Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Int J Mol Sci ; 15(5): 9160-72, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24857924

ABSTRACT

Activation of peroxisome proliferator-activated receptor (PPAR) α disrupts growth-related activities in a variety of human cancers. This study was designed to determine whether fenofibrate, a PPARα agonist, can suppress 4-nitroquinoline 1-oxide (4-NQO)-induced proliferative lesions in the lung of obese hyperlipidemic mice. Male Tsumura Suzuki Obese Diabetic mice were subcutaneously injected with 4-NQO to induce lung proliferative lesions, including adenocarcinomas. They were then fed a diet containing 0.01% or 0.05% fenofibrate for 29 weeks, starting 1 week after 4-NQO administration. At week 30, the incidence and multiplicity (number of lesions/mouse) of pulmonary proliferative lesions were lower in mice treated with 4-NQO and both doses of fenofibrate compared with those in mice treated with 4-NQO alone. The incidence and multiplicity of lesions were significantly lower in mice treated with 4-NQO and 0.05% fenofibrate compared with those in mice treated with 4-NQO alone (p<0.05). Both doses of fenofibrate significantly reduced the proliferative activity of the lesions in 4-NQO-treated mice (p<0.05). Fenofibrate also significantly reduced the serum insulin and insulin-like growth factor (IGF)-1 levels, and decreased the immunohistochemical expression of IGF-1 receptor (IGF-1R), phosphorylated Akt, and phosphorylated Erk1/2 in lung adenocarcinomas. Our results indicate that fenofibrate can prevent the development of 4-NQO-induced proliferative lesions in the lung by modulating the insulin-IGF axis.


Subject(s)
Fenofibrate/pharmacology , Hypolipidemic Agents/pharmacology , PPAR alpha/agonists , Pulmonary Alveoli/drug effects , Animals , Cell Proliferation/drug effects , Fenofibrate/therapeutic use , Hyperlipidemias/drug therapy , Hyperlipidemias/metabolism , Hyperlipidemias/pathology , Hypolipidemic Agents/therapeutic use , Insulin/blood , Insulin-Like Growth Factor I/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice , Mice, Obese , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , PPAR alpha/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Receptor, IGF Type 1/metabolism
2.
BMC Cancer ; 12: 448, 2012 Oct 03.
Article in English | MEDLINE | ID: mdl-23031214

ABSTRACT

BACKGROUND: Chemotherapeutic bioassay for colorectal cancer (CRC) with a rat model bearing chemically-induced CRCs plays an important role in the development of new anti-tumor drugs and regimens. Although several protocols to induce CRCs have been developed, the incidence and number of CRCs are not much enough for the efficient bioassay. Recently, we established the very efficient system to induce CRCs with a chemically induced-colon carcinogenesis-prone Apc-mutant rat, Kyoto Apc Delta (KAD) rat. Here, we applied the KAD rat to the chemotherapeutic bioassay for CRC and showed the utility of the KAD rat. METHODS: The KAD rat has been developed by the ENU mutagenesis and carries a homozygous nonsense mutation in the Apc gene (S2523X). Male KAD rats were given a single subcutaneous injection of AOM (20 mg/kg body weight) at 5 weeks of age. Starting at 1 week after the AOM injection, they were given 2% DSS in drinking water for 7 days. Tumor-bearing KAD rats were divided into experimental and control groups on the basis of the number of tumors observed by endoscopy at week 8. The 5-fluorouracil (5-FU) was administrated intravenously a dose of 50 or 75 mg/kg weekly at week 9, 10, and 11. After one-week interval, the 5-FU was given again at week 13, 14, and 15. At week 16, animals were sacrificed and tumor number and volume were measured macroscopically and microscopically. RESULTS: In total 48 tumors were observed in 27 KAD rats with a 100% incidence at week 8. The maximum tolerated dose for the KAD rat was 50 mg/kg of 5-FU. Macroscopically, the number or volume of tumors in the 5-FU treated rats was not significantly different from the control. Microscopically, the number of adenocarcinoma in the 5-FU treated rats was not significantly different (p < 0.02) from that of the control. However, the volume of adenocarcinomas was significantly lower than in the control. Anticancer effect of the 5-FU could be obtained only after the 16 weeks of experimental period. CONCLUSION: The use of the AOM/DSS-treated tumor-bearing KAD rats could shorten the experimental period and reduce the number of animals examined in the chemotherapeutic bioassay. The efficient bioassay with the AOM/DSS-treated tumor-bearing KAD rats would promote the development of new anti-tumor drugs and regimens.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Fluorouracil/pharmacology , Mutation , Adenocarcinoma/chemically induced , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Animals , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacology , Azoxymethane , Colon/drug effects , Colon/pathology , Colonic Neoplasms/chemically induced , Dextran Sulfate , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Screening Assays, Antitumor/methods , Fluorouracil/administration & dosage , Injections, Intravenous , Male , Rats , Reproducibility of Results
3.
Psychiatry Res ; 198(2): 194-201, 2012 Jul 30.
Article in English | MEDLINE | ID: mdl-22421064

ABSTRACT

We examined whether augmentation with olanzapine would be superior to increased risperidone dose among acute schizophrenia patients showing early non-response to risperidone. We performed a rater-blinded, randomized controlled trial at psychiatric emergency sites. Eligible patients were newly admitted patients with acute schizophrenia. Early response was defined as Clinical Global Impressions-Improvement Scale score ≤3 following 2 weeks of treatment. Early non-responders were allocated to receive either augmentation with olanzapine (RIS+OLZ group) or increased risperidone dose (RIS+RIS group). The 78 patients who completed 2 weeks of treatment were divided into 52 early responders to risperidone and 26 early non-responders to risperidone (RIS+OLZ group, n=13; RIS+RIS group, n=13). No difference in the achievement of ≥50% improvement in Positive and Negative Syndrome Scale total score was observed between RIS+OLZ and RIS+RIS groups. Although time to treatment discontinuation for any cause was significantly shorter in the RIS+RIS group (6.8 weeks [95% confidence interval, 5.2-8.4]) than in early responders to risperidone (8.6 weeks [7.9-9.3]; P=0.018), there was no significant difference between the RIS+OLZ group (7.9 weeks [6.3-9.5]) and early responders to risperidone. Secondary outcomes justify the inclusion of augmentation arms in additional, larger studies comparing strategies for early non-responders.


Subject(s)
Benzodiazepines/administration & dosage , Drug Resistance/drug effects , Drug Therapy, Combination/psychology , Risperidone/therapeutic use , Schizophrenia/drug therapy , Adult , Antipsychotic Agents/administration & dosage , Antipsychotic Agents/therapeutic use , Benzodiazepines/therapeutic use , Female , Humans , Male , Olanzapine , Psychiatric Status Rating Scales/statistics & numerical data , Single-Blind Method
4.
Carcinogenesis ; 33(3): 702-7, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22223845

ABSTRACT

Obese people and diabetic patients are known to be high risk of colorectal cancer (CRC), suggesting need of a new preclinical animal model, by which to extensively study the diverse mechanisms, therapy and prevention. The present study aimed to determine whether experimental obese and diabetic mice produced by monosodium glutamate (MSG) treatment are susceptible to azoxymethane (AOM)-induced colon tumorigenesis using early biomarkers, aberrant crypts foci (ACF) and ß-catenin-accumulated crypts (BCACs), of colorectal carcinogenesis. Male Crj:CD-1 (ICR) newborns were daily given four subcutaneous injections of MSG (2 mg/g body wt) to induce diabetes and obesity. They were then given four intraperitoneal injections of AOM (15 mg/kg body wt) or saline (0.1 ml saline/10 g body wt). Ten weeks after the last injection of AOM, the MSG-AOM mice had a significant increase in the multiplicity of BCAC (13.83 ± 7.44, P < 0.002), but not ACF (78.00 ± 11.20), when compare to the Saline-AOM mice (5.45 ± 1.86 of BCAC and 69.27 ± 8.06 of ACF). Serum biochemical profile of the MSG-treated mice with or without AOM showed hyperinsulinemia, hypercholesteremia and hyperglycemia. The mRNA expression of insulin-like growth factor-1 receptor (IGF-1R, P<0.01) was increased in the MSG-AOM mice, when compared with the mice given AOM alone. IGF-1R was immunohistochemically expressed in the BCAC, but not ACF, in the AOM-treated mice. Our findings suggest that the MSG mice are highly susceptible to AOM-induced colorectal carcinogenesis, suggesting potential utility of our MSG-AOM mice for further investigation of the possible underlying events that affect the positive association between obese/diabetes and CRC.


Subject(s)
Colonic Neoplasms/chemically induced , Diabetes Mellitus, Experimental/complications , Disease Models, Animal , Animals , Azoxymethane/administration & dosage , Azoxymethane/pharmacology , Colonic Neoplasms/complications , Diabetes Mellitus, Experimental/chemically induced , Disease Susceptibility , Hypercholesterolemia , Hyperglycemia , Hyperinsulinism , Insulin-Like Growth Factor I , Male , Mice , Mice, Inbred ICR , Precancerous Conditions/chemically induced , RNA, Messenger/biosynthesis , Receptor, IGF Type 1/biosynthesis , Receptor, IGF Type 1/genetics , Sodium Glutamate/administration & dosage , Sodium Glutamate/pharmacology
5.
Int J Mol Sci ; 12(11): 8133-45, 2011.
Article in English | MEDLINE | ID: mdl-22174655

ABSTRACT

The numbers of obese people and diabetic patients are ever increasing. Obesity and diabetes are high-risk conditions for chronic diseases, including certain types of cancer, such as colorectal cancer (CRC). The aim of this study was to develop a novel animal model in order to clarify the pathobiology of CRC development in obese and diabetic patients. We developed an animal model of obesity and colorectal cancer by breeding the C57BL/KsJ-db/db (db/db) mouse, an animal model of obesity and type II diabetes, and the C57BL/6J-Apc(Min/+) (Min/+) mouse, a model of familial adenomatous polyposis. At 15 weeks of age, the N9 backcross generation of C57BL/KsJ-db/db-Apc(Min/+) (db/db-Min/+) mice developed an increased incidence and multiplicity of adenomas in the intestinal tract when compared to the db/m-Min/+ and m/m-Min/+ mice. Blood biochemical profile showed significant increases in insulin (8.3-fold to 11.7-fold), cholesterol (1.2-fold to 1.7-fold), and triglyceride (1.2-fold to 1.3-fold) in the db/db-Min/+ mice, when compared to those of the db/m-Min/+ and m/m-Min/+ mice. Increases (1.4-fold to 2.6-fold) in RNA levels of insulin-like growth factor (IGF)-1, IRF-1R, and IGF-2 were also observed in the db/db- Min/+ mice. These results suggested that the IGFs, as well as hyperlipidemia and hyperinsulinemia, promoted adenoma formation in the db/db-Min/+ mice. Our results thus suggested that the db/db-Min/+ mice should be invaluable for studies on the pathogenesis of CRC in obese and diabetes patients and the therapy and prevention of CRC in these patients.


Subject(s)
Colorectal Neoplasms/pathology , Disease Models, Animal , Obesity/pathology , Animals , Blood Glucose/metabolism , Cholesterol/blood , Colorectal Neoplasms/etiology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/pathology , Female , Insulin/blood , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/complications , RNA, Messenger/genetics , RNA, Messenger/metabolism , Risk Factors , Triglycerides/blood
6.
Cancer Sci ; 102(9): 1659-64, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21668584

ABSTRACT

Colon carcinogenesis is a multistage process involving genetic alterations of various tumor suppressor genes and oncogenes. Repressor element 1 silencing factor (REST ), which was originally discovered as a transcriptional repressor of neuronal genes, plays an important role in neuronal differentiation. In a previous genetic screening for tumor suppressor genes in human cancers, REST was identified as a candidate tumor suppressor gene in colorectal carcinogenesis. However, the role of Rest in colon carcinogenesis in vivo remains unclear because of the embryonic lethal phenotype of the conventional Rest knockout mouse. In the present study, we conditionally deleted the Rest gene in the intestinal epithelium and investigated the effect of Rest ablation in mouse colon tumorigenesis. A conditional ablation of Rest in the colonic crypts led to a rapid upregulation of Rest-targeted genes, such as Syt4, Bdnf, and Tubb3, suggesting that Rest actually suppresses the expression of its target genes in the colon. However, Rest ablation did not lead to any significant effect on the development of colon tumors in two independent mouse models of colon carcinogenesis. In addition, despite the upregulation of neuronal genes in the colonic crypts, no neuronal differentiation was observed in the colonic crypts and tumors after the Rest ablation. These results indicate that the loss of Rest expression by itself does not promote the development of colon tumors in mice, and suggest that REST may exert a tumor suppressing activity in conjunction with the additional genetic/epigenetic abnormalities that occur during colon carcinogenesis.


Subject(s)
Colonic Neoplasms/genetics , Genes, Tumor Suppressor , Repressor Proteins/genetics , Animals , Female , Gene Deletion , Intestinal Mucosa/metabolism , Mice
7.
EMBO Mol Med ; 3(6): 320-33, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21542132

ABSTRACT

The stratum corneum (SC), the outermost layer of the epidermis, acts as a barrier against the external environment. It is hydrated by endogenous humectants to avoid desiccation. However, the molecular mechanisms of SC hydration remain unclear. We report that skin-specific retroviral-like aspartic protease (SASPase) deficiency in hairless mice resulted in dry skin and a thicker and less hydrated SC with an accumulation of aberrantly processed profilaggrin, a marked decrease of filaggrin, but no alteration in free amino acid composition, compared with control hairless mice. We demonstrated that recombinant SASPase directly cleaved a linker peptide of recombinant profilaggrin. Furthermore, missense mutations were detected in 5 of 196 atopic dermatitis (AD) patients and 2 of 28 normal individuals. Among these, the V243A mutation induced complete absence of protease activity in vitro, while the V187I mutation induced a marked decrease in its activity. These findings indicate that SASPase activity is indispensable for processing profilaggrin and maintaining the texture and hydration of the SC. This provides a novel approach for elucidating the complex pathophysiology of atopic dry skin.


Subject(s)
Aspartic Acid Endopeptidases/metabolism , Epidermis/physiology , Intermediate Filament Proteins/metabolism , Protein Precursors/metabolism , Amino Acid Substitution , Animals , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/physiology , Dermatitis, Atopic/enzymology , Dermatitis, Atopic/genetics , Epidermis/enzymology , Filaggrin Proteins , Humans , Intermediate Filament Proteins/genetics , Mice , Mice, Hairless , Mutation, Missense , Protein Precursors/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Water/metabolism
8.
Carcinogenesis ; 31(9): 1627-33, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20622006

ABSTRACT

Global DNA hypomethylation and concomitant site-specific gene hypermethylation are among the most common molecular alterations in human neoplasia. Although site-specific DNA hypermethylation has been shown to be associated with the development of various tumors accompanied by transcriptional silencing of target genes, the functional significance of global DNA hypomethylation in tumorigenesis remains unclear. Previous studies have revealed that a genetic reduction of the DNA methylation levels leads to opposing effects on tumor development, depending on the tumor cell type and the stage of tumorigenesis. In the present study, we investigated the effect of DNA hypomethylation on gastric carcinogenesis in mice. The genetic reduction of DNA methylation levels suppressed the incidence, number and size of gastric tumors in two different mouse models for gastric tumorigenesis: the N-methyl-N-nitrosourea-induced model and the Apc(Min/+) mouse model that spontaneously develops gastric tumors with aging. Histological analyses revealed DNA hypomethylation to completely inhibit the development of invasive gastric tumors. These findings indicate that the reduction of DNA methylation levels suppresses gastric carcinogenesis and suggest that DNA methylation is closely associated with gastric tumorigenesis.


Subject(s)
DNA Methylation , Genes, APC/physiology , Methylnitrosourea/toxicity , Stomach Neoplasms/prevention & control , Animals , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/physiology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Stomach Neoplasms/genetics
9.
Cancer Sci ; 101(7): 1701-7, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20398056

ABSTRACT

Obesity and related metabolic abnormalities are risk factors for colorectal cancer. A state of chronic inflammation and adipocytokine imbalance may play a role in colorectal carcinogenesis. Statins, which are commonly used for the treatment of hyperlipidemia, are known to possess anti-inflammatory effects. Statins also exert chemopreventive properties against various cancers. The present study examined the effects of pitavastatin, a recently developed lipophilic statin, on the development of azoxymethane (AOM)-initiated colonic premalignant lesions in C57BL/KsJ-db/db (db/db) obese mice. Male db/db mice were administrated weekly subcutaneous injections of AOM (15 mg/kg body weight) for 4 weeks and then were subsequently fed a diet containing 1 ppm or 10 ppm pitavastatin for 8 weeks. Feeding with either dose of pitavastatin significantly reduced the number of colonic premalignant lesions, beta-catenin accumulated crypts, by inhibiting proliferation and the surrounding inflammation. Pitavastatin increased the serum levels of adiponectin while conversely decreasing the serum levels of total cholesterol, tumor necrosis factor-alpha (TNF-alpha), interleukin (IL)-6, IL-18, and leptin. Pitavastatin also caused a significant increase in the expression of phosphorylated form of the AMP-activated kinase (AMPK) protein on the colonic mucosa of AOM-treated mice. In addition, the expression levels of TNF-alpha, IL-6, IL-18, and COX-2 mRNAs on the colonic mucosa of AOM-treated mice were decreased by treatment with this agent. These findings suggest that pitavastatin attenuates chronic inflammation and improves the imbalance of adipocytokines, both of which are caused by the presence of excess adipose tissues, thereby preventing the development of colonic premalignancies in an obesity-related colon cancer model. Therefore, some types of statins, including pitavastatin, may be a useful chemoprevention modality for colon cancer in obese individuals.


Subject(s)
Azoxymethane/toxicity , Colonic Neoplasms/prevention & control , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Precancerous Conditions/prevention & control , Quinolines/therapeutic use , Animals , Carcinogens/toxicity , Cell Division/drug effects , Colonic Neoplasms/chemically induced , Colonic Neoplasms/complications , Colonic Neoplasms/pathology , Cytokines/drug effects , Cytokines/metabolism , Humans , Inflammation/prevention & control , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/complications , Obesity/drug therapy , Precancerous Conditions/chemically induced , Precancerous Conditions/pathology
10.
Clin Cancer Res ; 15(9): 3068-75, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19366832

ABSTRACT

PURPOSE: Obesity and related metabolic abnormalities, including insulin resistance and activation of the insulin-like growth factor (IGF)/IGF-I receptor (IGF-IR) axis, are risk factors for colon cancer. Supplementation with branched-chain amino acids (BCAA) reduces the risk of liver cancer in cirrhotic patients who are obese, and this has been associated with an improvement of insulin resistance. The present study examined the effects of BCAA on the development of azoxymethane (AOM)-initiated colonic premalignant lesions in C57BL/KsJ-db/db (db/db) mice that were obese and had hyperinsulinemia. EXPERIMENTAL DESIGN: Male db/db mice were given 4 weekly s.c. injections of AOM (15 mg/kg of body weight) and then they were fed a diet containing 3.0% BCAA or casein, a nitrogenc content-matched control diet, for 7 weeks. RESULTS: Feeding with BCAA caused a significant reduction in the number of total aberrant crypt foci and beta-catenin accumulated crypts, both of which are premalignant lesions of the colon, compared with the control diet-fed groups. BCAA supplementation caused a marked decrease in the expression of IGF-IR, the phosphorylated form of IGF-IR, phosphorylated glycogen synthase kinase 3beta, phosphorylated Akt, and cyclooxygenase-2 proteins on the colonic mucosa of AOM-treated mice. The serum levels of insulin, IGF-I, IGF-II, triglyceride, total cholesterol, and leptin were also decreased by supplementation with BCAA. CONCLUSION: BCAA supplementation in diet improves insulin resistance and inhibits the activation of the IGF/IGF-IR axis, thereby preventing the development of colonic premalignancies in an obesity-related colon cancer model that was also associated with hyperlipidemia and hyperinsulinemia. BCAA, therefore, may be a useful chemoprevention modality for colon cancer in obese people.


Subject(s)
Amino Acids, Branched-Chain/therapeutic use , Azoxymethane/toxicity , Carcinogens/toxicity , Colonic Neoplasms/prevention & control , Diabetes Mellitus, Experimental/drug therapy , Obesity/drug therapy , Precancerous Conditions/prevention & control , Animals , Body Weight/drug effects , Colonic Neoplasms/chemically induced , Colonic Neoplasms/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Dietary Supplements , Insulin/blood , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor II/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Obesity/metabolism , Obesity/pathology , Organ Size/drug effects , Precancerous Conditions/chemically induced , Precancerous Conditions/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Receptor, IGF Type 1/metabolism , Receptor, IGF Type 2/metabolism , Tumor Cells, Cultured , beta Catenin/metabolism
11.
Carcinogenesis ; 29(3): 666-72, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18204079

ABSTRACT

Apc(Min/+) mouse, a mouse model for human familial adenomatosis polyposis, contains a truncating mutation in the Apc gene and spontaneously develops intestinal tumors. Our previous study revealed two distinct stages of tumorigenesis in the colon of Apc(Min/+) mouse: microadenomas and macroscopic tumors. Microadenomas already have lost their remaining allele of the Apc and all microadenomas show accumulation of beta-catenin, indicating that activation of the canonical Wnt pathway is an initiating event in the tumorigenesis. This study shows that expression of nuclear beta-catenin in macroscopic tumors is further upregulated in comparison with that in microadenomas. Furthermore, transcriptional activity of beta-catenin/T-cell factor (Tcf) signaling, assessed using beta-catenin/Tcf reporter transgenic mice, is higher in the macroscopic tumors than that in microadenomas. In addition, the expression level of Dickkopf-1, which is known to be a negative modifier of the canonical Wnt pathway, was reduced only in colon tumors. These results suggest that activation of beta-catenin/Tcf transcription plays a role not only in the initiation stage but also in the promotion stage of colon carcinogenesis in Apc(Min/+) mice.


Subject(s)
Colonic Neoplasms/pathology , T Cell Transcription Factor 1/genetics , Transcription, Genetic , Up-Regulation , beta Catenin/genetics , Animals , Colonic Neoplasms/genetics , Flow Cytometry , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Mutant Strains , Reverse Transcriptase Polymerase Chain Reaction , Wnt Proteins/antagonists & inhibitors
12.
Cancer Prev Res (Phila) ; 1(4): 298-304, 2008 Sep.
Article in English | MEDLINE | ID: mdl-19138973

ABSTRACT

Obesity and diabetes mellitus are risk factors for colon cancer. The activation of the insulin-like growth factor (IGF)/IGF-IR axis plays a critical role in this carcinogenesis. (-)-Epigallocatechin gallate (EGCG), the major constituent of green tea, seems to have both antiobesity and antidiabetic effects. This study examined the effects of EGCG on the development of azoxymethane-induced colonic premalignant lesions in C57BL/KsJ-db/db (db/db) mice, which are obese and develop diabetes mellitus. Male db/db mice were given four weekly s.c. injections of azoxymethane (15 mg/kg body weight) and then they received drinking water containing 0.01% or 0.1% EGCG for 7 weeks. At sacrifice, drinking water with EGCG caused a significant decrease in the number of total aberrant crypt foci, large aberrant crypt foci, and beta-catenin accumulated crypts in these mice, all of which are premalignant lesions of the colon. The colonic mucosa of db/db mice expressed high levels of the IGF-IR, phosphorylated form of IGF-IR (p-IGF-IR), p-GSK-3beta, beta-catenin, cyclooxygenase-2, and cyclin D1 proteins, and EGCG in drinking water caused a marked decrease in the expression of these proteins. Treating these mice with EGCG also caused an increase in the serum level of IGFBP-3 while conversely decreasing the serum levels of IGF-I, insulin, triglyceride, cholesterol, and leptin. EGCG overcomes the activation of the IGF/IGF-IR axis, thereby inhibiting the development of colonic premalignant lesions in an obesity-related colon cancer model, which was also associated with hyperlipidemia, hyperinsulinemia, and hyperleptinemia. EGCG may be, therefore, useful in the chemoprevention or treatment of obesity-related colorectal cancer.


Subject(s)
Azoxymethane/toxicity , Carcinoma/prevention & control , Catechin/analogs & derivatives , Colonic Neoplasms/prevention & control , Precancerous Conditions/chemically induced , Animals , Anticarcinogenic Agents/pharmacology , Anticarcinogenic Agents/therapeutic use , Carcinoma/chemically induced , Carcinoma/metabolism , Catechin/pharmacology , Catechin/therapeutic use , Cholesterol/blood , Colonic Neoplasms/chemically induced , Colonic Neoplasms/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Drug Evaluation, Preclinical , Insulin/blood , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Leptin/blood , Male , Mice , Mice, Inbred C57BL , Precancerous Conditions/blood , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Receptor, IGF Type 1/metabolism , Triglycerides/blood
13.
Cancer Res ; 67(9): 4079-87, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17483318

ABSTRACT

Although several lines of evidence suggest the involvement of the Wnt pathway in the development of gastric cancers, the functional significance of the pathway in gastric carcinogenesis is still poorly defined. To examine the role of the Apc/beta-catenin signaling pathway in the development of gastric cancers, we investigated the gastric mucosa of the Apc(Min/+) mouse, which is a murine model for familial adenomatous polyposis, carrying a germ-line mutation at codon 850 of Apc. We found that aged Apc(Min/+) mice spontaneously develop multiple tumors in the stomach, which are accompanied by loss of heterozygosity of Apc. Such tumors consisted of adenomatous glands with strong nuclear accumulation of beta-catenin. Even a single adenomatous gland already showed nuclear accumulation of beta-catenin, suggesting that Apc/beta-catenin pathway is an initiating event in gastric tumorigenesis in Apc(Min/+) mice. Myc and cyclin D1 expressions, which are transcriptional targets of beta-catenin/Tcf, increased in the adenomatous lesions. Furthermore, beta-catenin/Tcf reporter transgenic mice with Apc(Min) allele showed higher levels of the transcriptional activity of beta-catenin/Tcf in the gastric tumors. We also treated Apc(Min/+) and wild-type mice with N-methyl-N-nitrosourea (MNU), an alkylating agent that induces adenomas and adenocarcinomas in the stomach. Consequently, MNU-treated Apc(Min/+) mice significantly enhanced the tumor development in comparison with Apc(Min/+) mice or MNU-treated wild-type mice. Several gastric tumors in MNU-treated Apc(Min/+) mice showed invasion into the submucosal layer. These results indicate that the Apc/beta-catenin pathway may play an important role in at least subset of gastric carcinomas. In addition, Apc(Min/+) mice combined with MNU could be a useful short-term model to investigate multistage carcinogenesis in the stomach.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , T Cell Transcription Factor 1/metabolism , beta Catenin/metabolism , Adenocarcinoma/pathology , Animals , Cell Nucleus/metabolism , Female , Gene Expression , Genes, APC , Loss of Heterozygosity , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Invasiveness , Signal Transduction , Stomach Neoplasms/pathology , Transcription, Genetic
14.
Oncol Rep ; 17(2): 297-304, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17203164

ABSTRACT

Recent epidemiological studies have indicated that high dietary consumption of fruit and vegetables results in lower risk of bladder cancer. To confirm these findings, we investigated in the current study the effects of dietary administration with beta-cryptoxanthin extracted from Citras unshiu oranges on N-butyl-N-(4-hydroxybutyl)nitrosamine (OH-BBN)-induced urinary bladder carcinogenesis in mice. Male ICR mice were divided into 6 experimental and control groups. Groups 1 through 4 were given OH-BBN (500 ppm) in drinking water for 6 weeks to induced urinary bladder neoplasms. Mice in groups 2, 3 and 4 were fed the diets mixed with 1, 5 and 25 ppm of beta-cryptoxanthin, respectively, starting 1 week after the cessation of OH-BBN exposure, and kept on these diets for 24 weeks until the termination of the study. Group 5 was treated with the diet containing the test compound (25 ppm) alone, and group 6 served as an untreated control. All animals were sacrificed at week 32 for histopathology and immunohistochemistry (cyclin D1). Feeding with beta-cryptoxanthin decreased the incidence and multiplicity of preneoplastic and neoplastic lesions of urinary bladder. Notably, the highest dose (25 ppm) of the test chemical significantly lowered the occurrence of bladder carcinoma, in conjunction with reducing the cyclin D1-positive cell ratio. These findings suggest that beta-cryptoxanthin is able to prevent OH-BBN-induced bladder carcinogenesis in mice.


Subject(s)
Anticarcinogenic Agents/pharmacology , Butylhydroxybutylnitrosamine , Carcinogens , Urinary Bladder Neoplasms/chemically induced , Urinary Bladder Neoplasms/drug therapy , Xanthophylls/pharmacology , Animals , Cryptoxanthins , Cyclin D1/biosynthesis , Male , Mice , Mice, Inbred ICR , Urinary Bladder Neoplasms/prevention & control
15.
Cancer Lett ; 246(1-2): 63-8, 2007 Feb 08.
Article in English | MEDLINE | ID: mdl-16517058

ABSTRACT

Sesame, which has been reported to have preventive effects against various disordered conditions, contains small quantities of lignans and several precursors to them such as sesaminol glucosides (SG). The lignans have the potent antioxidative activity and are suggested to have chemopreventive property. In the present study, we evaluated the modulating effect of SG on the development of colon precancerous lesions, aberrant crypt foci (ACF) and beta-catenin-accumulated crypts (BCAC), in the azoxymethane (AOM)-induced short-term model using male F344 rats. Dietary SG (500 ppm) significantly decreased the incidence of AOM-induced ACF when compared to the control (P<0.01). The incidences of AOM-induced BCAC in the SG-treated groups (250 or 500 ppm) were also significantly lower than that of the control group (P<0.01). Interestingly, administration of 500 ppm SG clearly decreased serum triglyceride level and mRNA expression of intestinal fatty acid-binding protein in the colonic mucosa, as compared to the control. These findings indicate that dietary SG inhibits AOM-induced carcinogenesis and suggest SG as a possible chemopreventive agent.


Subject(s)
Colonic Neoplasms/prevention & control , Dietary Supplements , Dioxoles/therapeutic use , Furans/therapeutic use , Glucosides/therapeutic use , Precancerous Conditions/prevention & control , Animals , Azoxymethane , Colon/drug effects , Colon/metabolism , Colon/pathology , Colonic Neoplasms/chemically induced , Dioxoles/administration & dosage , Dioxoles/chemistry , Fatty Acid-Binding Proteins/genetics , Furans/administration & dosage , Furans/chemistry , Gene Expression/drug effects , Glucosides/administration & dosage , Glucosides/chemistry , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Molecular Structure , Precancerous Conditions/chemically induced , Precancerous Conditions/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Reverse Transcriptase Polymerase Chain Reaction , Sesamum/chemistry , Triglycerides/blood
16.
Int J Cancer ; 120(2): 252-8, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17066427

ABSTRACT

The modulatory effects of dietary citrus unshiu segment membrane (CUSM) on the occurrence of aberrant crypt foci (ACF) and beta-catenin accumulated crypts (BCACs) were determined in male C57BL/KsJ-db/db (db/db) mice initiated with azoxymethane (AOM). Male db/db, db/+ and +/+ mice were given 5 weekly subcutaneous injections of AOM (15 mg/kg body weight), and then they were fed the diet containing 0.02%, 0.1% or 0.5% CUSM for 7 weeks. At Week 12, a significant increase in the numbers of ACF and BCAC was noted in the db/db mice in comparison with the db/+ and +/+ mice. Feeding with CUSM caused reduction in the frequency of ACF in all genotypes of mice and the potency was high in order of the db/db mice, db/+ mice and +/+ mice. The number of BCACs was also reduced by feeding with CUSM, thus resulting in a 28-61% reduction in the db/db mice, possibly due to suppression of cell proliferation activity in the lesions by feeding with CUSM-containing diet. Clinical chemistry revealed a low serum level of triglyceride in mice fed CUSM. In addition, CUSM feeding inhibited fatty metamorphosis and fibrosis in the liver of db/db mice. Our findings show that CUSM in the diet has a chemopreventive ability against the early phase of AOM-induced colon carcinogenesis in the db/db as well as db/+ and +/+ mice, indicating potential use of CUSM in cancer chemoprevention in obese people.


Subject(s)
Colonic Neoplasms/prevention & control , Diet , Dietary Supplements , Fatty Liver/prevention & control , Plant Extracts/administration & dosage , Precancerous Conditions/prevention & control , Animals , Azoxymethane/toxicity , Carcinogens/toxicity , Cholesterol/blood , Colon/chemistry , Colon/pathology , Colonic Neoplasms/chemically induced , Colonic Neoplasms/pathology , Insulin/blood , Leptin/blood , Liver/chemistry , Liver/pathology , Male , Mice , Mice, Inbred Strains , Precancerous Conditions/chemically induced , Proliferating Cell Nuclear Antigen/analysis , Triglycerides/blood
17.
Oncol Rep ; 15(3): 533-8, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16465408

ABSTRACT

Fermented brown rice by Aspergillus oryzae (FBRA) has been shown to be a potent anti-carcinogenic compound. Here, we investigated the modifying effects of dietary feeding with a naturally occurring anti-oxidant FBRA on N-butyl-N-(4-hydroxybutyl)-nitrosamine (OH-BBN)-induced urinary bladder carcinogenesis in male ICR mice. Five-week-old male ICR mice were divided into 7 groups, and groups 1-5 were given OH-BBN (500 ppm) in drinking water for 6 weeks starting at 7 weeks of age. Groups 2 and 3 were fed the diet containing 5% and 10% FBRA during the initiation phase, respectively, whereas groups 4 and 5 were fed these diets during the post-initiation phase. Group 6 was given the diet containing 10% FBRA throughout the experiment, and group 7 was kept on the basal diet alone and served as an untreated control. At the end of the study (week 32), the incidences of simple hyperplasia, dysplasia and carcinoma in the bladders of group 1 (OH-BBN alone) were 92%, 49% and 38%, respectively. Those of group 5 (64%, 23% and 10%) and the incidence of carcinoma of group 4 (17%) was significantly less than that of group 1. Furthermore, the multiplicity of simple hyperplasia and carcinoma of group 5 was significantly less than that of group 1. Post-initiation exposure of 10% FBRA significantly decreased the number/nucleus of silver-stained nucleolar organizer region proteins (AgNORs), an index of cell proliferation, in the non-lesional transitional epithelium when compared to that of the control. Our results indicate that FBRA exerts chemopreventive effects against chemically induced urinary bladder carcinogenesis through anti-proliferative mechanisms. FBRA could be a promising chemopreventive agent for human urinary bladder cancer.


Subject(s)
Diet , Oryza , Urinary Bladder Neoplasms/prevention & control , Analysis of Variance , Animals , Butylhydroxybutylnitrosamine/administration & dosage , Butylhydroxybutylnitrosamine/toxicity , Cell Proliferation/drug effects , Hyperplasia/chemically induced , Hyperplasia/prevention & control , Male , Mice , Mice, Inbred ICR , Nucleolus Organizer Region/chemistry , Precancerous Conditions/chemically induced , Precancerous Conditions/prevention & control , Silver Staining , Urinary Bladder/chemistry , Urinary Bladder/drug effects , Urinary Bladder/pathology , Urinary Bladder Neoplasms/chemically induced
18.
Cancer Lett ; 238(1): 69-75, 2006 Jul 08.
Article in English | MEDLINE | ID: mdl-16046055

ABSTRACT

Effect of degraded lambda-carrageenan, which induces colitis in rodents, on the development of beta-catenin-accumulated crypts (BCAC) being putative precancer lesions of colon cancer was investigated in male DBA/2J mice initiated with azoxymethane (AOM). In a preliminary experiment, male DBA/2J mice among seven different strains (A/J, BALB/c, C3H/HeN, C57BL/6J, CBA/N, DBA/1J, and DBA/2J) of male mice were most sensitive to degraded lambda-carrageenan. Therefore, male DBA/2J mice were intraperitonially injected AOM (10 mg/kg body weight), and then 2% degraded lambda-carrageenan in drinking water for one or two weeks, starting one week after dosing of AOM. Thereafter animals were no further treated up to week 26. At week 26, the frequency of BCAC in the colonic mucosa was 12.50+/-2.46 in the AOM alone group, 11.30+/-3.50 in the AOM/degraded lambda-carrageenan (for one week) group, and 11.60+/-2.27 in the AOM/degraded lambda-carrageenan (for two weeks) group. The findings suggest that degraded lambda-carrageenan treatment for one or two weeks did not affect the occurrence of BCAC. Our results may indicate no enhancing or promoting effects of degraded lambda-carrageenan on colon carcinogenesis in mice initiated with AOM.


Subject(s)
Azoxymethane/toxicity , Carcinogens/toxicity , Carrageenan/pharmacology , Intestinal Mucosa/drug effects , Precancerous Conditions/chemically induced , beta Catenin/biosynthesis , Animals , Biomarkers, Tumor/analysis , Biomarkers, Tumor/biosynthesis , Carrageenan/chemistry , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred DBA , Mice, Inbred Strains , Precancerous Conditions/pathology , Species Specificity , beta Catenin/analysis
19.
Int J Cancer ; 118(1): 25-34, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16049979

ABSTRACT

The mouse model for familial adenomatous polyposis, Apc(Min/+) mouse, contains a truncating mutation in the Apc gene and spontaneously develops numerous adenomas in the small intestine but few in the large bowel. Our study investigated whether dextran sodium sulfate (DSS) treatment promotes the development of colonic neoplasms in Apc(Min/+) mice. Apc(Min/+) and Apc+/+ mice of both sexes were exposed to 2% dextran sodium sulfate in drinking water for 7 days, followed by no further treatment for 4 weeks. Immunohistochemistry for cyclooxygenase-2, inducible nitric oxide synthase, beta-catenin, p53, and nitrotyrosine, and mutations of beta-catenin and K-ras and loss of wild-type allele of the Apc gene in the colonic lesions were examined. Sequential observation of female Apc(Min/+) mice that received DSS was also performed up to week 5. At week 5, numerous colonic neoplasms developed in male and female Apc(Min/+) mice but did not develop in Apc+/+ mice. Adenocarcinomas developed in Apc(Min/+) mice that received DSS showed loss of heterozygosity of Apc and no mutations in the beta-catenin and K-ras genes. The treatment also significantly increased the number of small intestinal polyps. Sequential observation revealed increase in the incidences of colonic neoplasms and dysplastic crypts in female Apc(Min/+) mice given DSS. DSS treatment increased inflammation scores, associated with high intensity staining of beta-catenin, cyclooxygenase-2, inducible nitric oxide synthase and nitrotyrosine. Interestingly, strong nuclear staining of p53 was specifically observed in colonic lesions of Apc(Min/+) mice treated with DSS. Our results suggest a strong promotion effect of DSS in the intestinal carcinogenesis of Apc(Min/+) mice. The findings also suggest that strong oxidative/nitrosative stress caused by DSS-induced inflammation may contribute to the colonic neoplasms development.


Subject(s)
Adenocarcinoma/chemically induced , Adenocarcinoma/genetics , Anticoagulants/toxicity , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/genetics , Dextran Sulfate/toxicity , Genes, APC , Inflammation/chemically induced , Adenocarcinoma/physiopathology , Animals , Cell Transformation, Neoplastic , Colorectal Neoplasms/physiopathology , Cyclooxygenase 2/metabolism , Female , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress , beta Catenin/metabolism
20.
Cancer Lett ; 239(1): 123-8, 2006 Jul 28.
Article in English | MEDLINE | ID: mdl-16168560

ABSTRACT

Although Apc(Min/+) mice are widely used for an animal model of human familial adenomatous polyposis (FAP), a majority of intestinal polyps locate in the small intestine. We recently reported that numerous beta-catenin-accumulated crypts (BCAC), which are reliable precursor lesions for colonic adenocarcinoma, develop in the large bowel of aged Apc(Min/+) mice. In this study, we determined the presence and location of BCAC in the large intestine of juvenile Apc(Min/+) mice (3 and 5 weeks of age). Surprisingly, BCAC were noted in the colon of even Apc(Min/+) mice of both ages, and mainly located in the distal and middle segments of the colon. Also, a few microadenomas were detected in Apc(Min/+) mice of 5-week old. Our results may indicate need of further investigation of the colorectal mucosa of Apc(Min/+) mice for examining colorectal carcinogenesis using Apc(Min/+) mice.


Subject(s)
Adenoma/metabolism , Adenomatous Polyposis Coli/metabolism , Colon/metabolism , Colonic Neoplasms/metabolism , Intestinal Mucosa/metabolism , beta Catenin/metabolism , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/pathology , Animals , Animals, Newborn , Female , Genes, APC/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...