDH1b1 markers) with functioning p53 and shRNA-attenuated p53 were treated with PA at 5.0 µg/ml. Effects of baked PP (20% wt/wt) against colon CSCs were also tested in vivo in mice with azoxymethane-induced colon tumorigenesis. Effects of PA/PP were compared to positive control sulindac. In vitro, PA suppressed proliferation and elevated apoptosis in a p53-independent manner in colon CSCs. PA, but not sulindac, suppressed levels of Wnt pathway effector ß-catenin (a critical regulator of CSC proliferation) and its downstream proteins (c-Myc and cyclin D1) and elevated Bax and cytochrome c, proteins-mediating mitochondrial apoptosis. In vivo, PP reduced the number of crypts containing cells with nuclear ß-catenin (an indicator of colon CSCs) via induction of apoptosis and suppressed tumor incidence similar to that of sulindac. Combined, our data suggest that PP may contribute to reduced colon CSCs number and tumor incidence in vivo via suppression of Wnt/ß-catenin signaling and elevation of mitochondria-mediated apoptosis.


Anthocyanins/chemistry , Colonic Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Solanum tuberosum/chemistry , Animals , Antineoplastic Agents/chemistry , Apoptosis , Azoxymethane/chemistry , Carcinogenesis , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cell Transformation, Neoplastic/metabolism , Colonic Neoplasms/diet therapy , Colonic Neoplasms/prevention & control , Cytochromes c/metabolism , Food , Humans , In Situ Nick-End Labeling , Lentivirus , Male , Mice , Mitochondria/metabolism , Neoplastic Stem Cells/cytology , RNA, Small Interfering/metabolism , Sulindac/chemistry , Tumor Suppressor Protein p53/metabolism , Wnt Proteins/metabolism , bcl-2-Associated X Protein/metabolism , beta Catenin/metabolism
9.
J Nutr Biochem ; 26(12): 1547-58, 2015 Dec.
Article En | MEDLINE | ID: mdl-26355019

Patients with inflammatory bowel disease (IBD) are at increased risk for developing ulcerative colitis-associated colorectal cancer (CRC). The interleukin-6 (IL-6)/signal transducer and activator of transcription (STAT)-3 signaling regulates survival and proliferation of intestinal epithelial cells and play an important role in the pathogenesis of IBD and CRC. Cocoa is enriched with polyphenols that known to possess antioxidant, anti-inflammatory and antitumor activities. Here, we explored the antitumor effects and mechanisms of cocoa diet on colitis-associated cancer (CAC) using the azoxymethane/dextran sulfate sodium model, with a particular focus on whether cocoa exerts its anticancer effect through the IL-6/STAT3 pathway. We found that cocoa significantly decreased the tumor incidence and size in CAC-induced mice. In addition to inhibiting proliferation of tumor epithelial cells, cocoa suppressed colonic IL-6 expression and subsequently activation of STAT3. Thus, our findings demonstrated that cocoa diet suppresses CAC tumorigenesis, and its antitumor effect is partly mediated by limiting IL-6/STAT3 activation. In addition, cocoa induces apoptosis by increased the expressions of Bax and caspase 3 and decreased Bcl-xl. Thus, we conclude that cocoa may be a potential agent in the prevention and treatment of CAC.


Cacao/chemistry , Colorectal Neoplasms/prevention & control , Diet , Interleukin-6/metabolism , STAT3 Transcription Factor/metabolism , Animals , Anti-Inflammatory Agents/chemistry , Antineoplastic Agents/chemistry , Antioxidants/chemistry , Apoptosis , Azoxymethane/chemistry , Caspase 3/metabolism , Colitis/complications , Colitis/metabolism , Colorectal Neoplasms/metabolism , Dextran Sulfate/chemistry , Epithelial Cells/metabolism , Female , Gene Expression Regulation , Inflammatory Bowel Diseases/complications , Inflammatory Bowel Diseases/metabolism , Intestinal Mucosa/metabolism , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Neoplasms/pathology , Peroxidase/metabolism , Polyphenols/chemistry , Signal Transduction , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
10.
Cancer Prev Res (Phila) ; 8(11): 1076-83, 2015 Nov.
Article En | MEDLINE | ID: mdl-26323483

We have shown that dietary fish oil is protective against experimentally induced colon cancer, and the protective effect is enhanced by coadministration of pectin. However, the underlying mechanisms have not been fully elucidated. We hypothesized that fish oil with butyrate, a pectin fermentation product, protects against colon cancer initiation by decreasing cell proliferation and increasing differentiation and apoptosis through a p27(Kip1)-mediated mechanism. Rats were provided diets of corn or fish oil, with/without butyrate, and terminated 12, 24, or 48 hours after azoxymethane (AOM) injection. Proliferation (Ki-67), differentiation (Dolichos Biflorus Agglutinin), apoptosis (TUNEL), and p27(Kip1) (cell-cycle mediator) were measured in the same cell within crypts in order to examine the coordination of cell cycle as a function of diet. DNA damage (N(7)-methylguanine) was determined by quantitative IHC analysis. Dietary fish oil decreased DNA damage by 19% (P = 0.001) and proliferation by 50% (P = 0.003) and increased differentiation by 56% (P = 0.039) compared with corn oil. When combined with butyrate, fish oil enhanced apoptosis 24 hours after AOM injection compared with a corn oil/butyrate diet (P = 0.039). There was an inverse relationship between crypt height and apoptosis in the fish oil/butyrate group (r = -0.53, P = 0.040). The corn oil/butyrate group showed a positive correlation between p27(Kip1) expression and proliferation (r = 0.61, P = 0.035). These results indicate the in vivo effect of butyrate on apoptosis and proliferation is dependent on dietary lipid source. These results demonstrate the presence of an early coordinated colonocyte response by which fish oil and butyrate protects against colon tumorigenesis.


Apoptosis , Butyrates/administration & dosage , Colon/pathology , Colonic Neoplasms/prevention & control , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Fish Oils/administration & dosage , Animals , Azoxymethane/chemistry , Cell Differentiation , Cell Proliferation , Colonic Neoplasms/pathology , Corn Oil/administration & dosage , DNA Damage , Dietary Fats, Unsaturated/administration & dosage , Fermentation , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Ki-67 Antigen/metabolism , Male , Pectins/chemistry , Rats , Rats, Sprague-Dawley
11.
Cancer Prev Res (Phila) ; 8(11): 1093-101, 2015 Nov.
Article En | MEDLINE | ID: mdl-26304465

p21-activated kinase 1 (PAK1) is a serine/threonine kinase that is overexpressed in colorectal cancer. PAK1 is a target of mesalamine [5-aminosylicylic acid (5-ASA)], a common drug for the treatment of ulcerative colitis with prospective chemopreventive properties. Here, we investigated whether PAK1 deletion impedes tumorigenesis in murine intestinal cancer models. Ten-week-old APC(min) or APC(min)/PAK1(-/-) mice were monitored for 8 weeks, euthanized, and assessed for tumor number and size. Six- to 8-week-old PAK1(-/-) and wild-type (WT) mice received one 10 mg/kg intraperitoneal injection of azoxymethane (AOM) and four cycles of 1.7% dextran sodium sulfate (DSS) for 4 days followed by 14 days of regular water. Mice also received 5-ASA via diet. Tumor incidence and size was assessed via colonoscopy and pathology. Molecular targets of PAK1 and 5-ASA were evaluated via immunohistochemistry (IHC) in both models. PAK1 deletion reduced tumor multiplicity and tumor burden but did not alter average tumor size in APC(min) mice. IHC revealed that PAK1 deletion reduced p-AKT, ß-catenin, and c-Myc expression in APC(min) adenomas. Colonoscopy and pathologic analysis revealed that PAK1 deletion reduced tumor multiplicity without affecting tumor size in AOM/DSS-treated mice. 5-ASA treatment and PAK1 deletion impeded tumor multiplicity and dysplastic lesions in AOM/DSS mice. IHC further revealed that 5-ASA blocked ß-catenin signaling via inhibition of PAK1/p-AKT. These data indicate that PAK1 contributes to initiation of intestinal carcinogenesis.


Gene Expression Regulation, Neoplastic , Intestinal Neoplasms/metabolism , p21-Activated Kinases/metabolism , Animals , Azoxymethane/chemistry , Carcinogenesis , Colonoscopy , Dextrans/chemistry , Female , Gene Deletion , Genotype , Heterozygote , Immunohistochemistry , Inflammation , Male , Mesalamine/chemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , beta Catenin/metabolism
12.
Cancer Prev Res (Phila) ; 8(9): 786-95, 2015 Sep.
Article En | MEDLINE | ID: mdl-26069204

Despite widespread use as well as epidemiologic indications, there have been no investigations into the effect of St. John's wort (SJW) extract on colorectal carcinogenesis in vivo. This study reports a systematic evaluation of the impact of dietary supplementation of SJW extract on azoxymethane-induced colorectal carcinogenesis in mice. Mice were fed with either AIN-93G (control) diet or SJW extract-supplemented diet (SJW diet) prior to azoxymethane treatment. SJW diet was found to significantly improve the overall survival of azoxymethane-treated mice. Pretreatment with the SJW diet significantly reduced body weight loss as well as decrease of serum albumin and cholesterol levels associated with azoxymethane-induced colorectal tumorigenesis. SJW diet-fed mice showed a significant decrease in tumor multiplicity along with a decrease in incidence of large tumors and a trend toward decreased total tumor volume in a dose-dependent manner. A short-term study, which examined the effect of SJW prior to rectal bleeding, also showed decrease in colorectal polyps in SJW diet-fed mice. Nuclear factor kappa B (NF-κB) and extracellular signal-regulated kinase (ERK1/2) pathways were attenuated by SJW administration. SJW extract resulted in early and continuous attenuation of these pathways in the colon epithelium of SJW diet-fed mice under both short-term and long-term treatment regimens. In conclusion, this study demonstrated the chemopreventive potential of SJW extract against colorectal cancer through attenuation of proinflammatory processes.


Anticarcinogenic Agents/therapeutic use , Carcinogenesis/drug effects , Colorectal Neoplasms/prevention & control , Hypericum/chemistry , Inflammation/drug therapy , Plant Extracts/therapeutic use , Animals , Azoxymethane/chemistry , Cell Transformation, Neoplastic/drug effects , Colon/pathology , Colorectal Neoplasms/drug therapy , Diet , Dietary Supplements , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , Mice , NF-kappa B/metabolism , Oligonucleotide Array Sequence Analysis , Oligonucleotides/chemistry , Signal Transduction/drug effects
13.
Cancer Prev Res (Phila) ; 6(10): 1111-9, 2013 Oct.
Article En | MEDLINE | ID: mdl-23983085

Current fecal tests (occult blood, methylation, DNA mutations) target minute amounts of tumor products among a large amount of fecal material and thus have suboptimal performance. Our group has focused on exploiting field carcinogenesis as a modality to amplify the neoplastic signal. Specifically, we have shown that endoscopically normal rectal brushings have striking nano-architectural alterations which are detectable using a novel optical technique, partial wave spectroscopic microscopy (PWS). We therefore wished to translate this approach to a fecal assay. We examined mucus layer fecal colonocytes (MLFC) at preneoplastic and neoplastic time points (confirmed with rat colonoscopy) in the azoxymethane (AOM)-treated rat model and conducted PWS analysis to derive the nano-architectural parameter, disorder strength (Ld). We confirmed these results with studies in a genetic model (the Pirc rat). We showed that MLFC appeared microscopically normal, consistent with field carcinogenesis. Ld was elevated at an early time point (5 weeks post-AOM injection, effect size = 0.40, P = 0.024) and plateaued before adenoma formation (10 weeks post-AOM, effect size = 0.66, P = 0.001), with no dramatic increase once tumors developed. We replicated these data in the preneoplastic Pirc rat with an effect size in the MLFC that replicated the rectal brushings (increase vs. age-matched controls of 62% vs. 74%, respectively). We provide the first demonstration of a biophotonics approach to fecal assay. Furthermore, targeting the nano-architectural changes of field carcinogenesis rather than the detection of tumor products may provide a novel paradigm for colorectal cancer screening.


Adenoma/ultrastructure , Carcinogenesis , Colon/cytology , Colorectal Neoplasms/ultrastructure , Intestinal Mucosa/ultrastructure , Adenoma/pathology , Animals , Azoxymethane/chemistry , Colonoscopy , Colorectal Neoplasms/pathology , Disease Models, Animal , Early Detection of Cancer , Endoscopy , Feces , Intestinal Mucosa/pathology , Male , Mass Screening , Microscopy , Occult Blood , Optics and Photonics , Rats , Rats, Inbred F344
14.
Br J Nutr ; 109(4): 630-8, 2013 Feb 28.
Article En | MEDLINE | ID: mdl-23021249

Inflammatory bowel disease (IBD) is a risk factor for the development of colon cancer. Environmental factors including diet and the microflora influence disease outcome. Folate and homocysteine have been associated with IBD-mediated colon cancer but their roles remain unclear. We used a model of chemically induced ulcerative colitis (dextran sodium sulphate (DSS)) with or without the colon carcinogen azoxymethane (AOM) to determine the impact of dietary folic acid (FA) on colonic microflora and the development of colon tumours. Male mice (n 15 per group) were fed a FA-deficient (0 mg/kg), control (2 mg/kg) or FA-supplemented (8 mg/kg) diet for 12 weeks. Folate status was dependent on the diet (P< 0·001) and colitis-induced treatment (P= 0·04) such that mice with colitis had lower circulating folate. FA had a minimal effect on tumour initiation, growth and progression, although FA-containing diets tended to be associated with a higher tumour prevalence in DSS-treated mice (7-20 v. 0%, P= 0·08) and the development of more tumours in the distal colon of AOM-treated mice (13-83% increase, P= 0·09). Folate deficiency was associated with hyperhomocysteinaemia (P< 0·001) but homocysteine negatively correlated with tumour number (r - 0·58, P= 0·02) and load (r - 0·57, P= 0·02). FA had no effect on the intestinal microflora. The present data indicate that FA intake has no or little effect on IBD or IBD-mediated colon cancer in this model and that hyperhomocysteinaemia is a biomarker of dietary status and malabsorption rather than a cause of IBD-mediated colon cancer.


Diet , Folic Acid/chemistry , Inflammation/pathology , Microbiota , Neoplasms/prevention & control , Animals , Azoxymethane/chemistry , Biomarkers/metabolism , Colitis, Ulcerative/complications , Colitis, Ulcerative/microbiology , Colon/microbiology , Colonic Neoplasms/complications , Colonic Neoplasms/microbiology , Dextran Sulfate/chemistry , Dextrans/chemistry , Disease Progression , Male , Mice , Mice, Inbred C57BL , RNA, Ribosomal, 16S/genetics , Sulfates/chemistry
15.
PLoS One ; 7(7): e41132, 2012.
Article En | MEDLINE | ID: mdl-22815940

Sialic acids are acidic monosaccharides that bind to the sugar chains of glycoconjugates and change their conformation, intermolecular interactions, and/or half-life. Thus, sialidases are believed to modulate the function of sialoglycoconjugates by desialylation. We previously reported that the membrane-associated mammalian sialidase NEU3, which preferentially acts on gangliosides, is involved in cell differentiation, motility, and tumorigenesis. The NEU3 gene expression is aberrantly elevated in several human cancers, including colon, renal, prostate, and ovarian cancers. The small interfering RNA-mediated knock-down of NEU3 in cancer cell lines, but not in normal cell-derived primary cultures, downregulates EGFR signaling and induces apoptosis. Here, to investigate the physiological role of NEU3 in tumorigenesis, we established Neu3-deficient mice and then subjected them to carcinogen-induced tumorigenesis, using a sporadic and a colitis-associated colon cancer models. The Neu3-deficient mice showed no conspicuous accumulation of gangliosides in the brain or colon mucosa, or overt abnormalities in their growth, development, behavior, or fertility. In dimethylhydrazine-induced colon carcinogenesis, there were no differences in the incidence or growth of tumors between the Neu3-deficient and wild-type mice. On the other hand, the Neu3-deficient mice were less susceptible than wild-type mice to the colitis-associated colon carcinogenesis induced by azoxymethane and dextran sodium sulfate. These results suggest that NEU3 plays an important role in inflammation-dependent tumor development.


Colitis/etiology , Colitis/genetics , Colonic Neoplasms/etiology , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Neuraminidase/chemistry , Animals , Azoxymethane/chemistry , Cell Membrane/enzymology , Dextrans/chemistry , Gangliosides/chemistry , Gene Expression Profiling , Genetic Predisposition to Disease , Genetic Vectors , Genotype , Glycoconjugates/chemistry , Glycolipids/chemistry , Mice , Mice, Transgenic , Neuraminidase/genetics , Sialic Acids/chemistry , Sulfates/chemistry
16.
Anticancer Res ; 31(12): 4273-5, 2011 Dec.
Article En | MEDLINE | ID: mdl-22199291

BACKGROUND/AIM: The cancer stem cell model suggests that only a rare subpopulation, known as cancer stem cells (CSC) are responsible for tumor initiation. CSC from several human carcinomas are characterized by specific cell surface markers, such as CD133. The CD133 role in colon tumorigenesis remains controversial. MATERIALS AND METHODS: CD133 was evaluated by immunohistochemistry in a mouse model of colitis-related colon tumorigenesis induced by a combined treatment with azoxymethane (AOM) and dextran sodium sulphate (DSS). RESULTS: In normal tissue rare scattered positive cells were detectable at the bottom of the crypts. The percentage of positive cells significantly increased in dysplastic lesions and appeared to progressively decrease in the passage from dysplasia to adenoma and then to cancer, although always remaining greater in number than in the normal tissue. CONCLUSION: An increased CD133 expression occurs at early stages of colon tumorigenesis in the mouse. CD133-expressing cells might play an important role from the earlier phase and throughout the entire process of colon cancer development.


Antigens, CD/biosynthesis , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glycoproteins/biosynthesis , AC133 Antigen , Animals , Azoxymethane/chemistry , Carcinogens/pharmacology , Cell Line, Tumor , Colon/metabolism , Dextran Sulfate/chemistry , Mice , Mutation , Neoplastic Stem Cells/cytology , Peptides
17.
Acta cir. bras ; 26(2): 125-128, abr. 2011. tab
Article En | LILACS | ID: lil-579639

PURPOSE: To study the antitumor action of Tabebuia avellanedae in experimentally induced colon carcinogenesis by azoxymethane in mice. METHODS: Fifty (n=50) mice were divided into five groups: in group I azoxymethane (AOM) was administered, in Group II - β-lapachone, in group III - vehicle (diluent) and in group IV - vehicle + AOM and finally in group V - β-lapachone + AOM. RESULTS: It was observed the presence of aberrant crypt foci in all animals of groups I and IV, 50 percent in group II and 90 percent in group V. CONCLUSION: The β-lapachone extracted from the Tabebuia avellanedae showed no protective effect of lesions induced by azoxymethane in colon of mice.


OBJETIVO: Estudar a ação antitumoral da Tabebuia avellanedae (Ipê-Roxo) na carcinogênese colônica induzida experimentalmente pelo azoximetano em camundongos. MÉTODOS: Foram utilizados 50 camundongos divididos em 5 grupos: grupo I administrado Azoximetano (AOM); grupo II - β-lapachona; grupo III - veículo (diluente); grupo IV - veículo + AOM; e grupo V - β-lapachona + AOM. RESULTADOS: Observou-se presença de focos de criptas aberrantes em todos os animais dos grupos I e IV, 50 por cento no grupo II e 90 por cento no grupo V. CONCLUSÃO: A β-lapachona extraída da Tabebuia avellanedae não apresentou efeito protetor das lesões induzidas pelo azoximetano em cólon de camundongos.


Animals , Antineoplastic Agents/analysis , Mice/classification , Azoxymethane/chemistry , Tabebuia/classification
18.
Carcinogenesis ; 30(12): 2077-84, 2009 Dec.
Article En | MEDLINE | ID: mdl-19825969

We have hypothesized that dietary modulation of intestinal non-coding RNA [microRNA (miRNA)] expression may contribute to the chemoprotective effects of nutritional bioactives (fish oil and pectin). To fully understand the effects of these agents on the expression of miRNAs, Sprague-Dawley rats were fed diets containing corn oil or fish oil with pectin or cellulose and injected with azoxymethane (AOM, a colon-specific carcinogen) or saline (control). Real-time polymerase chain reaction using miRNA-specific primers and Taq Man probes was carried out to quantify effects on miRNA expression in colonic mucosa. From 368 mature miRNAs assayed, at an early stage of cancer progression (10 week post AOM injection), let-7d, miR-15b, miR-107, miR-191 and miR-324-5p were significantly (P < 0.05) affected by diet x carcinogen interactions. Overall, fish oil fed animals exhibited the smallest number of differentially expressed miRNAs (AOM versus saline treatment). With respect to the tumor stage (34 week post AOM injection), 46 miRNAs were dysregulated in adenocarcinomas compared with normal mucosa from saline-injected animals. Of the 27 miRNAs expressed at higher (P < 0.05) levels in tumors, miR-34a, 132, 223 and 224 were overexpressed at >10-fold. In contrast, the expression levels of miR-192, 194, 215 and 375 were dramatically reduced (< or = 0.32-fold) in adenocarcinomas. These results demonstrate for the first time the utility of the rat AOM model and the novel role of fish oil in protecting the colon from carcinogen-induced miRNA dysregulation.


Carcinogens , Colon/metabolism , Fatty Acids, Unsaturated/metabolism , MicroRNAs/metabolism , Adenocarcinoma/chemically induced , Animals , Azoxymethane/chemistry , Colonic Neoplasms/chemically induced , Epigenesis, Genetic , Fish Oils , Gene Expression Profiling , Male , Pectins/chemistry , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
19.
Nat Protoc ; 2(8): 1998-2004, 2007.
Article En | MEDLINE | ID: mdl-17703211

Colorectal cancer is a life-threatening disease that can develop spontaneously or as a complication of inflammatory bowel diseases. Mouse models are essential tools for the preclinical testing of novel therapeutic options in vivo. Here, we provide a highly reliable protocol for an experimental mouse model to study the development of colon cancers. It is based on the mutagenic agent azoxymethane (AOM), which exerts colonotropic carcinogenicity. Repeated intraperitoneal administration of AOM results in the development of spontaneous tumors within 30 weeks. As an alternative option, inflammation-dependent tumor growth can be investigated by combining the administration of AOM with the inflammatory agent dextran sodium sulfate in drinking water, which causes rapid growth of multiple colon tumors per mouse within 10 weeks. Different scoring systems including number of tumors and tumor size identify factors promoting or inhibiting tumor initiation and/or tumor progression, respectively.


Colonic Neoplasms/chemically induced , Disease Models, Animal , Mice , Animals , Azoxymethane/chemistry , Carcinogens/chemistry , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Dextran Sulfate , Disease Progression , Inflammation Mediators , Mutagens/chemistry
20.
Carcinogenesis ; 18(5): 949-55, 1997 May.
Article En | MEDLINE | ID: mdl-9163680

The 80%-ethanol extract of lemon grass (Cymbopogon citratus Stapf), a medicinal plant in Thailand, has been reported to be antimutagenic against various known mutagens in the Salmonella mutation assay. To investigate chemoprevention in an animal carcinogenesis model, we examined inhibitory effects of the lemon grass extract on the formation of azoxymethane (AOM)-induced DNA adducts and aberrant crypt foci (ACF) in the rat colon. One week after the start of the treatment with lemon grass extract at doses of 0.5 or 5 g/kg body wt by gavage, F344 rats received two s.c. injections of 15 mg of AOM per kg body weight at 1 week apart. For DNA adduct analysis of the colon and liver, the rats were killed 12 h after the second AOM injection. The DNA from the liver and colon were used for O6-methylguanine and N7-methylguanine analysis. For ACF analysis in the initiation stage, AOM-injected rats were continuously treated with lemon grass extract and were killed 3 weeks after the second AOM injection. For analysis in the promotion stage the treatment with the lemon grass extract (0.5 g/kg) started 2 weeks after the second AOM injection and continued for 12 weeks until the animals were killed. Lemon grass treatment significantly inhibited DNA adduct formation in both the colonic mucosa and the muscular layer but not in the liver. In addition, lemon grass extract treatment significantly inhibited ACF formation in both the initiation stage and the promotion stage. Especially in the promotion stage, lemon grass treatment inhibited the formation of larger ACF (with four or more crypts per focus), which was predictive of tumor incidence. Furthermore, lemon grass extract inhibited fecal beta-glucuronidase competitively and had antioxidant activity. These results suggest that the lemon grass extract inhibits the release of activated aglycon, methylazoxymethanol, from a glucuronide conjugate in the colon, and decreases the DNA adducts and ACF formation in the rat colon.


Antimutagenic Agents/pharmacology , Azoxymethane/chemistry , Colorectal Neoplasms/chemically induced , DNA Adducts/chemistry , Plant Extracts/pharmacology , Plants, Medicinal , Animals , Colon/chemistry , Colon/drug effects , Colon/enzymology , Colon/pathology , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Glucuronidase/metabolism , Male , Poaceae/chemistry , Rats , Rats, Inbred F344 , Thailand
...