Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Arch Toxicol ; 98(5): 1485-1498, 2024 May.
Article in English | MEDLINE | ID: mdl-38483585

ABSTRACT

Accumulating evidence indicates that chronic circadian rhythm disruption is associated with the development of neurodegenerative diseases induced by exposure to neurotoxic chemicals. Herein, we examined the relationship between cellular circadian rhythm disruption and cytotoxicity in neural cells. Moreover, we evaluated the potential application of an in vitro cellular circadian rhythm assay in determining circadian rhythm disruption as a sensitive and early marker of neurotoxicant-induced adverse effects. To explore these objectives, we established an in vitro cellular circadian rhythm assay using human glioblastoma (U87 MG) cells stably transfected with a circadian reporter vector (PER2-dLuc) and determined the lowest-observed-adverse-effect levels (LOAELs) of several common neurotoxicants. Additionally, we determined the LOAEL of each compound on multiple cytotoxicity endpoints (nuclear size [NC], mitochondrial membrane potential [MMP], calcium ions, or lipid peroxidation) using a multiparametric high-content screening (HCS) assay using transfected U87 MG cells treated with the same neurotoxicants for 24 and 72 h. Based on our findings, the LOAEL for cellular circadian rhythm disruption for most chemicals was slightly higher than that for most cytotoxicity indicators detected using HCS, and the LOAEL for MMP in the first 24 h was the closest to that for cellular circadian rhythm disruption. Dietary antioxidants (methylselenocysteine and N-acetyl-l-cysteine) prevented or restored neurotoxicant-induced cellular circadian rhythm disruption. Our results suggest that cellular circadian rhythm disruption is as sensitive as cytotoxicity indicators and occurs early as much as cytotoxic events during disease development. Moreover, the in vitro cellular circadian rhythm assay warrants further evaluation as an early screening tool for neurotoxicants.


Subject(s)
Circadian Rhythm , Neurons , Humans
2.
Front Nutr ; 6: 132, 2019.
Article in English | MEDLINE | ID: mdl-31555652

ABSTRACT

Nutrient metabolism is under circadian regulation. Disruption of circadian rhythms by lifestyle and behavioral choices such as work schedules, eating patterns, and social jetlag, seriously impacts metabolic homeostasis. Metabolic dysfunction due to chronic misalignment of an organism's endogenous rhythms is detrimental to health, increasing the risk of obesity, metabolic and cardiovascular disease, diabetes, and cancer. In this paper, we review literature on recent findings on the mechanisms that communicate metabolic signals to circadian clocks and vice versa, and how human behavioral changes imposed by societal and occupational demands affect the physiological networks integrating peripheral clocks and metabolism. Finally, we discuss factors possibly contributing to inter-individual variability in response to circadian changes in the context of metabolic (dys)function.

3.
Sleep Med ; 16(11): 1388-1394, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26498241

ABSTRACT

BACKGROUND: Epidemiological studies indicate that disruption of circadian rhythm by shift work increases the risk of breast and prostate cancer. Our studies demonstrated that carcinogens disrupt the circadian expression of circadian genes (CGs) and circadian-controlled genes (CCGs) during the early stages of rat mammary carcinogenesis. A chemopreventive regimen of methylselenocysteine (MSC) restored the circadian expression of CGs and CCGs, including PERIOD 2 (PER2) and estrogen receptor ß (ERS2), to normal. The present study evaluated whether changes in CG and CCG expression in whole blood can serve as indicators of circadian disruption in shift workers. METHODS: Fifteen shift workers were recruited to a crossover study. Blood samples were drawn before (6 PM) and after (8 AM) completing a night shift after at least seven days on floating night-shift rotation, and before (8 AM), during (1 PM), and after (6 PM) completing seven days on day shift. The plasma melatonin level and messenger RNA (mRNA) expression of PER2, nuclear receptor subfamily 1, group d, member 1 (NR1D1), and ERS2 were measured, and the changes in levels of melatonin and gene expression were evaluated with statistical analyses. RESULTS: The mRNA expression of PER2 was affected by shift (p = 0.0079); the levels were higher in the evening for the night shift, but higher in the morning for the day shift. Increased PER2 expression (p = 0.034) was observed in the evening on the night versus day shifts. The melatonin level was higher in the morning for both day shifts (p = 0.013) and night shifts (p <0.0001). CONCLUSION: Changes in the level of PER2 gene expression can serve as a biomarker of disrupted circadian rhythm in blood cells. Therefore, they can be a useful intermediate indicator of efficacy in future MSC-mediated chemoprevention studies.


Subject(s)
Circadian Rhythm/physiology , Gene Expression , Internship and Residency , Melatonin/blood , Period Circadian Proteins/genetics , Sleep Disorders, Circadian Rhythm/genetics , Work Schedule Tolerance , Adult , Animals , Biomarkers/blood , Circadian Clocks , Cross-Over Studies , Female , Humans , Male , Period Circadian Proteins/metabolism , RNA, Messenger/metabolism , Rats , Young Adult
4.
PLoS One ; 8(9): e70930, 2013.
Article in English | MEDLINE | ID: mdl-24023717

ABSTRACT

Rat strains differ dramatically in their susceptibility to mammary carcinogenesis. On the assumption that susceptibility genes are conserved across mammalian species and hence inform human carcinogenesis, numerous investigators have used genetic linkage studies in rats to identify genes responsible for differential susceptibility to carcinogenesis. Using a genetic backcross between the resistant Copenhagen (Cop) and susceptible Fischer 344 (F344) strains, we mapped a novel mammary carcinoma susceptibility (Mcs30) locus to the centromeric region on chromosome 12 (LOD score of ∼8.6 at the D12Rat59 marker). The Mcs30 locus comprises approximately 12 Mbp on the long arm of rat RNO12 whose synteny is conserved on human chromosome 13q12 to 13q13. After analyzing numerous genes comprising this locus, we identified Fry, the rat ortholog of the furry gene of Drosophila melanogaster, as a candidate Mcs gene. We cloned and determined the complete nucleotide sequence of the 13 kbp Fry mRNA. Sequence analysis indicated that the Fry gene was highly conserved across evolution, with 90% similarity of the predicted amino acid sequence among eutherian mammals. Comparison of the Fry sequence in the Cop and F344 strains identified two non-synonymous single nucleotide polymorphisms (SNPs), one of which creates a putative, de novo phosphorylation site. Further analysis showed that the expression of the Fry gene is reduced in a majority of rat mammary tumors. Our results also suggested that FRY activity was reduced in human breast carcinoma cell lines as a result of reduced levels or mutation. This study is the first to identify the Fry gene as a candidate Mcs gene. Our data suggest that the SNPs within the Fry gene contribute to the genetic susceptibility of the F344 rat strain to mammary carcinogenesis. These results provide the foundation for analyzing the role of the human FRY gene in cancer susceptibility and progression.


Subject(s)
Genetic Predisposition to Disease/genetics , Mammary Neoplasms, Animal/genetics , Proteins/metabolism , Quantitative Trait Loci/genetics , Animals , Blotting, Northern , Cell Line, Tumor , Cells, Cultured , Female , Genotype , Humans , In Situ Hybridization, Fluorescence , Male , Phenotype , Polymerase Chain Reaction , Proteins/genetics , Rats , Rats, Inbred F344
6.
Cancer Prev Res (Phila) ; 3(5): 640-52, 2010 May.
Article in English | MEDLINE | ID: mdl-20424134

ABSTRACT

Epidemiologic and animal studies indicate that disruption of circadian rhythm increases breast cancer risk. Previously, we showed that methylselenocysteine reduced the incidence of N-nitroso-N-methylurea (NMU)-induced mammary carcinomas in Fischer 344 rats by 63%. Methylselenocysteine also increased the expression of Period 2 (Per2) and D-binding protein (DBP), providing evidence for a link between circadian rhythm and chemoprevention. Here, we report that NMU disrupted the expression of core circadian genes (Per1, Per2, Cry1, Cry2, and RevErbAalpha) and circadian-controlled genes, including melatonin receptor 1alpha (MTNR1A), estrogen receptors (ERalpha and ERbeta), and growth-regulatory genes (Trp53, p21, Gadd45alpha, and c-Myc) in mammary glands of Fischer 344 rats. By contrast, dietary methylselenocysteine (3 ppm selenium) given for 30 days, significantly enhanced the circadian expression of these genes (except for Cry1 and Cry2). The largest effect was on the levels of the Per2, MTNR1A, and ERbeta mRNAs, which showed 16.5-fold, 4.7-fold, and 9.5-fold increases in their rhythm-adjusted means, respectively, and 44.5-fold, 6.5-fold, and 9.7-fold increases in amplitude as compared with the control diet, respectively. Methylselenocysteine also shifted the peak expression times of these genes to Zeitgeber time 12 (ZT12; lights off). Methylselenocysteine also induced rhythmic expression of Trp53, p21, and Gadd45alpha mRNAs with peak levels at ZT12, when c-Myc expression was at its lowest level. However, methylselenocysteine had no significant effect on the circadian expression of these genes in liver. These results suggest that dietary methylselenocysteine counteracted the disruptive effect of NMU on circadian expression of genes essential to normal mammary cell growth and differentiation.


Subject(s)
Alkylating Agents/toxicity , Anticarcinogenic Agents/administration & dosage , Circadian Rhythm/drug effects , Cysteine/analogs & derivatives , Mammary Glands, Animal/drug effects , Methylnitrosourea/toxicity , Organoselenium Compounds/administration & dosage , Administration, Oral , Animals , Blotting, Western , Cell Proliferation , Circadian Rhythm/genetics , Cysteine/administration & dosage , Diet , Female , Gene Expression/drug effects , Gene Expression Regulation , Immunohistochemistry , Mammary Glands, Animal/metabolism , Rats , Rats, Inbred F344 , Reverse Transcriptase Polymerase Chain Reaction , Selenocysteine/analogs & derivatives
7.
Clin Cancer Res ; 11(19 Pt 1): 7033-41, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16203797

ABSTRACT

PURPOSE: We have previously shown the reactivation of some methylation-silenced genes in cancer cells by (-)-epigallocatechin-3-gallate, the major polyphenol from green tea. To determine whether other polyphenolic compounds have similar activities, we studied the effects of soy isoflavones on DNA methylation. EXPERIMENTAL DESIGN: Enzyme assay was used to determine the inhibitory effect of genistein on DNA methyltransferase activity in nuclear extracts and purified recombinant enzyme. Methylation-specific PCR and quantitative real-time PCR were employed to examine the DNA methylation and gene expression status of retinoic acid receptor beta (RARbeta), p16INK4a, and O6-methylguanine methyltransferase (MGMT) in KYSE 510 esophageal squamous cell carcinoma cells treated with genistein alone or in combination with trichostatin, sulforaphane, or 2'-deoxy-5-aza-cytidine (5-aza-dCyd). RESULTS: Genistein (2-20 micromol/L) reversed DNA hypermethylation and reactivated RARbeta, p16INK4a, and MGMT in KYSE 510 cells. Genistein also inhibited cell growth at these concentrations. Reversal of DNA hypermethylation and reactivation of RARbeta by genistein were also observed in KYSE 150 cells and prostate cancer LNCaP and PC3 cells. Genistein (20-50 micromol/L) dose-dependently inhibited DNA methyltransferase activity, showing substrate- and methyl donor-dependent inhibition. Biochanin A and daidzein were less effective in inhibiting DNA methyltransferase activity, in reactivating RARbeta, and in inhibiting cancer cell growth. In combination with trichostatin, sulforaphane, or 5-aza-dCyd, genistein enhanced reactivation of these genes and inhibition of cell growth. CONCLUSIONS: These results indicate that genistein and related soy isoflavones reactivate methylation-silenced genes, partially through a direct inhibition of DNA methyltransferase, which may contribute to the chemopreventive activity of dietary isoflavones.


Subject(s)
Anticarcinogenic Agents/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA Methylation , Esophageal Neoplasms/drug therapy , Genistein/pharmacology , Glycine max/metabolism , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Receptors, Retinoic Acid/metabolism , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cell Proliferation , Decitabine , Dose-Response Relationship, Drug , Esophageal Neoplasms/metabolism , Humans , Hydroxamic Acids/pharmacology , Isoflavones/chemistry , Isoflavones/pharmacology , Isothiocyanates , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sulfites/pharmacology , Sulfoxides , Tea , Thiocyanates/pharmacology
8.
Int J Oncol ; 26(3): 615-22, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15703815

ABSTRACT

The purpose of this study was to characterize the role of DNA hypermethylation in the loss of expression of O(6)-methylguanine-DNA methyltransferase (MGMT) during the development of esophageal squamous cell carcinoma (ESCC) and to investigate its reactivation in cell lines. Samples were collected from Linzhou City of the Henan province in northern China, a high-risk area of this disease. The hypermethylation of promoter CpG islands of the MGMT gene was examined by methylation-specific PCR in ESCC and neighboring non-tumorous tissues, as well as in laser capture microdissected samples with normal epithelium, basal cell hyperplasia (BCH), and dysplasia (DYS). The MGMT mRNA and protein expression were determined with RT-PCR and immunohistochemistry, respectively. Five of 17 (29%) normal, 10 of 20 (50%) BCH, 8 of 12 (67%) DYS, and 13 of 18 (72%) ESCC samples had DNA hypermethylation in the MGMT promoter region, showing a gradual increase with the progression of carcinogenesis. The frequency of the loss of MGMT mRNA and protein expression progressively decreased from normal to BCH, DYS, and ESCC, and it was highly correlated with MGMT promoter hypermethylation according to Fisher's exact tests. When each individual sample was considered, good concordance between DNA hypermethylation and the loss of expression of MGMT was also observed. In samples from the same patient, if hypermethylation was detected in earlier lesions, it was usually observed in more severe lesions. In the ESCC cell line KYSE 510, treatment with a DNA methyltransferase inhibitor, 2'-deoxy-5-azacytidine partially reversed the CpG hypermethylation status and restored mRNA expression of the MGMT gene. Similar reactivation of MGMT gene by dietary polyphenols, (-)-epigallocatechin-3-gallate and genistein, has also been observed. The results suggest that the DNA hypermethylation of MGMT is an important mechanism for MGMT gene silencing in the development of ESCC, and this epigenetic event may be prevented or reversed by these polyphenols for the prevention of carcinogenesis.


Subject(s)
Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , DNA Methylation , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Gene Silencing , O(6)-Methylguanine-DNA Methyltransferase/biosynthesis , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Carcinoma, Squamous Cell/enzymology , CpG Islands , Diet , Epigenesis, Genetic , Esophageal Neoplasms/enzymology , Flavonoids , Humans , Phenols , Polymerase Chain Reaction , Polyphenols , Promoter Regions, Genetic/genetics , Tumor Cells, Cultured
9.
Carcinogenesis ; 25(6): 865-71, 2004 Jun.
Article in English | MEDLINE | ID: mdl-14764456

ABSTRACT

Gastrin-releasing peptide (GRP) is known as an autocrine growth factor for a number of gastrointestinal cancers. There is, however, little information on the expression of GRP in the squamous epithelia and squamous cell carcinoma, particularly in the esophagus. With a differential display approach, up-regulated GRP was observed in human esophageal squamous cell carcinoma (ESCC) samples obtained from a high-risk area for esophageal cancer, Linzhou in northern China. Up-regulation of phosphoglycerate mutase and P311 HUM (3.1) and down-regulation of keratin 13, cystatin B, endoglin and annexin I were observed. Using a reverse transcription-polymerase chain reaction (RT-PCR) method, significant over-expression of GRP was observed in 10 out of 12 ESCC samples (83.3%) and all four ESCC cell lines. With in situ hybridization, GRP mRNA expression was detected in nine out of 21 (42.8%) samples with basal cell hyperplasia (BCH), five out of seven (71.4%) samples with dysplasia (DYS) and 17 out of 24 (70.9%) ESCC samples. In contrast, GRP was expressed only in three out of 16 (18.7%) normal epithelium. Digital image analysis revealed that the mean value of GRP expression index, determined by intensity and area ratio of staining, was 0.19 in normal epithelium, 1.23 in BCH, 2.94 in DYS and 2.38 in ESCC, showing a progressive increase. Studies on ESCC cell lines showed GRP increased cell growth in a dose-dependent pattern in GRP receptor-positive ESCC cells, but not in GRP receptor-negative ESCC cells. GRP (1 mM) also increased cyclooxygenase-2 protein expression by 3.4-fold. This is the first demonstration that GRP is over-expressed in ESCC, and its over-expression may play a role in ESCC development and growth.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Esophageal Neoplasms/metabolism , Gastrin-Releasing Peptide/metabolism , Base Sequence , Carcinoma, Squamous Cell/pathology , Cell Transformation, Neoplastic , DNA Primers , Esophageal Neoplasms/pathology , Gastrin-Releasing Peptide/genetics , Humans , In Situ Hybridization , Reverse Transcriptase Polymerase Chain Reaction
10.
Cancer Res ; 63(22): 7563-70, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-14633667

ABSTRACT

Hypermethylation of CpG islands in the promoter regions is an important mechanism to silence the expression of many important genes in cancer. The hypermethylation status is passed to the daughter cells through the methylation of the newly synthesized DNA strand by 5-cytosine DNA methyltransferase (DNMT). We report herein that (-)-epigallocatechin-3-gallate (EGCG), the major polyphenol from green tea, can inhibit DNMT activity and reactivate methylation-silenced genes in cancer cells. With nuclear extracts as the enzyme source and polydeoxyinosine-deoxycytosine as the substrate, EGCG dose-dependently inhibited DNMT activity, showing competitive inhibition with a K(i) of 6.89 microM. Studies with structural analogues of EGCG suggest the importance of D and B ring structures in the inhibitory activity. Molecular modeling studies also support this conclusion, and suggest that EGCG can form hydrogen bonds with Pro(1223), Glu(1265), Cys(1225), Ser(1229), and Arg(1309) in the catalytic pocket of DNMT. Treatment of human esophageal cancer KYSE 510 cells with 5-50 microM of EGCG for 12-144 h caused a concentration- and time-dependent reversal of hypermethylation of p16(INK4a), retinoic acid receptor beta (RARbeta), O(6)-methylguanine methyltransferase (MGMT), and human mutL homologue 1 (hMLH1) genes as determined by the appearance of the unmethylation-specific bands in PCR. This was accompanied by the expression of mRNA of these genes as determined by reverse transcription-PCR. The re-expression of RARbeta and hMLH1 proteins by EGCG was demonstrated by Western blot. Reactivation of some methylation-silenced genes by EGCG was also demonstrated in human colon cancer HT-29 cells, esophageal cancer KYSE 150 cells, and prostate cancer PC3 cells. The results demonstrate for the first time the inhibition of DNA methylation by a commonly consumed dietary constituent and suggest the potential use of EGCG for the prevention or reversal of related gene-silencing in the prevention of carcinogenesis.


Subject(s)
Carcinoma, Squamous Cell/genetics , Catechin/analogs & derivatives , Catechin/pharmacology , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Esophageal Neoplasms/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing/drug effects , Adaptor Proteins, Signal Transducing , Anticarcinogenic Agents/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/enzymology , Carrier Proteins , Cell Division/drug effects , Cell Line, Tumor , DNA Methylation/drug effects , Esophageal Neoplasms/enzymology , Gene Expression Regulation, Neoplastic/physiology , Gene Silencing/physiology , Genes, p16/drug effects , Genes, p16/physiology , Humans , Models, Molecular , MutL Protein Homolog 1 , Neoplasm Proteins/genetics , Nuclear Proteins , O(6)-Methylguanine-DNA Methyltransferase/genetics , Receptors, Retinoic Acid/genetics
11.
Clin Cancer Res ; 9(14): 5257-63, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-14614007

ABSTRACT

PURPOSE: The purpose of this study was to investigate the mechanism of altered retinoic acid receptor beta (RARbeta) expression during esophageal squamous carcinogenesis. EXPERIMENTAL DESIGN: Samples were collected from Linzhou, China. The hypermethylation of CpG islands in the promoter region of the RARbeta gene was examined by methylation-specific PCR in human esophageal squamous cell carcinoma (ESCC) samples, as well as in neighboring tissues with normal epithelium, basal cell hyperplasia, and dysplasia. RARbeta mRNA expression was determined by in situ hybridization. The DNA methyltransferase inhibitor 2'-deoxy-5-azacytidine was used to treat the ESCC cell line, and the DNA hypermethylation status and mRNA expression level were examined. RESULTS: Two of 17 (12%) normal, 9 of 21 basal cell hyperplasia (43%), 7 of 12 dysplasia (58%), and 14 of 20 ESCC (70%) samples had hypermethylation of the RARbeta promoter region. The loss of RARbeta mRNA expression was highly concordant with RARbeta promoter CpG island hypermethylation when individual samples were considered in the correlation analysis. Good statistical correlation between hypermethylation and loss of RARbeta expression was revealed. Frequencies of hypermethylation appeared to increase with the progression of carcinogenesis. In samples from the same patients, if hypermethylation was detected in earlier lesions, it was usually observed in more severe lesions. In the ESCC cell line KYSE 510, 2'-deoxy-5-azacytidine partially reversed CpG island hypermethylation and restored RARbeta mRNA expression. CONCLUSIONS: The results suggest that hypermethylation of RARbeta promoter region is an important mechanism for RARbeta gene silencing in esophageal squamous carcinogenesis.


Subject(s)
Carcinoma, Squamous Cell/genetics , DNA Methylation , Esophageal Neoplasms/genetics , Receptors, Retinoic Acid/genetics , Carcinoma, Squamous Cell/metabolism , CpG Islands , DNA Primers , Down-Regulation , Epithelium/metabolism , Esophageal Neoplasms/metabolism , Gene Silencing , Humans , In Situ Hybridization , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Promoter Regions, Genetic/genetics , RNA, Messenger/metabolism , Receptors, Retinoic Acid/metabolism , Tumor Cells, Cultured
12.
Toxicology ; 177(2-3): 253-65, 2002 Aug 15.
Article in English | MEDLINE | ID: mdl-12135628

ABSTRACT

Cadmium (Cd), a carcinogenic metal in human and rodents, has been shown to transform cells in vitro. However, the carcinogenic mechanisms of Cd as a mutagen and/or promoter are not well clarified. We already reported that CdCl2 in a range of 1.5 approximately 360 ng/ml enhanced transformation of Balb/3T3 A31 cells induced by N-methyl-N'-nitro-N-nitrosoguanidine (MNNG, 0.1 microg/ml) in a dose-dependent manner (Fang et al., Toxicol. In Vitro 15(3) (2001a) 51-7). In previous study, we observed that Cd stimulated cell proliferation on MNNG-initiated cells through inactivation of p53 and p27 and increase of proliferating cell nuclear antigen (PCNA) expression after 24 h treatment (Fang et al., Toxicology 163 (2001b) 175-84). The aim of this study is to further elucidate the long-term effect of Cd in terms of cell cycle control gene expressions during the promotion stage of in vitro two-stage transformation. For the purpose, we determined the expression levels of the genes involved in growth regulation, such as p53, p27, c-myc, mdm2, cyclins D1 and B1, CDK4, and PCNA in the cells treated with Cd for 14 days after MNNG-initiation. In MNNG+CdCl2 group, cells apparently expressed cellular tumor antigen p53 mRNA, but did not express the wild-type p53 protein; the protein and mRNA levels of p27 were reduced apparently in the cells of MNNG+CdCl2 group compared to the cells of control and MNNG group. In addition, the protein levels of cyclin D1, CDK4, PCNA, c-myc, and mdm2, and cyclin B1 mRNA level were higher in MNNG+CdCl2 group than control and MNNG group. Together with previous data (Fang et al., Toxicology 163 (2001b) 175-84), our results indicated that during the transformation process of MNNG-treated cells, Cd may activate oncogenes such as c-myc, mdm2, and cellular tumor antigen p53, inhibit the tumor suppressor genes such as wild-type p53 and p27, and consequently accelerate the proliferation of initiated cells. This work firstly demonstrates that Cd affects the genes involved in growth regulation on initiated cells during the promotion stage of in vitro cell transformation.


Subject(s)
Cadmium/toxicity , Cell Transformation, Neoplastic/chemically induced , Muscle Proteins , Nuclear Proteins , 3T3 Cells , Animals , Cell Division/drug effects , DNA Damage , Gene Expression/drug effects , Genes, myc , Methylnitronitrosoguanidine , Mice , Microfilament Proteins/analysis , Microfilament Proteins/genetics , Proliferating Cell Nuclear Antigen/analysis , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins c-mdm2 , RNA, Messenger/analysis , Tumor Suppressor Protein p53/analysis , Tumor Suppressor Protein p53/genetics
13.
J Vet Med Sci ; 64(3): 201-5, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11999438

ABSTRACT

At present, the effect of cyclin D2 implicated in cell cycle regulation, differentiation, and oncogenic transformation is not fully confirmed. To better elucidate the role of cyclin D2 in controling the cell proliferation, cyclin D2 expression level was determined at the early initiation and promotion stages during the in vitro two-stage transformation process of Balb/3T3 A31 cells. N-methyl-N'-nitro-N-nitrosoguanidine (MNNG)-induced G2/M-arrested cells expressed low level of cyclin D2 mRNA, while the contact-inhibited nonproliferating cells expressed high level of cyclin D2 mRNA. In the transformed proliferating cells at the promotion stage, cyclin D2 mRNA was not expressed. These data suggest that cyclin D2 expression may be associated with the type of growth arrest and nonproliferating state, but not with the cell proliferation and transformation.


Subject(s)
3T3 Cells/metabolism , Cell Transformation, Neoplastic/metabolism , Cyclins/biosynthesis , 3T3 Cells/cytology , Animals , Blotting, Northern , Cell Division/drug effects , Cell Division/physiology , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/pathology , Cyclin D2 , Cyclins/genetics , G2 Phase/physiology , Methylnitronitrosoguanidine/pharmacology , Mice , Mice, Inbred BALB C , Mitosis/physiology , Proliferating Cell Nuclear Antigen/biosynthesis , Proliferating Cell Nuclear Antigen/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...