Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 293(10): 3663-3674, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29358331

ABSTRACT

The pluripotency-controlling stem-cell protein SRY-box 2 (SOX2) plays a pivotal role in maintaining the self-renewal and pluripotency of embryonic stem cells and also of teratocarcinoma or embryonic carcinoma cells. SOX2 is monomethylated at lysine 119 (Lys-119) in mouse embryonic stem cells by the SET7 methyltransferase, and this methylation triggers ubiquitin-dependent SOX2 proteolysis. However, the molecular regulators and mechanisms controlling SET7-induced SOX2 proteolysis are unknown. Here, we report that in human ovarian teratocarcinoma PA-1 cells, methylation-dependent SOX2 proteolysis is dynamically regulated by the LSD1 lysine demethylase and a methyl-binding protein, PHD finger protein 20-like 1 (PHF20L1). We found that LSD1 not only removes the methyl group from monomethylated Lys-117 (equivalent to Lys-119 in mouse SOX2), but it also demethylates monomethylated Lys-42 in SOX2, a reaction that SET7 also regulated and that also triggered SOX2 proteolysis. Our studies further revealed that PHF20L1 binds both monomethylated Lys-42 and Lys-117 in SOX2 and thereby prevents SOX2 proteolysis. Down-regulation of either LSD1 or PHF20L1 promoted SOX2 proteolysis, which was prevented by SET7 inactivation in both PA-1 and mouse embryonic stem cells. Our studies also disclosed that LSD1 and PHF20L1 normally regulate the growth of pluripotent mouse embryonic stem cells and PA-1 cells by preventing methylation-dependent SOX2 proteolysis. In conclusion, our findings reveal an important mechanism by which the stability of the pluripotency-controlling stem-cell protein SOX2 is dynamically regulated by the activities of SET7, LSD1, and PHF20L1 in pluripotent stem cells.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Histone Demethylases/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Neoplasm Proteins/metabolism , Ovarian Neoplasms/metabolism , Protein Processing, Post-Translational , SOXB1 Transcription Factors/metabolism , Amino Acid Substitution , Animals , Cell Line, Tumor , Cells, Cultured , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , HEK293 Cells , Histone Demethylases/antagonists & inhibitors , Histone Demethylases/chemistry , Histone Demethylases/genetics , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/genetics , Humans , Methylation , Mice, Inbred C57BL , Mutation , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology , Protein Stability , Proteolysis , RNA Interference , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , SOXB1 Transcription Factors/chemistry , SOXB1 Transcription Factors/genetics , Teratocarcinoma/enzymology , Teratocarcinoma/metabolism , Teratocarcinoma/pathology
2.
Nucleic Acids Res ; 44(10): 4665-83, 2016 06 02.
Article in English | MEDLINE | ID: mdl-26883630

ABSTRACT

Cell differentiation is a central process in development and in cancer growth and dissemination. OCT4 (POU5F1) and NANOG are essential for cell stemness and pluripotency; yet, the mechanisms that regulate their expression remain largely unknown. Repetitive elements account for almost half of the Human Genome; still, their role in gene regulation is poorly understood. Here, we show that the dioxin receptor (AHR) leads to differentiation of human carcinoma cells through the transcriptional upregulation of Alu retrotransposons, whose RNA transcripts can repress pluripotency genes. Despite the genome-wide presence of Alu elements, we provide evidences that those located at the NANOG and OCT4 promoters bind AHR, are transcribed by RNA polymerase-III and repress NANOG and OCT4 in differentiated cells. OCT4 and NANOG repression likely involves processing of Alu-derived transcripts through the miRNA machinery involving the Microprocessor and RISC. Consistently, stable AHR knockdown led to basal undifferentiation, impaired Alus transcription and blockade of OCT4 and NANOG repression. We suggest that transcripts produced from AHR-regulated Alu retrotransposons may control the expression of stemness genes OCT4 and NANOG during differentiation of carcinoma cells. The control of discrete Alu elements by specific transcription factors may have a dynamic role in genome regulation under physiological and diseased conditions.


Subject(s)
Alu Elements , Basic Helix-Loop-Helix Transcription Factors/physiology , Cell Differentiation/genetics , Gene Expression Regulation, Neoplastic , Receptors, Aryl Hydrocarbon/physiology , Teratocarcinoma/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma/pathology , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Nucleus/metabolism , Humans , Mice , MicroRNAs/metabolism , Nanog Homeobox Protein/genetics , Octamer Transcription Factor-3/genetics , Promoter Regions, Genetic , RNA Polymerase III/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Teratocarcinoma/enzymology , Teratocarcinoma/metabolism , Teratocarcinoma/pathology , Teratoma/genetics , Teratoma/metabolism , Transcription, Genetic , Tretinoin/pharmacology
3.
J Mol Biol ; 377(1): 28-46, 2008 Mar 14.
Article in English | MEDLINE | ID: mdl-18237746

ABSTRACT

Rex1 (Zfp42), first identified as a gene that is transcriptionally repressed by retinoic acid (RA), encodes a zinc finger transcription factor expressed at high levels in F9 teratocarcinoma stem cells, embryonic stem cells, and other stem cells. Loss of both alleles of Rex1 by homologous recombination alters the RA-induced differentiation of F9 cells, a model of pluripotent embryonic stem cells. We identified Suppressor of Cytokine Signaling-3 (SOCS-3) as a gene that exhibits greatly increased transcriptional activation in RA, cAMP, and theophylline (RACT)-treated F9 Rex1(-/-) cells (approximately 25-fold) as compared to wild-type (WT) cells ( approximately 2.5-fold). By promoter deletion, mutation, and transient transfection analyses, we have shown that this transcriptional increase is mediated by the STAT3 DNA-binding elements located between -99 to -60 in the SOCS-3 promoter. Overexpression of STAT3 dominant-negative mutants greatly diminishes this SOCS-3 transcriptional increase in F9 Rex1(-/-) cells. This increase in SOCS-3 transcription is associated with a four- to fivefold higher level of tyrosine-phosphorylated STAT3 in the RACT-treated F9 Rex1(-/-) cells as compared to WT. Dominant-negative Src tyrosine kinase, Jak2, and protein kinase A partially reduce the transcriptional activation of the SOCS 3 gene in RACT-treated F9 Rex1 null cells. In contrast, parathyroid hormone peptide enhances the effect of RA in F9 Rex1(-/-) cells, but not in F9 WT. Thus, Rex1, which is highly expressed in stem cells, inhibits signaling via the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway, thereby modulating the differentiation of F9 cells.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplastic Stem Cells/metabolism , STAT3 Transcription Factor/genetics , Suppressor of Cytokine Signaling Proteins/genetics , Teratocarcinoma/genetics , Transcription Factors/deficiency , Transcriptional Activation/genetics , Animals , Binding Sites , Bucladesine/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Embryonal Carcinoma Stem Cells , Genes, Reporter , Janus Kinase 2/metabolism , Mercaptoethanol/pharmacology , Mice , Microarray Analysis , Neoplastic Stem Cells/drug effects , Parathyroid Hormone/pharmacology , Peptides/pharmacology , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins pp60(c-src)/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Retinoic Acid/metabolism , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/metabolism , Teratocarcinoma/enzymology , Theophylline/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects , Tretinoin/pharmacology
4.
Mol Cell Biochem ; 274(1-2): 31-7, 2005 Jun.
Article in English | MEDLINE | ID: mdl-16335526

ABSTRACT

Knocking out the regulatory beta subunit of protein kinase CK2 in mice leads to early embryonic lethality. Heterozygous CK2beta (CK2beta+/-) knockout mice do not show an obvious phenotype. However, the number of heterozygous offsprings from CK2B+/- inter-crossings is lower than expected, meaning that some heterozygous embryos do not survive. Interestingly, CK2beta+/- ES (Embryonic Stem) cells express a considerably lower level of CK2beta than wild-type ES cells, whereas the level of CK2beta in organs from heterozygous adult mice does not significantly differ from those of wild-type mice. The data suggest a compensatory mechanism that adjusts CK2beta levels during development in the majority of, but not in all, cases (Mol Cell Biol 23: 908-915, 2003). In order to find an explanation for the gene dosage effect observed for heterozygous offsprings, we analysed embryos at mid-gestation (E10.5) as well as wild-type and CK2beta+/- ES cells for differences in growth rate and response to different stress agents. Analysis of E10.5 embryos generated from heterozygous matings revealed about 20% of smaller retarded CK2beta+/- embryos. No correlation between CK2beta levels in normal looking and retarded CK2beta+/- embryos were found. However, a different post-translational form of CK2beta protein has been detected in these retarded embryos. Cellular parameters such as growth rate and G1-, G2-checkpoints in ES cells were identical in both wild-type and CK2beta+/- cells. When ES cells were injected to induce differentiated teratocarcinoma in syngenic mice, the size of the tumours correlated with the level of CK2beta.


Subject(s)
Casein Kinase II/metabolism , Embryo, Mammalian/abnormalities , Gene Dosage , Protein Subunits/metabolism , Stem Cells/physiology , Animals , Casein Kinase II/genetics , Cell Cycle , Embryo, Mammalian/enzymology , Heterozygote , Mice , Mice, Knockout , Phenotype , Protein Subunits/genetics , Stem Cells/enzymology , Teratocarcinoma/enzymology , Teratocarcinoma/pathology
5.
Toxicol In Vitro ; 19(1): 1-10, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15582350

ABSTRACT

Nitrofen is a diphenyl ether herbicide that produces a spectrum of fetal abnormalities in rodents. To characterize the molecular mechanisms of nitrofen-mediated birth defects at the cellular level, we explored its effects on undifferentiated P19 teratocarcinoma cells. Nitrofen induces a time-dependent cell death of P19 cells that is associated with increases in TUNEL-positivity and caspase-3 cleavage suggesting that nitrofen induces P19 cell apoptosis. In addition, the increase in TUNEL-positive cells was inhibited with zVAD-fmk, suggesting that nitrofen induces a caspase-dependent apoptosis. Nitrofen treatment was associated with increased p38 MAP kinase activity, though pretreatment of cells with multiple p38 inhibitors did not affect nitrofen-mediated caspase-3 cleavage, suggesting caspase-3 cleavage is p38-independent. Nitrofen induced a dose-dependent increase in reactive oxygen species (ROS), which was accompanied by a decrease in the ratio of reduced/oxidized glutathione, indicating that nitrofen alters the cellular redox state of these cells. Furthermore, pretreatment of cells with N-acetyl cysteine gave a dose- and time-dependent reduction of caspase-3 cleavage, supporting the observations that caspase-3 cleavage is cell-redox-dependent. Therefore, nitrofen induces P19 cell apoptosis that is cell-redox-dependent and is associated with increases in p38 activity and ROS and may play a role in nitrofen-mediated birth defects.


Subject(s)
Apoptosis/drug effects , Herbicides/toxicity , Phenyl Ethers/toxicity , Teratocarcinoma/pathology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Line, Tumor/drug effects , Cell Line, Tumor/enzymology , Cell Line, Tumor/pathology , Cell Survival/drug effects , Dose-Response Relationship, Drug , In Situ Nick-End Labeling , Mice , Oxidation-Reduction , Teratocarcinoma/drug therapy , Teratocarcinoma/enzymology
6.
Biochem Biophys Res Commun ; 306(3): 650-9, 2003 Jul 04.
Article in English | MEDLINE | ID: mdl-12810068

ABSTRACT

Telomerase is active in about 90% of cancers and contributes to the immortality of cancer cells by maintaining the lengths of the ends of chromosomes. Undifferentiated embryonic human teratocarcinoma (HT) cells were found to express high levels of hTERT, the catalytic subunit of telomerase, and the hTERT promoter was unmethylated in these cells. Retinoic acid (RA)-induced differentiation led to hTERT gene silencing and increased methylation of the hTERT promoter. Treatment with trichostatin A, a histone deacetylase inhibitor, resulted in hTERT reactivation only in very early differentiating HT cells. After methylation patterns had been established within the hTERT promoter region in late differentiating cells, 5-azacytidine, a common demethylating agent, activated the hTERT gene but trichostatin A had no effect on hTERT transcription. These studies suggest that histone deacetylation is involved in early hTERT gene down-regulation and that DNA methylation may maintain silencing of the hTERT gene in these cells.


Subject(s)
Gene Expression Regulation, Enzymologic , Telomerase/metabolism , Teratocarcinoma/enzymology , Antineoplastic Agents/metabolism , Azacitidine/metabolism , Catalytic Domain , Cell Differentiation , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA-Binding Proteins , Enzyme Inhibitors/metabolism , Humans , Hydroxamic Acids/metabolism , Methylation , Promoter Regions, Genetic , Telomerase/genetics , Teratocarcinoma/genetics , Teratocarcinoma/pathology , Tretinoin/metabolism , Tumor Cells, Cultured
7.
Scand J Immunol ; 56(3): 303-9, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12193232

ABSTRACT

Redistribution, post-translational modifications and coclustering with viral antigens contribute to the immunogenicity of apoptotic cell-derived autoantigens. Almost all known targets of the humoral autoimmune response in systemic lupus erythematosus (SLE) are cleaved by caspases or granzyme B during apoptosis. Antibodies against retroviral proteins can frequently be detected in the sera of SLE patients without overt retroviral infections. These antibodies may represent cross-reactive antibodies or may have been induced by proteins encoded by endogenous retroviral sequences. We used Tera-1 cells that abundantly express a group-specific antigen of human endogenous retroviruses, HERV-K10gag polyprotein, to investigate its processing during apoptosis. Tera-1 cells induced to undergo apoptosis showed an altered HERV-K10gag processing compared with viable cells. In addition, granzyme B was able to cleave HERV-K10gag isolated from viable Tera-1 cells. Similar to nuclear autoantigens, endogenous retroviral proteins are cleaved during the execution phase of apoptosis. These post-translational modifications may result in the generation of T-cell neoepitopes or a changed epitope hierarchy of retroviral proteins. Therefore, immunogenicity of retroviral antigens in SLE patients may result from a similar mechanism as described for nuclear autoantigens.


Subject(s)
Apoptosis , Caspases/metabolism , Endogenous Retroviruses , Gene Products, gag/metabolism , Lupus Erythematosus, Systemic/immunology , Serine Endopeptidases/metabolism , Amino Acid Sequence , Antibodies, Viral/blood , Caspase Inhibitors , Cell Extracts/analysis , Cysteine Proteinase Inhibitors/pharmacology , Gene Products, gag/chemistry , Granzymes , Humans , Immunoblotting , Teratocarcinoma/enzymology , Teratocarcinoma/metabolism , Teratocarcinoma/pathology , Tumor Cells, Cultured , Viral Proteins
8.
J Neurochem ; 76(4): 957-65, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11181815

ABSTRACT

Patients with Down's syndrome (DS) show elevated levels of copper, zinc-containing superoxide dismutase (SOD1) and appear to have increased lipid peroxidation and oxidative damage to DNA as well as elevated glutathione peroxidase activity. Increasing SOD1 levels by gene transfection in NT-2 and SK-N-MC cell lines also led to a rise in glutathione peroxidase activity, but this was nevertheless accompanied by decreased proliferation rates, increased lipid peroxidation and protein carbonyls, and a trend to a rise in 8-hydroxyguanine and protein-bound 3-nitrotyrosine. Transfection of these cell lines with DNA encoding two mutant SOD1 enzymes (G37R and G85R) associated with familial amyotrophic lateral sclerosis (FALS), produced similar, but more severe changes, i.e. even lower growth rates, higher lipid peroxidation, 3-nitrotyrosine and protein carbonyl levels, decreased GSH levels, raised GSSG levels and higher glutathione peroxidase activities. Since G85R has little SOD activity, these changes cannot be related to increased O(2)(-) scavenging. In no case was SOD2 (mitochondrial Mn-SOD) level altered. Our cellular systems reproduce many of the biochemical changes observed in patients with DS or ALS, and in transgenic mice overexpressing mutant SOD1. They also show the potentially deleterious effects of SOD1 overexpression on cellular proliferation, which may be relevant to abnormal development in DS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Antioxidants/metabolism , Down Syndrome/genetics , Guanine/analogs & derivatives , Oxidative Stress/genetics , Superoxide Dismutase/biosynthesis , Tyrosine/analogs & derivatives , Aldehydes/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Cell Division/genetics , Cell Line , Cell Survival/genetics , Down Syndrome/metabolism , Gene Expression , Glutathione/metabolism , Glutathione Disulfide/metabolism , Guanine/metabolism , Humans , Ketones/metabolism , Lipid Peroxidation/genetics , Mutation , Neuroblastoma/enzymology , Neuroblastoma/genetics , Oxidative Stress/drug effects , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Teratocarcinoma/enzymology , Teratocarcinoma/genetics , Transfection , Tyrosine/metabolism
9.
Int J Cancer ; 85(3): 370-5, 2000 Feb 01.
Article in English | MEDLINE | ID: mdl-10652429

ABSTRACT

Cell cycle regulators govern cellular proliferation, modulate differentiation and, when defective, contribute to oncogenesis. Here, we examined expression of cyclins A, B1 and E, and cyclin-dependent kinase (CDK) inhibitors p18INK4C (p18), p21WAF1/Cip1 (p21) and p27KiP1 (p27), in normal human adult testis (n = 5), and 53 testicular tumours, including 23 carcinomas in situ (CIS) and 30 germ cell tumours (GCTs). Immunohistochemical analysis revealed a correlation between proliferation and abundance of the cyclin proteins, and abundant p18 and the lack of p21 and p27 in normal spermatogenesis. Expression of p21 and/or p27 was induced in some differentiated structures seen in teratomas, and was recapitulated in cell culture, using human NTera2/D1 teratocarcinoma cells induced to differentiate into neurons. CIS lesions showed abundant p18, low cyclin E, and moderate p27, in contrast with most invasive seminomas and embryonal carcinomas with very low-to-negative p18, often elevated cyclin E, and, unexpectedly, sustained or increased p27. Our results suggest increased abundance of cyclin E, and particularly downmodulation or loss of p18INK4C as the features that correlate with progression from CIS to invasive germ cell tumours of the human testis.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/metabolism , Enzyme Inhibitors/metabolism , Germinoma/enzymology , Germinoma/pathology , Microtubule-Associated Proteins/metabolism , Teratocarcinoma/enzymology , Teratocarcinoma/pathology , Testicular Neoplasms/enzymology , Testicular Neoplasms/pathology , Tumor Suppressor Proteins , Adult , Carcinoma in Situ/enzymology , Carcinoma in Situ/pathology , Cyclin-Dependent Kinase Inhibitor p18 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Disease Progression , Electrophoresis, Polyacrylamide Gel , Germinoma/physiopathology , Humans , Immunoblotting , Luminescent Measurements , Male , Neoplasm Invasiveness , Teratocarcinoma/physiopathology , Testicular Neoplasms/physiopathology , Tumor Cells, Cultured
10.
FEBS Lett ; 454(3): 197-200, 1999 Jul 09.
Article in English | MEDLINE | ID: mdl-10431806

ABSTRACT

Phospholipase D has been recognized as playing an important role in signal transduction in many types of cells. We investigated the expression of phospholipase D during the differentiation of F9 embryonal teratocarcinoma cells. The ADP ribosylation factor-dependent phospholipase D activity, as measured by an in vitro assay, and H2O2-induced phospholipase D activity and phospholipase D protein content in whole cells were decreased during the differentiation of F9 cells induced by a combination of dibutyryl cyclic AMP and all-trans retinoic acid. In contrast, these changes were not observed when cells were induced by retinoic acid. These results suggest that down-regulation of phospholipase D protein is associated with differentiation of F9 cells to a parietal endoderm lineage.


Subject(s)
Phospholipase D/biosynthesis , Teratocarcinoma/enzymology , Teratocarcinoma/pathology , Animals , Antineoplastic Agents/pharmacology , Cell Differentiation/drug effects , Cyclic AMP/pharmacology , Down-Regulation/drug effects , Mice , Tretinoin/pharmacology , Tumor Cells, Cultured
11.
Mol Reprod Dev ; 49(4): 343-55, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9508085

ABSTRACT

Extracellular glutathione peroxidase (EGPx) is a secreted selenium-dependent enzyme that reduces hydroperoxides and organic hydroperoxides. Selenium deficiency in females is associated with infertility and spontaneous abortion, suggesting a role for selenium-requiring proteins during embryonic development. To gain insight into functions of EGPx in vivo, we determined sites of murine EGPx synthesis by in situ hybridization during embryogenesis and in adult tissues. At E7.5 of development, high EGPx expression was found in the maternally derived deciduum, with lower levels of accumulation in the embryonic visceral endoderm. At E9.5, the major sites of expression were the yolk sac endoderm and heart musculature. By E16.5, EGPx mRNA expression persisted in yolk sac endoderm but also accumulated significantly in atrially derived myocytes, ossification centers, adipose tissue, intestinal epithelium, and in a ventral-to-dorsal gradient in developing skin. Glutathione peroxidase activity due to EGPx protein was identified in the fluids surrounding the developing mouse embryo at midgestation. The expression of EGPx in tissues at the maternal-fetal interface--deciduum, visceral yolk sac, and skin--suggests that EGPx may serve to protect the embryo from oxidant damage. In adult mice, we identified the S1 segment of the kidney proximal tubules as the primary site of EGPx mRNA accumulation, with lower EGPx levels in atrial cardiac muscle, intestine, skin, and adipose tissue. These findings suggest that EGPx may serve a wider antioxidant role than previously recognized in the interstitium of multiple localized tissues, particularly those associated with the active transport of lipids.


Subject(s)
Antioxidants/metabolism , Decidua/enzymology , Extracellular Space/enzymology , Glutathione Peroxidase/biosynthesis , Skin/enzymology , Yolk Sac/enzymology , Amniotic Fluid/enzymology , Animals , Cell Differentiation , Decidua/embryology , Female , Kidney/enzymology , Kidney/growth & development , Mice , Myocardium/enzymology , Organ Specificity/genetics , RNA, Messenger/biosynthesis , Skin/embryology , Teratocarcinoma/enzymology , Teratocarcinoma/pathology , Tumor Cells, Cultured , Yolk Sac/embryology
12.
Cancer Lett ; 134(2): 121-8, 1998 Dec 25.
Article in English | MEDLINE | ID: mdl-10025871

ABSTRACT

Treatment with all-trans-retinoic acid (ATRA) induces complete remission in many acute promyelocytic leukemia patients. However, plasma drug levels progressively decrease following prolonged treatment with oral ATRA. This decrease is due, at least in part, to the induced cytochrome P-450-dependent metabolism of ATRA. To investigate if incorporation of ATRA in liposomes could alter its metabolism, we compared the cellular metabolism of liposomal-ATRA (L-ATRA) with free drug. Microsomes isolated from the rat liver metabolized L-ATRA to a significantly lower extent than they did free-ATRA. Similarly, in F9 cells, L-ATRA was metabolized at a slower rate than the free drug. These results suggest that L-ATRA may have important clinical implications in terms of slowing down the rate of ATRA metabolism and producing long-term remission in APL patients.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Drug Carriers , Liposomes , Microsomes, Liver/enzymology , Teratocarcinoma/enzymology , Tretinoin/metabolism , Animals , Cytochrome P-450 Enzyme Inhibitors , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Male , Rats , Testicular Neoplasms/enzymology , Tumor Cells, Cultured
13.
J Biol Chem ; 272(7): 4359-66, 1997 Feb 14.
Article in English | MEDLINE | ID: mdl-9020157

ABSTRACT

When ornithine decarboxylase, the initial and highly regulated enzyme in polyamine biosynthesis, is irreversibly inactivated by alpha-difluoromethylornithine, F9 teratocarcinoma stem cells are depleted of putrescine and spermidine and as a result differentiate into a cell type which phenotypically resembles the parietal endoderm cells of the early mouse embryo. Simultaneously the level of decarboxylated S-adenosylmethionine (dcAdoMet), the aminopropyl group donor in spermidine and spermine synthesis, increases dramatically, as the aminopropyl group acceptor molecules (putrescine and spermidine) become limiting. When this excessive accumulation of dcAdoMet is prevented by specific inhibition of the AdoMet decarboxylase activity, the differentiative effect is counteracted, despite the fact that the extent of polyamine depletion remains almost identical. Therefore, it may be concluded that dcAdoMet plays an important role in the induction of differentiation. Moreover, this key metabolite acts as a competitive inhibitor of DNA methyltransferase and is therefore capable of interfering with the maintenance methylation of newly replicated DNA. During the course of F9 cell differentiation, the highly methylated genome is gradually demethylated, and its pattern of gene expression is changed. Our present findings, that the DNA remains highly methylated and that the differentiative process is counteracted when the build-up of dcAdoMet is prevented, provide strong evidence for a causative relation between the level of dcAdoMet and the state of DNA methylation as well as cell differentiation.


Subject(s)
Cell Differentiation , DNA Methylation , DNA Modification Methylases/antagonists & inhibitors , Polyamines/metabolism , Teratocarcinoma/metabolism , Adenosylmethionine Decarboxylase/antagonists & inhibitors , Animals , Cytosine/metabolism , DNA Modification Methylases/genetics , Eflornithine/pharmacology , Enzyme Inhibitors/pharmacology , Mice , Ornithine Decarboxylase Inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , S-Adenosylmethionine/analogs & derivatives , S-Adenosylmethionine/pharmacology , Teratocarcinoma/enzymology , Teratocarcinoma/genetics , Tumor Cells, Cultured
14.
Exp Cell Res ; 219(2): 494-8, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7641802

ABSTRACT

gamma-Glutamyl transpeptidase (gamma GT) is a crucial enzyme for the metabolism of xenobiotics and endogenous mediators of biological functions (leukotrienes, prostaglandins, and hepoxillins). Yet little is known about its potential role during development. It is a single copy gene expressed from at least seven promoters. Using histochemistry and immunohistochemistry we demonstrate that gamma GT first appears in the midgestational yolk sacs of mouse embryos. Established cell lines with phenotypic features of yolk sac endoderm (JC-44) or embryonic stem cells were also assayed for the expression of gamma GT. Significant levels were detected in JC-44 cells and higher levels were found in JC-44-derived embryoid bodies. Because this cell line appears to be a good in vitro counterpart of yolk sac differentiation, we characterized the gamma GT mRNA types expressed in JC-44 cells. By ribonuclease protection analysis, gamma GT RNA types IV and VI represent about 80% of the total gamma GT RNA in JC-44 embryoid bodies. Reverse transcription-mediated polymerase chain reaction detected low amounts of gamma GT RNA types I, III, and V. Expression of gamma GT in yolk sac follows a pattern seen in many tissues in which one or two gamma GT RNA types dominate the expression profile; however, the reason for this tissue specificity is unknown.


Subject(s)
Endodermal Sinus Tumor/enzymology , Ovarian Neoplasms/enzymology , Teratocarcinoma/enzymology , Yolk Sac/enzymology , gamma-Glutamyltransferase/biosynthesis , Animals , Cell Line, Transformed , Embryonic and Fetal Development , Female , Immunochemistry , Mice , Pregnancy
15.
Cell Mol Biol Res ; 40(5-6): 587-92, 1994.
Article in English | MEDLINE | ID: mdl-7735333

ABSTRACT

The effect of cis-diaminedichloroplatinum(II) (cisplatin) on the induction of p53 and protein kinase CK2 activity was studied in the mouse teratocarcinoma cell line F9. Treatment of the cells with the chemotherapeutic agent cisplatin led to the detection of p53 3 h after addition of the drug. F9 cell extracts treated with and without cisplatin were analyzed by ion exchange chromatography for protein kinase CK2 alpha/beta subunits and p53 distribution. The following results were obtained: (a) in crude extracts of cisplatin-treated cells, CK2 activity was sometimes reduced by as much as 50%; (b) after separation by anionic exchange chromatography (MA7Q, BioRad) of the crude cellular extracts from cisplatin-treated cells, lower CK2 activity was found in the peak fractions confirming the results obtained with crude cellular extracts; (c) besides the detection of CK2 alpha subunit by immunostaining, we have detected, at a concentration of approximately 200 mM NaCl, a protein of approximately 46 kDa which reacted with the CK2 alpha-specific antibody. This fraction was devoid of CK2 activity; and (d) cisplatin-treated cells exhibited p53 protein, which was mostly eluting ahead but also partly together with CK2 holoenzyme.


Subject(s)
Cisplatin/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Neoplasm Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Teratocarcinoma/enzymology , Tumor Suppressor Protein p53/metabolism , Amino Acid Sequence , Animals , Casein Kinase II , Chromatography, Ion Exchange , Mice , Molecular Sequence Data , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Phosphorylation/drug effects , Protein Conformation , Protein Processing, Post-Translational/drug effects , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Recombinant Fusion Proteins/immunology , Teratocarcinoma/pathology , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
16.
Int J Cancer ; 56(2): 219-23, 1994 Jan 15.
Article in English | MEDLINE | ID: mdl-8314305

ABSTRACT

Secretion of proteolytic enzymes by cells has been implicated in tissue remodeling during embryonic development as well as in invasive neoplastic diseases. We studied the regulation of type-IV-collagenase activity in Tera 2 human embryonal carcinoma cells, which in the undifferentiated state proliferate rapidly and are tumorigenic. The undifferentiated cells produced relatively low levels of matrix-metalloproteinase-2 (MMP-2) activity. This activity was not markedly affected by exogenous basic fibroblast growth factor (bFGF) or 12-O-tetradecanoyl-phorbol-13-acetate (TPA), even though the plasminogen activator activity of the cells was increased by these agents. Tera 2 cells can be induced by retinoic acid to differentiate into quiescent cells, of which many express neuronal characteristics. The type-IV-collagenase activity of the cells increased markedly during the differentiation. This increase was mainly due to increased expression of MMP-2. Expression of tissue inhibitors of metalloproteinases (TIMP-1 and TIMP-2) was not markedly affected by the differentiation of Tera 2 cells. The results show that in the Tera 2 cell system, increased expression of MMP-2 is characteristic of the differentiated derivatives. This is in contrast with many other model systems, where increased type-IV-collagenase activity is associated with the malignant phenotype. This pattern of regulation may reflect the facts that Tera 2 cells resemble early embryonic cells and that their differentiation mimics related cell-differentiation processes in the developing embryo.


Subject(s)
Carcinoma, Embryonal/embryology , Gelatinases/metabolism , Metalloendopeptidases/metabolism , Teratocarcinoma/enzymology , Testicular Neoplasms/enzymology , Carcinoma, Embryonal/metabolism , Carcinoma, Embryonal/pathology , Cell Differentiation/physiology , Collagen/metabolism , Collagenases/metabolism , Fibroblast Growth Factor 2/pharmacology , Gelatinases/antagonists & inhibitors , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Male , Matrix Metalloproteinase 2 , Matrix Metalloproteinase Inhibitors , Metalloendopeptidases/antagonists & inhibitors , Plasminogen Activators/metabolism , RNA, Messenger/genetics , Stimulation, Chemical , Teratocarcinoma/metabolism , Teratocarcinoma/pathology , Testicular Neoplasms/metabolism , Testicular Neoplasms/pathology , Tetradecanoylphorbol Acetate/pharmacology , Tissue Inhibitor of Metalloproteinases , Tumor Cells, Cultured/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...