Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Prostate ; 75(2): 151-60, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25284475

ABSTRACT

BACKGROUND: Ellagic acid (EA), a component of pomegranate fruit juice (PFJ), is a plant-derived polyphenol and has antioxidant properties. PFJ and EA have been reported to suppress various cancers, including prostate cancer. However, their chemopreventive effects on development and progression of prostate cancer using in vivo models have not been established yet. METHODS: The transgenic rat for adenocarcinoma of prostate (TRAP) model was used to investigate the modulating effects of PFJ and EA on prostate carcinogenesis. Three-week-old male transgenic rats were treated with EA or PFJ for 10 weeks. In vitro assays for cell growth, apoptosis, and Western blot were performed using the human prostate cancer cell lines, LNCaP (androgen-dependent), PC-3 and DU145 (androgen-independent). RESULTS: PFJ decreased the incidence of adenocarcinoma in lateral prostate, and both EA and PFJ suppressed the progression of prostate carcinogenesis and induced apoptosis by caspase 3 activation in the TRAP model. In addition, the level of lipid peroxidation in ventral prostate was significantly decreased by EA treatment. EA was able to inhibit cell proliferation of LNCaP, whereas this effect was not observed in PC-3 and DU145. As with the in vivo data, EA induced apoptosis in LNCaP by increasing Bax/Bcl-2 ratio and caspase 3 activation. Cell-cycle related proteins, p21(WAF) , p27(Kip) , cdk2, and cyclin E, were increased while cyclin D1 and cdk1 were decreased by EA treatment. CONCLUSIONS: The results indicate that PFJ and EA are potential chemopreventive agents for prostate cancer, and EA may be the active component of PFJ that exerts these anti-cancer effects.


Subject(s)
Androgen Antagonists/therapeutic use , Apoptosis/drug effects , Carcinogenesis/drug effects , Ellagic Acid/therapeutic use , Lythraceae , Prostatic Neoplasms/drug therapy , Androgen Antagonists/isolation & purification , Androgen Antagonists/pharmacology , Androgens/metabolism , Animals , Apoptosis/physiology , Beverages , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Ellagic Acid/isolation & purification , Ellagic Acid/pharmacology , Fruit , Humans , Male , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Rats , Rats, Sprague-Dawley , Rats, Transgenic
2.
Carcinogenesis ; 35(9): 1962-7, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24562575

ABSTRACT

There is a need for exploration of new therapeutic strategies that target distinct molecular mechanisms of castration-resistant prostate cancer (CRPC) because its emergence following androgen deprivation therapy is a major clinical problem. In this report, we investigated the role of glutathione peroxidase 2 (GPX2) in CRPC. GPX2 expression was analyzed in rat and human CRPC cells. Next, we determined the proliferation rate and level of reactive oxygen species (ROS) in GPX2-small interfering RNA (siRNA)-transfected CRPC cells. For in vivo analysis, siRNA-transfected cells were subcutaneously implanted into normal and castrated nude mice. Further, immunohistochemical and prognostic analyses of GPX2 were performed using human specimens. Silencing of GPX2 caused significant growth inhibition and increased intracellular ROS in both rat (PCai1) and human (PC3) CRPC cells. Flow cytometry and western blot analyses revealed that the decrease in proliferation rate of the GPX2-silenced cells was due to cyclin B1-dependent G2/M arrest. Furthermore, knockdown of Gpx2 inhibited tumor growth of PCai1 cells in castrated mice. Immunohistochemical analyses indicated that expression of GPX2 was significantly higher in residual cancer foci after neoadjuvant hormonal therapy than in hormone naive cancer foci. Moreover, patients with high GPX2 expression in biopsy specimen had significantly lower prostate-specific antigen recurrence-free survival and overall survival than those with no GPX2 expression. These findings suggest that GPX2 is a prognostic marker in CRPC and affects proliferation of prostate cancer under androgen depletion partially through protection against ROS signaling.


Subject(s)
Adenocarcinoma/enzymology , Cell Proliferation , Glutathione Peroxidase/metabolism , Prostatic Neoplasms, Castration-Resistant/enzymology , Adenocarcinoma/drug therapy , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Androgen Antagonists/pharmacology , Androgen Antagonists/therapeutic use , Animals , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Agents, Hormonal/therapeutic use , Cell Line, Tumor , Disease-Free Survival , Drug Resistance, Neoplasm , Gene Expression , Glutathione Peroxidase/genetics , Humans , Male , Mice , Mice, Nude , Multivariate Analysis , Neoplasm Transplantation , Neoplasm, Residual , Prognosis , Proportional Hazards Models , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/mortality , Prostatic Neoplasms, Castration-Resistant/pathology , RNA Interference , RNA, Small Interfering/genetics , Rats , Reactive Oxygen Species/metabolism
3.
Toxicology ; 311(3): 115-23, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23867582

ABSTRACT

Understanding of mechanisms of cancer progression is very important for reduction of cancer mortality. Of six rat hepatocellular carcinoma (HCC) cell lines, differing in their metastatic potential to the lung after inoculation into the tail vein of nude mice, the most metastatic featured particular overexpression of glutathione peroxidase 2 (GPX2). Therefore, we analyzed the influence of interference in highly metastatic L2 cells by siRNA transfection. Gpx2 siRNA significantly inhibited cell proliferation at 24 and 48h time points with induction of apoptosis but not cell cycle arrest. High expression of mutated p53 was detected in all HCC cell lines, with reduction in Gpx2 siRNA-transfected cells. Migration and invasion in vitro were also suppressed as compared to control siRNA-transfected cells and secretion of matrix metalloproteinase 9 was reduced. In vivo, the numbers and areas of metastatic nodules per area in the lungs were significantly reduced in the mice inoculated with Gpx2 siRNA-transfected cells as compared to control siRNA-transfected cells. In conclusion, expression of GPX2 is associated with cancer metastasis from rat HCCs both in vitro and in vivo. Together with immunohistochemical findings of elevated expression in rat and also human liver lesions, the results point to important roles in hepatocarcinogenesis.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Glutathione Peroxidase/metabolism , Liver Neoplasms/metabolism , Aged , Animals , Apoptosis , Carcinoma, Hepatocellular/pathology , Cell Cycle , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Gene Silencing , Glutathione Peroxidase/genetics , Humans , Liver Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Nude , Middle Aged , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Rats , Rats, Inbred F344 , Reactive Oxygen Species/metabolism
4.
Asian Pac J Cancer Prev ; 14(5): 2859-63, 2013.
Article in English | MEDLINE | ID: mdl-23803044

ABSTRACT

Polyphenolic compounds from pomegranate fruit extracts (PFEs) have been reported to possess antiproliferative, pro-apoptotic, anti-inflammatory and anti-invasion effects in prostate and other cancers. However, the mechanisms responsible for the inhibition of cancer invasion remain to be clarified. In the present study, we investigated anti-invasive effects of ellagic acid (EA) in androgen-independent human (PC-3) and rat (PLS10) prostate cancer cell lines in vitro. The results indicated that non-toxic concentrations of EA significantly inhibited the motility and invasion of cells examined in migration and invasion assays. The EA treatment slightly decreased secretion of matrix metalloproteinase (MMP)-2 but not MMP-9 from both cell lines. We further found that EA significantly reduced proteolytic activity of collagenase/gelatinase secreted from the PLS-10 cell line. Collagenase IV activity was also concentration-dependently inhibited by EA. These results demonstrated that EA has an ability to inhibit invasive potential of prostate cancer cells through action on protease activity.


Subject(s)
Cell Movement/drug effects , Collagenases/metabolism , Ellagic Acid/pharmacology , Gelatinases/metabolism , Neoplasm Invasiveness/pathology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Lythraceae , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Plant Extracts/pharmacology , Prostatic Neoplasms , Rats , Rats, Inbred F344
5.
Toxicol Pathol ; 40(5): 715-21, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22569583

ABSTRACT

Connexin 32 (Cx32) is a major gap junction protein in the liver. The authors previously demonstrated that transgenic rats carrying a dominant negative mutant of Cx32 (Cx32ΔTg) have much decreased capacity for gap junctional intercellular communication (GJIC) and increased susceptibility to diethylnitrosamine (DEN)-induced hepatocarcinogenesis as compared to littermate wild-type (wt) rats. To evaluate the age-dependent susceptibility to DEN-induced hepatocarcinogenesis and alteration of GJIC function, male Cx32ΔTg and wt rats at 10, 30, or 85 weeks old were given a single intraperitoneal administration of DEN (40 mg/rat) and sacrificed 12 weeks later. The number and area of glutathione S-transferase placental form (GST-P)-positive preneoplastic foci were significantly increased in the liver of 10- and 30-wk-old Cx32ΔTg rats compared with age-matched wt. However, in the 85-wk-old rats, both Cx32ΔTg and wt rats had similarly large number and area of GST-P-positive foci, and the difference was not significant. Interestingly, function of hepatic GJIC was reduced and protein and mRNA expression of Cx32 were decreased with aging in wt rats. These results suggest that a decline of hepatic intercellular communication through gap junction results in increased susceptibility to DEN-induced hepatocarcinogenesis in aged rats.


Subject(s)
Cell Communication/drug effects , Disease Susceptibility/metabolism , Gap Junctions/drug effects , Liver/drug effects , Animals , Carcinogens/pharmacology , Connexins/genetics , Connexins/metabolism , Diethylnitrosamine/pharmacology , Gap Junctions/metabolism , Glutathione Transferase/metabolism , Liver/metabolism , Liver/pathology , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/metabolism , Male , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Gap Junction beta-1 Protein
6.
Cancer Sci ; 103(5): 860-7, 2012 May.
Article in English | MEDLINE | ID: mdl-22320152

ABSTRACT

To reduce cancer mortality, understanding of mechanisms of cancer metastasis is crucial. We have established six rat hepatocellular carcinoma (HCC) cell lines, which exhibit differing metastatic potential to the lung after inoculation into the tail veins of nude mice. In the present experiment, we investigated the process of cell attachment to metastatic sites and possible regulating factors. One hour after inoculation, two of two HCC cell lines with high metastatic potential and one of two HCC cell lines with low metastatic potential exhibited many attached cells in the lung. One day after inoculation, lung metastatic foci were observed only with highly-metastatic cells with elevated connexin 43 (Cx43) expression as assessed by cDNA array analysis. Furthermore, 24 or 48 h after transfection of an siRNA targeting Cx43, in vitro invasion and migration were suppressed by 68% (P < 0.001) and 36% (P < 0.05) compared with control-siRNA transfected cells, despite no differences in cellular morphology, cell proliferation or apoptotic activity. Moreover, the number of metastatic nodules per lung area in nude mice was significantly (P < 0.01) reduced. In conclusion, suppression of Cx43 expression in tumor cells reduced in vitro migration and invasion capacity and in vivo metastatic ability so that Cx43 has potential as a molecular target for prevention of cancer metastasis with Cx43 overexpressing tumors.


Subject(s)
Carcinoma, Hepatocellular/genetics , Connexin 43/genetics , Liver Neoplasms/genetics , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Animals , Carcinoma, Hepatocellular/secondary , Cell Line, Tumor , Cell Movement/genetics , Gene Silencing , Liver Neoplasms/pathology , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation , Rats
7.
Food Chem Toxicol ; 50(3-4): 840-7, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22266361

ABSTRACT

In this study, we focused on the in vitro effects of Kuguacin J (KuJ), a purified component of bitter melon (Momordica charantia) leaf extract (BMLE), on the androgen-independent human prostate cancer cell line PC3 and the in vivo effect of dietary BMLE on prostate carcinogenesis using a PC3-xenograph model. KuJ exerted a strong growth-inhibitory effect on PC3 cells. Growth inhibition was mainly through G1-arrest: KuJ markedly decreased the levels of cyclins (D1 and E), cyclin-dependent kinases (Cdk2 and Cdk4) and proliferating cell nuclear antigen. Interestingly, KuJ also dramatically decreased the levels of survivin expressed by PC3 cells. In addition, KuJ exerted anti-invasive effects on PC3 cells, significantly inhibiting migration and invasion: KuJ inhibited secretion of the active forms of MMP-2, MMP-9 and uPA by PC3 cells. In addition, KuJ treatment significantly decreased the expression of membrane type 1-MMP (MT1-MMP) by PC3 cells. In vivo, 1% and 5% BMLE in the diet resulted in 63% and 57% inhibition of PC3 xenograft growth without adverse effect on host body weight. Our results suggest that KuJ is a promising new candidate chemopreventive and chemotherapeutic agent for prostate cancer.


Subject(s)
Androgens/pharmacology , Momordica/chemistry , Plant Leaves/chemistry , Prostatic Neoplasms/pathology , Triterpenes/analysis , Animals , Apoptosis/drug effects , Base Sequence , Blotting, Western , Cell Cycle/drug effects , Cell Line, Tumor , DNA Primers , Disease Progression , Humans , In Situ Nick-End Labeling , Male , Matrix Metalloproteinases/genetics , Mice , Mice, Nude , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Xenograft Model Antitumor Assays
8.
Pathol Int ; 62(2): 84-92, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22243777

ABSTRACT

Histone H1, one of the histone superfamilies, is known to determine chromatin structure and alter gene expression. It also contributes to regulation of cell proliferation in breast cancer. We hypothesized a similar association in prostate cancer, and therefore examined relationships between histone H1 expression and Gleason pattern, Ki-67 and androgen receptor levels in a series of prostate cancer tissues and cell lines. Histone H1 positive cancer cells increased with the Gleason pattern. Gleason pattern 3 tumors were divided into two groups, one with high histone H1 positivity (H1-high cases, 60-100% positivity) and the other with low histone H1 positivity (H1-low cases, 0-20% positivity). Ki-67 or androgen receptor positivity in H1-high cases was significantly higher than in H1-low cases. PC3 cells demonstrated more frequent histone H1 and Ki-67 positivity as compared to LNCaP cells. Silencing of histone H1 by siRNA transfection significantly reduced cell proliferation in LNCaP and PC3. These findings suggest that histone H1 expression is associated with the Gleason pattern, cell proliferation and androgen receptor expression in prostate cancers.


Subject(s)
Histones/metabolism , Prostate/metabolism , Prostatic Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Histones/genetics , Humans , Male , Neoplasm Grading , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Receptors, Androgen/genetics , Receptors, Androgen/metabolism
9.
Prostate ; 72(5): 533-41, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21748757

ABSTRACT

BACKGROUND: Elucidating the mechanisms of metastasis in prostate cancer, particularly to the bone, is a major issue for treatment of this malignancy. We previously reported that an androgen-independent variant had higher expression of glutathione S-transferase pi (Gst-pi) compared with a parent androgen-dependent transplantable rat prostate carcinoma which was established from the transgenic rat for adenocarcinoma of the prostate (TRAP). METHODS: A new cell line, PCai1, was established from the androgen-independent tumor and investigated its metastatic potential in nude mice. The tumorigenesis of PCai1 cells in vivo was studied by subcutaneous transplantations into nude mice. The growth in the microenvironment of the prostate was studied by orthotopic transplantation of PCai1 cells into nude mice. The metastatic potential of PCai1 cells was studied by tail vein injections. Effects of Gst-pi knocked down were analysis in PCai1 cells. RESULTS: PCai1 frequently formed metastatic lesions in the lung and lymph nodes after orthotopic implantation in the prostate. Intravenous injections of PCai1, metastasis to lung and bone were obvious. PCai1 had strong expression for Gst-pi, therefore we tried knocked down Gst-pi. Gst-pi-siRNA in vitro significantly suppressed cell proliferation rate. In addition, high levels of intracellular reactive oxygen species (ROS) were recognized in the Gst-pi knockout. CONCLUSIONS: Gst-pi expression of the prostate cancers are dependent on metastatic site, and that Gst-pi has an important role in adapting prostate cancer for growth and metastasis involving an alteration of ROS signals.


Subject(s)
Adenocarcinoma/secondary , Glutathione S-Transferase pi/genetics , Neoplasms, Hormone-Dependent/pathology , Prostatic Neoplasms/pathology , Adenocarcinoma/enzymology , Adenocarcinoma/genetics , Androgens/physiology , Animals , Bone Neoplasms/enzymology , Bone Neoplasms/genetics , Bone Neoplasms/secondary , Cell Line, Tumor , Gene Expression , Gene Knockdown Techniques , Gene Silencing , Glutathione S-Transferase pi/metabolism , Male , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms, Hormone-Dependent/enzymology , Neoplasms, Hormone-Dependent/genetics , Organ Specificity , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , RNA, Small Interfering/genetics
10.
Toxicol Pathol ; 39(7): 1084-90, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21934139

ABSTRACT

In a previous study, to identify genes of importance for hepatocellular carcinogenesis, and especially for processes involved in malignant transformation, the authors investigated differences in gene expression between adenomas and carcinomas by DNA microarray. In the present study, the authors investigated AW434047, one of the sequences that was upregulated in carcinomas. The investigation led to the identification of a novel gene, which the authors named hepatocyte malignant transforming factor (HMTF), of unknown function whose expression was increased in hepatocellular carcinomas. Northern blot and in situ hybridization also demonstrated high levels of HMTF in rat hepatocellular carcinoma (HCC) cell lines, lymphocytes in the spleen, colon mucosal epithelia, spermatocytes, and granule cells of the hippocampus. Reduction of HMTF by RNA interference (RNAi) in N1 cells, an HCC cell line, caused suppression of cell proliferation, invasion, and migration. Suppression of proliferation appeared to be due to cell cycle arrest without increased apoptosis. Decreased HMTF expression resulted in down-regulation of STAT3, PCNA, and cyclin D1 and upregulation of p27. These results suggest that HMTF is a new marker for rat HCC and is involved in HCC cell proliferation and may also be linked to cell proliferation in the spleen, colon, brain, and testis.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Transformation, Neoplastic/metabolism , Liver Neoplasms, Experimental/metabolism , Neoplasm Proteins/metabolism , Animals , Apoptosis , Biomarkers, Tumor/chemistry , Biomarkers, Tumor/genetics , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Liver Neoplasms, Experimental/chemistry , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/pathology , Male , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Organ Specificity , RNA Interference , RNA, Small Interfering/pharmacology , Rats , Rats, Inbred F344 , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics
11.
Cancer Lett ; 306(2): 142-50, 2011 Jul 28.
Article in English | MEDLINE | ID: mdl-21429659

ABSTRACT

In this study, we focused on the effects of a bitter melon (Momordica charantia) leaf extract (BMLE) and a purified component, Kuguacin J (KuJ), on androgen-dependent LNCaP human prostate cancer cells. Both treatments exerted growth inhibition through G1 arrest and induction of apoptosis. In addition, KuJ markedly decreased the levels of cyclins (D1 and E), cyclin-dependent kinases (Cdk2 and Cdk4) and proliferating cell nuclear antigen, and caused an increase in p21 and p27 levels. Its induction of apoptosis was accompanied by an increase in cleavage of caspase-3 and poly (ADP-ribose) polymerase, attributable to augment of Bax/Bcl-2 and Bad/Bcl-xL and reduction of survivin levels. BMLE and KuJ also reduced the expression of androgen receptor (AR), prostate-specific antigen (PSA) while induced P53 protein level. Down-regulation of p53 by RNA interference indicated that BMLE and KuJ inhibited cell growth partly through p53-dependent cell cycle arrest and apoptotic pathways. Both BMLE and KuJ caused less toxicity in a normal prostate cell line, PNT1A. Our results suggest that BMLE and a purified component, KuJ, from its diethyl ether fraction could be promising candidate new antineoplastic and chemopreventive agents for androgen-dependent prostate cancer and carcinogenesis.


Subject(s)
Apoptosis/drug effects , G1 Phase/drug effects , Momordica charantia/chemistry , Neoplasms, Hormone-Dependent/pathology , Plant Leaves/chemistry , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Triterpenes/pharmacology , Blotting, Western , Cell Cycle Proteins , Cell Proliferation/drug effects , Humans , Male , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/metabolism , Prostatic Neoplasms/metabolism , Tumor Cells, Cultured
12.
Pathol Int ; 61(4): 202-9, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21418392

ABSTRACT

Epidemiological data on the relationship between hypertension and prostate cancer development are conflicting. To cast light on this question, we performed animal experiments using hybrid rats generated by crossing the spontaneously hypertensive rat (SHR) or its normotensive control Wistar Kyoto (WKY) rat with a transgenic rat for adenocarcinoma of prostate (TRAP) that features development of adenocarcinoma at high incidence by 15 weeks of age. The number of adenocarcinomatous foci in the lateral prostate of hypertensive (TRAP × SHR)F1 rats was demonstrated to be significantly increased compared with those of normotensive (TRAP × WKY)F1 rats. In the ventral prostate, increase of carcinoma foci was also observed but did not reach significance. The number of cancer foci showing microinvasion in (TRAP × SHR)F1 rats was higher than that of (TRAP × WKY)F1 rats, but again without significance, while treatment with prazosin, an anti-hypertensive agent, tended to decrease microinvasive carcinoma foci in both the ventral and lateral prostate. In conclusion, the present study provided additional evidence that high blood pressure is associated with prostate cancer risk.


Subject(s)
Adenocarcinoma/complications , Hypertension/complications , Prostatic Neoplasms/complications , Animals , Disease Models, Animal , Female , Immunohistochemistry , Male , Rats , Rats, Inbred SHR , Rats, Transgenic , Rats, Wistar
13.
Nutr Cancer ; 63(2): 227-33, 2011.
Article in English | MEDLINE | ID: mdl-21298591

ABSTRACT

The current study was designed to investigate the effects of nobiletin (5,6,7,8,3',4'-hexamethoxy flavone) on 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP)-induced prostate and colon carcinogenesis. PhIP was administered to 6-wk-old F344 male rats intragastrically (100 mg/kg) twice a wk for 10 wk. The animals were given 0.05% nobiletin or the basal diet for 50 wk. At the end of the experiment, serum testosterone, estrogen, and leptin did not differ between the 2 groups. The body weights of nobiletin-treated rats were significantly higher than controls (P<0.05), and feeding of nobiletin significantly reduced the relative prostate (P<0.05) and testes (P<0.05) weights as well as the Ki67 labeling index in the normal epithelium in the ventral prostate (P<0.01). The incidence and multiplicity of adenocarcinomas in nobiletin-treated ventral prostate were 50% and 36%, respectively, of controls, but the differences were not statistically significant. However, nobiletin did significantly reduce the total number of colonic aberrant crypt foci (ACF) compared to the control value (P<0.05). Nobiletin, therefore, may have potential for chemoprevention of early changes associated with carcinogenesis in both the prostate and colon.


Subject(s)
Colonic Neoplasms/drug therapy , Flavones/pharmacology , Imidazoles/toxicity , Prostatic Neoplasms/drug therapy , Aberrant Crypt Foci , Adenocarcinoma/chemically induced , Animals , Carcinogens/toxicity , Chemoprevention , Colonic Neoplasms/chemically induced , Male , Organ Size , Prostate/pathology , Prostatic Neoplasms/chemically induced , Rats , Rats, Inbred F344 , Testis/pathology
14.
Cancer Sci ; 101(10): 2234-40, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20731662

ABSTRACT

Cancer metastasis is a major cause of death in cancer patients, with invasion as a first step greatly contributing to the failure of clinical treatments. Any compounds with an inhibitory influence on this process are therefore of prime interest. Momordica charantia (bitter melon) is widely consumed as a vegetable and especially as a folk medicine in Asia. Here, we investigated the anti-invasive effects of bitter melon leaf extract (BMLE) on a rat prostate cancer cell line (PLS10) in vitro and in vivo. The results indicated that non-toxic concentrations of BMLE significantly inhibited the migration and invasion of cells in vitro. The results of zymography showed that BMLE inhibited the secretion of MMP-2, MMP-9 and urokinase plasminogen activator (uPA) from PLS10. Real-time RT-PCR revealed that BMLE not only significantly decreased gene expression of MMP-2 and MMP-9, but also markedly increased the mRNA level of TIMP-2, known to have inhibitory effects on the activity of MMP-2. An EnzChek gelatinase/collagenase assay showed that collagenase type IV activity was partially inhibited by BMLE. In the in vivo study, intravenous inoculation of PLS10 to nude mice resulted in a 100% survival rate in the mice given a BMLE-diet as compared with 80% in the controls. The incidence of lung metastasis did not show any difference, but the percentage lung area occupied by metastatic lesions was slightly decreased in the 0.1% BMLE treatment group and significantly decreased with 1% BMLE treatment as compared with the control. Thus, the results indicate for the first time an anti-metastatic effect of BMLE both in vitro and in vivo.


Subject(s)
Momordica charantia , Plant Extracts/therapeutic use , Prostatic Neoplasms/drug therapy , Animals , Cell Line, Tumor , Disease Progression , Lung Neoplasms/secondary , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Phytotherapy , Plant Extracts/pharmacology , Plant Leaves , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Rats , Transforming Growth Factor beta/physiology
15.
Toxicol Pathol ; 38(4): 583-91, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20448087

ABSTRACT

Interaction of more than two chemicals from foods is a very important factor for carcinogenic risk assessment and management. 2-Amino-3,8-dimethylimidazo[4,5-f]quinoxaline (MeIQx), one of the most abundant carcinogenic heterocyclic amines in cooked foods, is speculated to be a human liver carcinogen. MeIQx is metabolically activated by CYP1A2 and then N-acetyltransferase (NAT), findings that suggest that its carcinogenic potential might be enhanced by simultaneous exposure to chemical(s) inducing CYP1A2. Therefore, we here investigated the effects of alpha- and beta-naphthoflavone as CYP1A2 inducers on MeIQx-induced rat hepatocarcinogenesis in a medium-term rat liver bioassay. Unexpectedly, no modifying influence of naphthoflavones on MeIQx-induced hepatocarcinogenesis was demonstrated with reference to glutathione S-transferase placental form (GST-P) positive foci in the liver, although up-regulation of CYP1A2 was detected on Western blot analysis. Activity of NAT was not affected. In MeIQx-treated rats, CYP1A expression was mainly detected in zone 3 of the liver where GST-P positive foci were preferentially located, while naphthoflavones alone or combinations of naphthoflavones and MeIQx induced CYP1A expression in zone 1. This difference in intralobular distribution of CYP1A might be related to the fact that MeIQx hepatocarcinogenesis was not modified by the two CYP1A inducers.


Subject(s)
Benzoflavones/toxicity , Cytochromes/biosynthesis , Enzyme Inhibitors/toxicity , Liver Neoplasms, Experimental/chemically induced , Quinoxalines/toxicity , beta-Naphthoflavone/toxicity , Analysis of Variance , Animals , Arylamine N-Acetyltransferase/genetics , Arylamine N-Acetyltransferase/metabolism , Body Weight/drug effects , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1A2 , Drug Synergism , Immunohistochemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Liver/metabolism , Liver/pathology , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Male , Organ Size/drug effects , Organ Specificity , Rats , Rats, Inbred F344
16.
Toxicol Pathol ; 38(2): 280-6, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20097795

ABSTRACT

Acetaminophen (APAP) is a widely used antipyretic and analgesic agent. However, overdosing and sometimes even a recommended dose can lead to serious and conceivably fatal liver toxicity. Therefore, it is important to clarify understand mechanisms of hepatotoxicity induced by APAP. Gap junctions, formed by connexin, have important roles in maintenance of tissue homeostasis and control of cell growth and differentiation. In the liver, Cx32 is a major gap junction protein whose expression is known to gradually decrease with chronic liver disease progression. In the present study, acute hepatotoxic effects of APAP were found to be reduced in Cx32 dominant negative transgenic rats lacking normal gap junctional intercellular communication in the liver. In littermate wild-type rats, the injured centrilobular hepatocytes were positive for TUNEL staining and featured elevated expre ssion of cleaved caspase-3 and Cx43, which is not expressed in normal hepatocytes. These results suggest that APAP hepatotoxicity involves apoptosis, and induction of Cx43 expression may play an important role in the apoptotic signaling. Moreover, gap junctional functions of Cx32 can play important roles in removing damaged hepatocytes by apoptosis for liver tissue homeostasis.


Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Chemical and Drug Induced Liver Injury/metabolism , Connexin 43/metabolism , Gap Junctions/drug effects , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Chemical and Drug Induced Liver Injury/pathology , Connexin 43/drug effects , Connexins/metabolism , Female , Male , Rats , Rats, Sprague-Dawley , Gap Junction beta-1 Protein
17.
Prostate ; 70(3): 229-38, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-19790239

ABSTRACT

BACKGROUND: In bone metastatic sites, prostate cancer cells proliferate on interacting with osteoclasts. Tranilast, which is used for an antiallergic drug, has been shown to inhibit growth of several cancers and stromal cells. The present study was conducted to assess suppressive effects of Tranilast on prostate cancer growth and osteoclast differentiation in vivo and in vitro. METHODS: In vivo, rat prostate cancer tissue was transplanted onto cranial bones of F344 rats and Tranilast was given for 9 days at doses of 0, 200, or 400 mg/kg/day. In vitro, human prostate cancer cell lines, LNCaP, PC3, and DU145, the rat prostate cancer cell line, PLS-10, and rat bone marrow cells were similarly treated with the agent. RESULTS: In vivo, tumor volumes were significantly decreased in the high dose group. While cell proliferation did not appear to be affected, apoptosis was induced and tumor necrosis was apparent. Cranial bone defects were decreased in the high dose group. In vitro, cell proliferation rates of all four cell lines were reduced by Tranilast and increased apoptosis was observed in LNCaP and PLS-10. In addition, Tranilast significantly reduced osteoclast differentiation of rat bone marrow cells. Western blot analysis of PLS-10 and LNCaP revealed that phospho-GSK3beta was up-regulated and phospho-Akt was down-regulated. CONCLUSIONS: Tranilast here suppressed rat prostate cancer growth and osteoclast differentiation. Growth of human prostate cancer cells was also inhibited. Thus, this agent deserves consideration as a candidate for conventional therapy of bone metastatic prostate cancer.


Subject(s)
Bone Marrow Cells/cytology , Cell Differentiation/drug effects , Osteoclasts/cytology , Prostatic Neoplasms/pathology , Prostatic Neoplasms/physiopathology , ortho-Aminobenzoates/administration & dosage , Animals , Apoptosis/drug effects , Blotting, Western , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Down-Regulation , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Male , Necrosis , Neoplasm Invasiveness/pathology , Neoplasm Transplantation , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Inbred F344 , Skull , Up-Regulation
18.
J Toxicol Pathol ; 23(3): 125-31, 2010 Sep.
Article in English | MEDLINE | ID: mdl-22272023

ABSTRACT

The effects of streptozotocin (STZ)-induced diabetes on induction of hepatic preneoplastic lesions by diethylnitrosamine (DEN) were investigated in male Fischer rats. A single dose of STZ was injected intravenously either 2 weeks before or after initiation with DEN. The blood glucose levels were significantly elevated from 1 week after STZ-injection until autopsy. The numbers of GST-P positive foci at 1 week after DEN administration in the STZ-injected rats were similar to those in the non-diabetic rats. In contrast, both the numbers and areas of GST-P positive foci > 2 mm in diameter 8 weeks after DEN administration were increased significantly in the rats treated with STZ after DEN exposure compared with the non-diabetic control rats. The results suggest that hepatic preneoplastic lesions initiated with DEN are promoted by STZ treatment-inducing diabetes.

19.
Pathol Int ; 59(11): 790-6, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19883429

ABSTRACT

The soybean-derived serine protease inhibitor, Bowman-Birk inhibitor (BBI), has been reported as a potent chemoprevention agent against several types of tumors. The present study was undertaken to evaluate the effects of BBI on androgen-sensitive/dependent prostate cancers using a human prostate cancer cell (LNCaP) and the transgenic rats developing adenocarcinoma of the prostate (TRAP) model. Treatment of LNCaP prostate cancer cells with 500 microg/mL BBI resulted in inhibition of viability measured on WST-1 assays, with induction of connexin 43 (Cx43) and cleaved caspase-3 protein expression. Feeding of 3% roughly prepared BBI (BBIC) to TRAP from the age 3 weeks to 13 weeks resulted in significant reduction of the relative epithelial areas within the acinus and multiplicity of the adenocarcinomas in the lateral prostate lobes. Cx43- and terminal deoxynucleotidyl transferase mediated dUTP-biotin end labeling of fragmented DNA (TUNEL)-positive apoptotic cancer cells were more frequently observed in the lateral prostates treated with BBIC than in the controls. These in vivo and in vitro results suggest that BBI possesses chemopreventive activity associated with induction of Cx43 expression and apoptosis.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Prostatic Neoplasms/drug therapy , Trypsin Inhibitor, Bowman-Birk Soybean/pharmacology , Animals , Animals, Genetically Modified , Blotting, Western , Cell Line, Tumor , Connexin 43/biosynthesis , Connexin 43/drug effects , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Male , Rats
20.
Toxicology ; 255(3): 160-70, 2009 Jan 31.
Article in English | MEDLINE | ID: mdl-19041683

ABSTRACT

To identify genes important in hepatocellular carcinogenesis, especially processes involved in malignant transformation, we focused on differences in gene expression between adenomas and carcinomas by DNA microarray. Eighty-one genes for which expression was specific in carcinomas were analyzed using Ingenuity Pathway Analysis software and Gene Ontology, and found to be associated with TP53 and regulators of cell proliferation. In the genes associated with TP53, we selected high mobility group box (HMGB) for detailed analysis. Immunohistochemistry revealed expression of HMGBs in carcinomas to be significantly higher than in other lesions among both human and rat liver, and a positive correlation between HMGBs and TP53 was detected in rat carcinomas. Knock-down of HMGB 2 expression in a rat hepatocellular carcinoma cell line by RNAi resulted in inhibition of cell growth, although no effects on invasion were evident in vitro. These results suggest that acquisition of malignant potential in the liver requires specific signaling pathways related to high cell proliferation associated with TP53. In particular, HMGBs appear to have an important role for progression and cell proliferation associated with loss of TP53 function in rat and in human hepatocarcinogenesis.


Subject(s)
Carcinoma, Hepatocellular/physiopathology , Cell Proliferation , High Mobility Group Proteins/physiology , Liver Neoplasms/physiopathology , Animals , Carcinoma, Hepatocellular/pathology , Cell Transformation, Neoplastic , Gene Expression Profiling , Gene Knockdown Techniques , High Mobility Group Proteins/genetics , Humans , Immunohistochemistry , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Oligonucleotide Array Sequence Analysis , Rats , Rats, Inbred F344 , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...