Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Oncogene ; 34(9): 1094-104, 2015 Feb 26.
Article in English | MEDLINE | ID: mdl-24662829

ABSTRACT

Most p53 mutations in human cancers are missense mutations resulting in a full-length mutant p53 protein. Besides losing tumor suppressor activity, some hotspot p53 mutants gain oncogenic functions. This effect is mediated in part, through gene expression changes due to inhibition of p63 and p73 by mutant p53 at their target gene promoters. Here, we report that the tumor suppressor microRNA let-7i is downregulated by mutant p53 in multiple cell lines expressing endogenous mutant p53. In breast cancer patients, significantly decreased let-7i levels were associated with missense mutations in p53. Chromatin immunoprecipitation and promoter luciferase assays established let-7i as a transcriptional target of mutant p53 through p63. Introduction of let-7i to mutant p53 cells significantly inhibited migration, invasion and metastasis by repressing a network of oncogenes including E2F5, LIN28B, MYC and NRAS. Our findings demonstrate that repression of let-7i expression by mutant p53 has a key role in enhancing migration, invasion and metastasis.


Subject(s)
Gene Regulatory Networks , MicroRNAs/genetics , Neoplasms/genetics , Neoplasms/pathology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line, Tumor , Cell Movement , DNA Mutational Analysis , Down-Regulation , Female , Gene Regulatory Networks/drug effects , Humans , Mice , Mice, Nude , MicroRNAs/antagonists & inhibitors , Mutation, Missense , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Neoplasm Transplantation , RNA, Small Interfering/pharmacology , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism
2.
Oncogene ; 32(35): 4139-47, 2013 Aug 29.
Article in English | MEDLINE | ID: mdl-22986525

ABSTRACT

The miR-106b-25 microRNA (miRNA) cluster is a candidate oncogene in human prostate cancer. Here, we report that miRNAs encoded by miR-106b-25 are upregulated in both primary tumors and distant metastasis. Moreover, increased tumor miR-106b expression was associated with disease recurrence and the combination of high miR-106b and low CASP7 (caspase-7) expressions in primary tumors was an independent predictor of early disease recurrence (adjusted hazard ratio=4.1; 95% confidence interval: 1.6-12.3). To identify yet unknown oncogenic functions of miR-106b, we overexpressed it in LNCaP human prostate cancer cells to examine miR-106b-induced global expression changes among protein-coding genes. The approach revealed that CASP7 is a direct target of miR-106b, which was confirmed by western blot analysis and a 3'-untranslated region reporter assay. Moreover, selected phenotypes induced by miR-106b knockdown in DU145 human prostate cancer cells did not develop when both miR-106b and CASP7 expression were inhibited. Further analyses showed that CASP7 is downregulated in primary prostate tumors and metastatic lesions across multiple data sets and is by itself associated with disease recurrence and disease-specific survival. Using bioinformatics, we also observed that miR-106b-25 may specifically influence focal adhesion-related pathways. This observation was experimentally examined using miR-106b-25-transduced 22Rv1 human prostate cancer cells. After infection with a miR-106b-25 lentiviral expression construct, 22Rv1 cells showed increased adhesion to basement membrane- and bone matrix-related filaments and enhanced soft agar growth. In summary, miR-106b-25 was found to be associated with prostate cancer progression and disease outcome and may do so by altering apoptosis- and focal adhesion-related pathways.


Subject(s)
Caspase 7/genetics , Focal Adhesions , Gene Expression Regulation, Neoplastic , MicroRNAs/physiology , Neoplasm Recurrence, Local/etiology , Prostatic Neoplasms/genetics , 3' Untranslated Regions , Cell Line, Tumor , Extracellular Matrix/metabolism , Humans , Male , Neoplasm Metastasis , Prostatic Neoplasms/etiology , Prostatic Neoplasms/pathology
3.
Cancer Res ; 60(13): 3333-7, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10910033

ABSTRACT

Ulcerative colitis (UC) is a chronic inflammatory disease that produces reactive oxygen and nitrogen species and increases the risk of colorectal cancer (CRC). The p53 tumor suppressor gene is frequently mutated in UC-associated dysplastic lesions and CRC. We are exploring the hypothesis that p53 mutations in the nontumorous colonic tissue in noncancerous UC cases indicate genetic damage from exposure to exogenous and endogenous carcinogens and may identify individuals at increased cancer risk. We are reporting, for the first time, the frequency of specific p53 mutated alleles in nontumorous colon tissue from donors either with or without UC by using a highly sensitive genotypic mutation assay. Higher p53 mutation frequencies of both G:C to A:T transitions at the CpG site of codon 248 and C:G to T:A transitions at codon 247 were observed in colon from UC cases when compared with normal adult controls (P = 0.001 and P = 0.001, respectively). In the UC cases, higher p53 codon 247 and 248 mutation frequencies were observed in the inflamed lesional regions when compared with the nonlesional regions of their colon (P < 0.001 and P = 0.001). The colonic nitric oxide synthase-2 activity was higher in UC cases than in non-UC adult controls (P = 0.02). Our data are consistent with the hypothesis that a higher frequency of p53 mutant cells can be generated under oxidative stress in people with UC. The increased frequency of specific p53 mutated alleles in noncancerous UC colon tissue may confer susceptibility to the development of CRC in an inflammatory microenvironment.


Subject(s)
Colitis, Ulcerative/genetics , Colonic Neoplasms/genetics , Genes, p53 , Point Mutation , Adult , Codon , Colitis, Ulcerative/complications , Colitis, Ulcerative/pathology , Colon/pathology , Colonic Neoplasms/etiology , Dinucleoside Phosphates/genetics , Genetic Predisposition to Disease , Genotype , Humans , Intestinal Mucosa/pathology , Nitric Oxide Synthase/analysis , Nitric Oxide Synthase Type II , Reference Values
4.
Cancer Res ; 59(22): 5773-7, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10582698

ABSTRACT

Although the therapeutic value of angiostatin, a proteolytic fragment of plasminogen, has been recognized for the treatment of cancer, the production of bioactive angiostatin remains a difficult task. Here we report that expression of a cDNA encoding a secreted, four-kringle human angiostatin inhibited tumor growth of B16F10 melanoma cells in mice but did not suppress tumor cell growth in culture. After transfection and selection, stable expression of the angiostatin cDNA was demonstrated in several B16F10 clones by quantitative mRNA analysis using the Taqman method. Cells that expressed angiostatin at either a low, medium, or high level were injected into C57BL/6 mice. s.c. Growth of B16F10 tumors was diminished by the angiostatin transgene, and the inhibition was directly proportional to the expression level of angiostatin in the transfected cells. However, suppression of s.c. tumor growth was transient, and eventually, tumors emerged with a strongly decreased expression of the transgene. Angiostatin expression also reduced lung metastasis from i.v.-injected B16F10 cells. Our data indicate that a cDNA encoding bioactive human angiostatin is potentially useful for gene therapy of human cancers, but the delivery of the transgene may require repeated dosing to achieve sustained dormancy of primary tumors and cancer metastases.


Subject(s)
Antineoplastic Agents/therapeutic use , Kringles , Melanoma, Experimental/drug therapy , Peptide Fragments/therapeutic use , Plasminogen/therapeutic use , Angiostatins , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , DNA, Complementary/metabolism , Drug Screening Assays, Antitumor , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Melanoma, Experimental/metabolism , Melanoma, Experimental/secondary , Mice , Mice, Inbred C57BL , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Plasmids/therapeutic use , Plasminogen/chemistry , Plasminogen/genetics , Plasminogen/metabolism , Transfection , Transgenes , Tumor Cells, Cultured/drug effects
7.
IARC Sci Publ ; (150): 295-302, 1999.
Article in English | MEDLINE | ID: mdl-10626229

ABSTRACT

Oxyradical overload disease develops in conditions involving chronic inflammation and may be of inherited etiology, e.g. haemochromatosis and Wilson disease, be acquired, e.g. infection with hepatitis B or C virus or Helicobactor pylori, or be chemically induced, e.g. acid reflux in Barrett oesophagus. Susceptibility to cancer is frequently a pathological consequence of extensive oxyradical damage that leads to a cycle of cell death and regeneration and causes mutations in cancer-related genes. In this brief review, we focus on the possible interactive effects of nitric oxide and the p53 tumour suppressor gene in human carcinogenesis.


Subject(s)
Colonic Neoplasms/etiology , Nitric Oxide/metabolism , Apoptosis , Colonic Neoplasms/genetics , DNA Damage , Disease Progression , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Genes, p53/genetics , Humans , Models, Biological , Mutation , Nitric Oxide Synthase/metabolism
8.
Nat Med ; 4(12): 1371-6, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9846573

ABSTRACT

The finding of frequent nitric oxide synthase expression in human cancers indicates that nitric oxide has a pathophysiological role in carcinogenesis. To determine the role of nitric oxide in tumor progression, we generated human carcinoma cell lines that produced nitric oxide constitutively. Cancer cells expressing inducible nitric oxide synthase that had wild-type p53 had reduced tumor growth in athymic nude mice, whereas those with mutated p53 had accelerated tumor growth associated with increased vascular endothelial growth factor expression and neovascularization. Our data indicate that tumor-associated nitric oxide production may promote cancer progression by providing a selective growth advantage to tumor cells with mutant p53, and that inhibitors of inducible nitric oxide synthase may have therapeutic activity in these tumors.


Subject(s)
Endothelial Growth Factors/physiology , Lymphokines/physiology , Neoplasms, Experimental/enzymology , Neoplasms, Experimental/pathology , Nitric Oxide Synthase/biosynthesis , Tumor Suppressor Protein p53/physiology , Animals , Apoptosis , Gene Transfer Techniques , Humans , Mice , Neoplasm Transplantation , Neovascularization, Pathologic , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Transplantation, Heterologous , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
9.
Proc Natl Acad Sci U S A ; 95(15): 8823-8, 1998 Jul 21.
Article in English | MEDLINE | ID: mdl-9671763

ABSTRACT

High concentrations of nitric oxide (NO) cause DNA damage and apoptosis in many cell types. Thus, regulation of NO synthase (NOS) activity is essential for minimizing effects of cytotoxic and genotoxic nitrogen oxide species. We have shown previously that NO-induced p53 protein accumulation down-regulates basal and cytokine-modulated inducible NOS (NOS2) expression in human cells in vitro. To further characterize the feedback loop between NOS2 and p53, we have investigated NO production, i.e., urinary nitrate plus nitrite excretion, and NOS2 expression in homozygous p53 knockout (KO) mice. We report here that untreated p53 KO mice excreted 70% more nitrite plus nitrate than mice with wild-type (wt) p53. NOS2 protein expression was constitutively detected in the spleen of untreated p53 KO mice, whereas it was undetectable in the spleen of wt p53 controls. Upon treatment with heat-inactivated Corynebacterium parvum, urinary nitrite plus nitrate excretion of p53 KO mice exceeded that of wt controls by approximately 200%. C. parvum treatment also induced p53 accumulation in the liver. Splenectomy reduced the NO output of C. parvum-treated p53 KO mice but not of wt p53 controls. Although NO production and NOS2 protein expression were increased similarly in KO and wt p53 mice 10 days after injection of C. parvum, NOS2 expression returned to baseline levels only in wt p53 controls while remaining up-regulated in p53 KO mice. These genetic and functional data indicate that p53 is an important transrepressor of NOS2 expression in vivo and attenuates excessive NO production in a regulatory negative feedback loop.


Subject(s)
Gene Expression Regulation, Enzymologic , Neoplasms, Experimental/genetics , Nitric Oxide Synthase/genetics , Up-Regulation , Animals , Feedback , Genetic Predisposition to Disease , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II , Promoter Regions, Genetic
10.
Br J Cancer ; 78(2): 233-9, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9683299

ABSTRACT

Vascular endothelial growth factor (VEGF) expression and mutations of cancer-related genes increase with cancer progression. This correlation suggests the hypothesis that oncogenes and tumour suppressors regulate VEGF, and a significant correlation between p53 alteration and increased VEGF expression in human lung cancer was reported recently. To further examine this hypothesis, we analysed VEGF protein expression and mutations in p53 and K-ras in 27 non-small-cell lung cancers (NSCLC): 16 squamous cell, six adenocarcinomas, one large cell, two carcinoids and two undifferentiated tumours. VEGF was expressed in 50% of the squamous cell carcinomas (SCC) and carcinoids but none of the others. p53 mutations occurred in 14 tumours (52%), and K-ras mutations were found in two adenocarcinomas and one SCC; there was no correlation between the mutations and VEGF expression. As nitric oxide also regulates angiogenesis, we examined NOS expression in NSCLC. The Ca2+-dependent NOS activity, which indicates NOS1 and NOS3 expression, was significantly reduced in lung carcinomas compared with adjacent non-tumour tissue (P < 0.004). Although the Ca2+-independent NOS activity, which indicates NOS2 expression, was low or undetectable in non-tumour tissues and most carcinomas, significant activity occurred in three SCC. In summary, our data do not show a direct regulation of VEGF by p53 in NSCLC. Finally, we did not find the up-regulation of NOS isoforms during NSCLC progression that has been suggested for gynaecological and breast cancers.


Subject(s)
Endothelial Growth Factors/analysis , Genes, p53 , Lung Neoplasms/chemistry , Lymphokines/analysis , Nitric Oxide Synthase/analysis , Carcinoma, Small Cell/chemistry , Carcinoma, Small Cell/genetics , Carcinoma, Squamous Cell/chemistry , Carcinoma, Squamous Cell/genetics , Genes, p53/physiology , Genes, ras , Humans , Lung Neoplasms/genetics , Mutation , Nitric Oxide Synthase Type II , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
11.
Cancer Res ; 58(2): 334-41, 1998 Jan 15.
Article in English | MEDLINE | ID: mdl-9443414

ABSTRACT

An increased expression of nitric oxide synthase (NOS) has been observed in human colon carcinoma cell lines as well as in human gynecological, breast, and central nervous system tumors. This observation suggests a pathobiological role of tumor-associated NO production. Hence, we investigated NOS expression in human colon cancer in respect to tumor staging, NOS-expressing cell type(s), nitrotyrosine formation, inflammation, and vascular endothelial growth factor expression. Ca2+-dependent NOS activity was found in normal colon and in tumors but was significantly decreased in adenomas (P < 0.001) and carcinomas (Dukes' stages A-D: P < 0.002). Ca2+-independent NOS activity, indicating inducible NOS (NOS2), is markedly expressed in approximately 60% of human colon adenomas (P < 0.001 versus normal tissues) and in 20-25% of colon carcinomas (P < 0.01 versus normal tissues). Only low levels were found in the surrounding normal tissue. NOS2 activity decreased with increasing tumor stage (Dukes' A-D) and was lowest in colon metastases to liver and lung. NOS2 was detected in tissue mononuclear cells (TMCs), endothelium, and tumor epithelium. There was a statistically significant correlation between NOS2 enzymatic activity and the level of NOS2 protein detected by immunohistochemistry (P < 0.01). Western blot analysis of tumor extracts with Ca2+-independent NOS activity showed up to three distinct NOS2 protein bands at Mr 125,000-Mr 138,000. The same protein bands were heavily tyrosine-phosphorylated in some tumor tissues. TMCs, but not the tumor epithelium, were immunopositive using a polyclonal anti-nitrotyrosine antibody. However, only a subset of the NOS2-expressing TMCs stained positively for 3-nitrotyrosine, which is a marker for peroxynitrite formation. Furthermore, vascular endothelial growth factor expression was detected in adenomas expressing NOS2. These data are consistent with the hypothesis that excessive NO production by NOS2 may contribute to the pathogenesis of colon cancer progression at the transition of colon adenoma to carcinoma in situ.


Subject(s)
Adenoma/enzymology , Colonic Neoplasms/enzymology , Neovascularization, Pathologic/enzymology , Nitric Oxide Synthase/metabolism , Adenoma/blood supply , Adenoma/pathology , Blotting, Western , Carcinoma/blood supply , Carcinoma/enzymology , Carcinoma/pathology , Colon/enzymology , Colonic Neoplasms/blood supply , Colonic Neoplasms/pathology , DNA Primers/chemistry , DNA, Neoplasm/analysis , Disease Progression , Endothelial Growth Factors/metabolism , Endothelium, Vascular/enzymology , Humans , Immunohistochemistry , Lymphokines/metabolism , Neoplasm Proteins/analysis , Nitric Oxide Synthase Type II , Phosphorylation , Polymerase Chain Reaction , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
12.
FASEB J ; 11(6): 443-8, 1997 May.
Article in English | MEDLINE | ID: mdl-9194524

ABSTRACT

The tumor suppressor gene product p53 plays an important role in the cellular response to DNA damage. DNA damage can lead to p53-mediated growth arrest and apoptosis. High concentrations of nitric oxide (NO) and NO metabolites such as peroxynitrite and NO2 cause DNA damage and have been shown to be mutagenic. Furthermore, NO induces p53 accumulation and, as part of a feedback loop, p53 mediates transcriptional transrepression of inducible nitric oxide synthase. Recent studies have shown increased expression and activity of nitric oxide synthase isoforms in human cancer. NO has both genotoxic and angiogenic properties, so that increased NO production may select mutant p53 cells and contribute to human carcinogenesis and tumor progression.


Subject(s)
Genes, p53/physiology , Neoplasms/genetics , Neoplasms/metabolism , Nitric Oxide/physiology , Animals , Disease Progression , Humans
13.
J Surg Res ; 69(2): 321-4, 1997 May.
Article in English | MEDLINE | ID: mdl-9224400

ABSTRACT

Chronic inflammatory states frequently lead to the increased production of nitric oxide (NO) via inducible NO synthase (NOS-2). In addition, NO may produce mutagenesis through several mechanisms such as DNA oxidation, DNA deamination, and the formation of N-nitroso compounds. As there is a strong association between human hepatitis C virus (HCV) infection and the development of hepatocellular carcinoma (HCC), we were interested in whether human HCV hepatitis leads to induction of NOS-2 and if the mutation repair system of p53/p21 was upregulated. Reverse transcriptase-polymerase chain reaction (RT-PCR) for human NOS-2 message was performed on RNA samples from both liver biopsies and whole liver from HCV-positive and control patients (normal liver from hepatic resections for metastases). Immunohistochemistry (IHC) for p53 and Western blot analysis for p21 were also performed on the whole liver samples. From the liver biopsies, 60% of HCV-positive patients expressed NOS-2 by RT-PCR. Looking at the whole liver samples, 100% of the HCV-positive patients expressed NOS-2 vs 12.5% in the normal samples. p53 was not detected in either group but there was upregulation of p21 over baseline expression in a number of the HCV-positive patients. Human HCV hepatitis leads to consistent upregulation of hepatic NOS-2 message, but message is not predictably present in "normal" human liver. There is also induction of p21 in some patients with HCV hepatitis. Chronic expression of NO in HCV hepatitis may play a role in DNA mutagenesis and the development of HCC.


Subject(s)
Hepatitis C/complications , Hepatitis C/enzymology , Liver Neoplasms/virology , Nitric Oxide Synthase/biosynthesis , Adult , Chronic Disease , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Enzyme Induction , Female , Hepacivirus , Humans , Male , RNA, Messenger/genetics , Tumor Suppressor Protein p53/metabolism
14.
Toxicol Appl Pharmacol ; 139(1): 186-94, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8685902

ABSTRACT

Isolated perfused livers from male Wistar rats were used to study acute and chronic toxic effects of carcinogenic aromatic amines. We investigated the hypothesis that aromatic amines can generate reactive oxygen species as part of their metabolism. Concentrations of 200-400 microM of 2-acetylaminofluorene (AAF), N-hydroxy-AAF, trans-4-acetylaminostilbene (AAS), N-hydroxy-AAS, and N-hydroxy-2-acetylaminophenanthrene in the recirculating perfusate were not toxic in a 2-hr exposure time as assessed by LDH efflux into the perfusate, glutathione excretion into bile, and changes of the beta-hydroxybutyrate/acetoacetate ratio in the perfusate. N-Acetoxy-AAF, however, was severely toxic. Menadione served as a positive control. It is concluded that exposures likely to occur in carcinogenicity studies with these aromatic amines will not be acutely toxic. In additional experiments the isolated perfused liver system was used to demonstrate chronic effects generated by feeding the carcinogenic dose of 0.02% AAF for up to 12 weeks. The following alterations were observed in livers from AAF-fed animals. excretion of glutathione into bile is drastically reduced after 5 or more weeks, increasingly less glucose is released into the perfusate, and oxygen consumption is constantly increased by 20% after 3 and more weeks of AAF feeding. Whereas the total glutathione level increased with time in homogenates of such livers, it decreased in the mitochondrial fraction. The results are interpreted as adaptive responses to chronic toxic effects of AAF which may be related to the promoting properties of this carcinogen.


Subject(s)
Amines/toxicity , Carcinogens/toxicity , Liver/drug effects , 2-Acetylaminofluorene/metabolism , 2-Acetylaminofluorene/toxicity , Amines/metabolism , Animals , Glutathione/metabolism , Hemostatics/pharmacology , Hydroxyacetylaminofluorene/metabolism , Hydroxyacetylaminofluorene/toxicity , Isotope Labeling , Liver/metabolism , Male , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Oxygen Consumption/drug effects , Perfusion , Phenanthrenes/metabolism , Phenanthrenes/toxicity , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Stilbenes/metabolism , Stilbenes/toxicity , Vitamin K/pharmacology
15.
Proc Natl Acad Sci U S A ; 93(6): 2442-7, 1996 Mar 19.
Article in English | MEDLINE | ID: mdl-8637893

ABSTRACT

The tumor suppressor gene product p53 plays an important role in the cellular response to DNA damage from exogenous chemical and physical mutagens. Therefore, we hypothesized that p53 performs a similar role in response to putative endogenous mutagens, such as nitric oxide (NO). We report here that exposure of human cells to NO generated from an NO donor or from overexpression of inducible nitric oxide synthase (NOS2) results in p53 protein accumulation. In addition, expression of wild-type (WT) p53 in a variety of human tumor cell lines, as well as murine fibroblasts, results in down-regulation of NOS2 expression through inhibition of the NOS2 promoter. These data are consistent with the hypothesis of a negative feedback loop in which endogenous NO-induced DNA damage results in WT p53 accumulation and provides a novel mechanism by which p53 safeguards against DNA damage through p53-mediated transrepression of NOS2 gene expression, thus reducing the potential for NO-induced DNA damage.


Subject(s)
Nitric Oxide Synthase/biosynthesis , Nitric Oxide/physiology , Tumor Suppressor Protein p53/physiology , Animals , Cells, Cultured , Colon/enzymology , DNA Damage , Down-Regulation , Enzyme Induction , Gene Expression Regulation, Enzymologic , Humans , Liver/enzymology , Mice , Promoter Regions, Genetic , Repressor Proteins/physiology , Tumor Cells, Cultured
16.
Carcinogenesis ; 16(9): 2069-74, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7554056

ABSTRACT

Nitric oxide (NO) is a cellular messenger which is mutagenic in bacteria and human TK6 cells and induces deamination of 5-methylcytosine (5meC) residues in vitro. The aims of this study were: (i) to investigate whether NO induces 5meC deamination in codon 248 of the p53 gene in cultured human bronchial epithelial cells (BEAS-2B); and (ii) to compare NO mutagenicity to that of ethylnitrosourea (ENU), a strong mutagen. Two approaches were used: (i) a novel genotypic assay, using RFLP/PCR technology on purified exon VII sequence of the p53 gene; and (ii) a phenotypic (HPRT) mutation assay using 6-thioguanine selection. BEAS-2B cells were either exposed to 4 mM DEA/NO (Et2N[N2O2]Na, an agent that spontaneously releases NO into the medium) or transfected with the inducible nitric oxide synthase (iNOS) gene. The genotypic mutation assay, which has a sensitivity of 1 x 10(-6), showed that 4 mM ENU induces detectable numbers of G --> A transitions in codon 248 of p53 while 5-methylcytosine deamination was not detected in either iNOS-transfected cells or cells exposed to 4 mM DEA/NO. Moreover, ENU was dose-responsively mutagenic in the phenotypic HPRT assay, reaching mutation frequencies of 24 and 96 times that of untreated control cells at ENU concentrations of 4 and 8 mM respectively; by contrast, 4 mM DEA/NO induced no detectable mutations in this assay, nor were any observed in cells transfected with murine iNOS. We conclude that if NO is at all promutagenic in these cells, it is significantly less so than the ethylating mutagen, ENU.


Subject(s)
Ethylnitrosourea/toxicity , Genes, p53/drug effects , Hypoxanthine Phosphoribosyltransferase/genetics , Mutagens/toxicity , Nitric Oxide/toxicity , Base Sequence , Bronchi/drug effects , Bronchi/metabolism , Bronchi/physiology , Cell Line , Codon , Cyclic GMP/biosynthesis , Cytosine/metabolism , Diethylamines/toxicity , Humans , Hypoxanthine Phosphoribosyltransferase/drug effects , Male , Methylation , Molecular Sequence Data , Mutagenicity Tests , Mutation , Nitrogen Oxides , Phenotype , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length , Transfection
17.
Toxicol In Vitro ; 9(2): 117-21, 1995 Apr.
Article in English | MEDLINE | ID: mdl-20650070

ABSTRACT

The aneuploidy-inducing carcinogenic oestrogen diethylstilboestrol (DES) causes an elevation of the cytoplasmic Ca(2+) concentration of C6 rat glioma cells. It is known that the intracellular calcium concentration is a critical factor in mitosis. DES induces mitotic disturbances resulting in aneuploidy; the mechanisms of these events are still largely unknown. It is therefore important to identify the calcium source for the DES-mediated calcium rise. Experiments with isolated rat liver mitochondria showed no release of calcium after DES treatment. However, DES releases calcium from the same stores as the tumour promotor thapsigargin, which are the inositol 1,4,5-triphosphate- and GTP-sensitive Ca(2+) stores. In contrast to DES, thapsigargin also induces a calcium influx into the cell. Administration of DES following thapsigargin treatment strongly reduced the thapsigargin-mediated calcium entry. Furthermore, DES inhibits the decay of the ionomycin-induced calcium signal, suggesting an interference of DES with the extrusion of calcium from the cell. In conclusion, thapsigargin and DES both cause release of calcium from the endoplasmic reticulum. However, for each compound the prolongation of the calcium signal is based on different mechanisms.

18.
Environ Health Perspect ; 102 Suppl 6: 173-6, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7889843

ABSTRACT

The growth of preneoplastic nodules during the feeding of a carcinogenic 2-acetylaminofluorene (2-AAF) regimen is preceded by several alterations in the physiologic homeostasis. Many of these alterations can be considered adaptive responses to the drug exposure. One property of AAF could be identified that clearly distinguishes this complete rat liver carcinogen from at least two other, incomplete rat liver carcinogens. Highly specific redox cycling in mitochondria was demonstrated in vitro, and this observation could well contribute an explanation of the morphologic and histochemical observations in vivo. It is emphasized that nongenotoxic effects may play an important role in the generation of tumors by genotoxic carcinogens.


Subject(s)
Amines/toxicity , Carcinogens/toxicity , 2-Acetylaminofluorene/toxicity , Animals , Oxidation-Reduction , Phenanthrenes/toxicity , Rats , Stilbenes/toxicity
19.
Am J Respir Cell Mol Biol ; 11(2): 159-64, 1994 Aug.
Article in English | MEDLINE | ID: mdl-7519434

ABSTRACT

Two major roles have been defined for nitric oxide (NO): cell-cell communication mediated by the stimulation of cyclic guanosine 3',5'-monophosphate (cGMP) synthesis and cytotoxicity by direct or indirect interaction of the free radical NO with cellular targets. Thus, pathologic states might result from an alteration of NO pathways, e.g., by deregulated activity of NO synthase. To investigate this hypothesis, we introduced the murine-inducible NO synthase (iNOS) sequence into immortalized human bronchial epithelial cells (BEAS-2B). iNOS activity, measured by conversion of [14C]arginine to [14C]citrulline in the presence of 1 mM EGTA, was higher than 100 pmol/min/mg protein in early passages of iNOS-transfected cells but decreased with cell subculturing. No iNOS activity could be detected in control vector-transfected cells. NO stimulated cGMP production in iNOS-transfected cells, and this effect was inhibited by the iNOS inhibitor NG-monomethyl-L-arginine. In addition, NO production induced c-fos expression and did not interfere with clonal cell growth. These results suggest that BEAS-2B cells constitute a suitable model to study the consequences of iNOS activity on signal transduction pathways in bronchial epithelium.


Subject(s)
Amino Acid Oxidoreductases/biosynthesis , Bronchi/metabolism , Cyclic GMP/metabolism , Gene Expression , Genes, fos , Proto-Oncogene Proteins c-fos/biosynthesis , Animals , Antigens, Polyomavirus Transforming/biosynthesis , Arginine/metabolism , Base Sequence , Blotting, Northern , Carbon Radioisotopes , Cell Division , Cell Line, Transformed , Citrulline/metabolism , DNA Primers , Enzyme Induction , Epithelium/metabolism , Humans , Kinetics , Mice , Molecular Sequence Data , Nitric Oxide Synthase , Polymerase Chain Reaction , Simian virus 40/genetics , Transfection
20.
Carcinogenesis ; 11(11): 2063-6, 1990 Nov.
Article in English | MEDLINE | ID: mdl-2121383

ABSTRACT

Aflatoxin B1 (AFB1) exposure from the diet is a major risk factor for the development of liver cancer in people living in regions of China and Africa. Rapid methods to assess the exposure status of these individuals to genotoxic damage imparted by AFB1 will be very important for cancer prevention strategies. Serum albumin is a readily accessible target protein for AFB1 and we report here the development of an accurate and sensitive method to quantitate the major AFB1 serum albumin adduct, aflatoxin-lysine, from less than 100 microliters of serum by combined immunoaffinity chromatography/high-performance liquid chromatography (IAC/HPLC) with fluorescence detection. For this method, serum is digested with Pronase and the adducts are purified by monoclonal antibody IAC and quantified by HPLC. Analysis of human serum samples obtained from an exposed population revealed a highly significant correlation coefficient (up to 0.82 for male samples) between aflatoxin-lysine adduct levels and AFB1 consumption. These data suggest that aflatoxin-lysine is an excellent molecular dosimeter for exposure assessment. To determine whether the liver is the sole site of aflatoxin-albumin adduct formation, preliminary experiments with isolated perfused rat liver were done. These data showed that AFB1 metabolites covalently react not only with albumin in the hepatocyte, but also with circulating proteins in the perfusate. This suggests that a reactive aflatoxin metabolite secreted by the liver may form serum albumin adducts in circulating blood. Taken together, the analysis of aflatoxin-lysine could prove a very useful tool for epidemiological studies.


Subject(s)
Aflatoxin B1 , Aflatoxins/analysis , Lysine/analysis , Serum Albumin/chemistry , Chromatography, High Pressure Liquid/methods , Environmental Exposure , Female , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL