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










Publication year range
1.
Stem Cells Dev ; 19(7): 951-60, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20064013

ABSTRACT

We have introduced 1 to 2 copies of a deletion mutant (betaDeltaC) of the human retinoic acid receptor beta into mouse embryonic stem (ES) cells. The betaDeltaC-expressing cells were 10 to 100 times less sensitive to RA-induced differentiation in comparison with their parental cells. In the presence of 10(-7) M RA in monolayer culture, they showed no growth arrest or differentiation, but remained pluripotent. Embryoid bodies (EBs) derived from betaDeltaC-expressing cells differentiated into cardiomyocytes rather than neurons after treatment with 10(-6) M RA, and became neurons upon exposure to 10(-5) or 10(-4) M RA. Remarkably, after 10 passages of continuous culture in the presence of 10(-7) M RA, they still were able to form chimeras after injection into blastocysts. These data suggest that appropriate levels of normal retinoid receptors are crucial for lineage-specific differentiation of mouse ES cells in vitro. The betaDeltaC mutant protein may prove to be useful in promoting "stemness" of ES cells in culture.


Subject(s)
Cell Differentiation/physiology , Cell Lineage , Embryo, Mammalian , Embryonic Stem Cells/physiology , Receptors, Retinoic Acid , Sequence Deletion , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line , Cell Proliferation/drug effects , Embryo, Mammalian/cytology , Embryo, Mammalian/physiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Humans , Mice , Neurons/cytology , Neurons/physiology , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Tretinoin/metabolism , Tretinoin/pharmacology
2.
J Endocrinol ; 203(3): 349-56, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19752149

ABSTRACT

Raloxifene is a selective oestrogen receptor modulator with tissue-specific effects. The mechanisms behind the effects of raloxifene are partly unclear, and the aim of the present study was to investigate whether raloxifene can activate the classical oestrogen-signalling pathway in vivo in three known oestrogen-responsive organs, uterus (reproductive organ), bone (non-reproductive organ) and thymus (immune organ). For this purpose, we have used reporter mice with a luciferase gene under control of oestrogen-responsive elements (EREs), enabling detection of in vivo activation of gene transcription via the classical oestrogen pathway. Three-month-old ovariectomized ERE-luciferase mice were treated with the raloxifene analogue (LY117018), oestradiol (OE(2)) or vehicle for 3 weeks. Luciferase activation was measured in bone, uterus and thymus, and compared to bone parameters, and uterus and thymus weights. The raloxifene analogue affected bone mineral density (BMD) to the same extent as OE(2), and both treatments resulted in increased luciferase activity in bone. As expected, OE(2) treatment resulted in increased uterus weight and increased uterine luciferase activity, while the effect of LY117018 on uterus weight and luciferase activity was modest and significantly lower than the effect of OE(2). LY117018 and OE(2) treatment resulted in similar luciferase activation in thymus. However, only OE(2) treatment resulted in thymic atrophy, while no effect on thymus weight was seen after LY117018 treatment. In summary, the raloxifene analogue LY117018 can activate the classical oestrogen pathway in bone, uterus and thymus in vivo, and this activation is associated with BMD and uterus weight, but not thymus weight.


Subject(s)
Estrogen Antagonists/pharmacology , Femur/drug effects , Pyrrolidines/pharmacology , Thiophenes/pharmacology , Thymus Gland/drug effects , Transcription, Genetic/drug effects , Uterus/drug effects , Animals , Bone Density/drug effects , Estradiol/pharmacology , Estrogens/metabolism , Estrogens/pharmacology , Female , Femur/metabolism , Luciferases/genetics , Mice , Mice, Inbred CBA , Mice, Transgenic , Organ Size/drug effects , Raloxifene Hydrochloride/analogs & derivatives , Raloxifene Hydrochloride/pharmacology , Receptors, Estrogen/metabolism , Response Elements , Thymus Gland/metabolism , Uterus/growth & development , Uterus/metabolism
3.
Mol Nutr Food Res ; 53(8): 996-1006, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19603405

ABSTRACT

Dietary phytoestrogens, such as the lignan metabolite enterolactone (ENL) and the isoflavone genistein (GEN), are suggested to modulate the risk of estrogen-dependent disease (e.g., breast cancer) through regulation of estrogen signaling. However, the effects of complex food items containing lignans or isoflavones on estrogen receptor (ER) transactivation have not been assessed so far. In this study, the modulation of ER-mediated signaling by dietary sources of lignans (cereals and flaxseed) and isoflavones (soy) was studied in vivo. Adult ovariectomized 3 x ERE-luciferase (luc) reporter mice received isocaloric diets supplemented with flaxseed, rye, wheat, or soy for 40 h or two weeks, and an additional group of mice was challenged with 17beta-estradiol (E(2)) following the two-week dietary intervention. In non-E(2)-treated mice, soy diet induced luc expression in liver, mammary gland, and pituitary gland while the other diets had no effects. Interestingly, all diets modulated the E(2)-induced luc expression. In particular rye diet efficiently reduced E(2)-induced luc expression as well as uterine growth, the hallmark of estrogen action in vivo. It is concluded that dietary sources of lignans and isoflavones can modulate estrogen signaling in vivo. The results suggest intriguing possibilities for the modulation of the risk of estrogen-dependent diseases by dietary means.


Subject(s)
Diet , Estradiol/pharmacology , Isoflavones/pharmacology , Lignans/pharmacology , Animals , Edible Grain , Female , Flax , Luciferases/genetics , Mice , Receptors, Estrogen/physiology , Weight Gain
4.
Proc Natl Acad Sci U S A ; 105(46): 17712-7, 2008 Nov 18.
Article in English | MEDLINE | ID: mdl-19004801

ABSTRACT

Refsum disease is caused by a deficiency of phytanoyl-CoA hydroxylase (PHYH), the first enzyme of the peroxisomal alpha-oxidation system, resulting in the accumulation of the branched-chain fatty acid phytanic acid. The main clinical symptoms are polyneuropathy, cerebellar ataxia, and retinitis pigmentosa. To study the pathogenesis of Refsum disease, we generated and characterized a Phyh knockout mouse. We studied the pathological effects of phytanic acid accumulation in Phyh(-/-) mice fed a diet supplemented with phytol, the precursor of phytanic acid. Phytanic acid accumulation caused a reduction in body weight, hepatic steatosis, and testicular atrophy with loss of spermatogonia. Phenotype assessment using the SHIRPA protocol and subsequent automated gait analysis using the CatWalk system revealed unsteady gait with strongly reduced paw print area for both fore- and hindpaws and reduced base of support for the hindpaws. Histochemical analyses in the CNS showed astrocytosis and up-regulation of calcium-binding proteins. In addition, a loss of Purkinje cells in the cerebellum was observed. No demyelination was present in the CNS. Motor nerve conduction velocity measurements revealed a peripheral neuropathy. Our results show that, in the mouse, high phytanic acid levels cause a peripheral neuropathy and ataxia with loss of Purkinje cells. These findings provide important insights in the pathophysiology of Refsum disease.


Subject(s)
Ataxia/pathology , Purkinje Cells/pathology , Refsum Disease/pathology , Animals , Ataxia/enzymology , Ataxia/physiopathology , Automation , Behavior, Animal/drug effects , Central Nervous System/abnormalities , Central Nervous System/drug effects , Central Nervous System/enzymology , Central Nervous System/pathology , Dietary Supplements , Disease Models, Animal , Gait/drug effects , Gene Targeting , Genetic Vectors , Lipidoses/enzymology , Lipidoses/pathology , Male , Mice , Mixed Function Oxygenases/deficiency , Mixed Function Oxygenases/genetics , Peripheral Nervous System Diseases/enzymology , Peripheral Nervous System Diseases/pathology , Phenotype , Phytanic Acid/blood , Phytol/administration & dosage , Phytol/pharmacology , Purkinje Cells/drug effects , Purkinje Cells/enzymology , Refsum Disease/enzymology , Refsum Disease/physiopathology , Spermatogonia/drug effects , Spermatogonia/enzymology , Spermatogonia/pathology
5.
Chem Biol Interact ; 174(2): 126-33, 2008 Jul 30.
Article in English | MEDLINE | ID: mdl-18501883

ABSTRACT

The present paper aims at clarifying to what extent seven food-associated compounds, shown before to be estrogenic in vitro, can induce estrogenic effects in male mice with an estrogen receptor (ER)-mediated luciferase (luc) reporter gene system. The luc induction was determined in different tissues 8h after dosing the ER-luc male mice intraperitoneally (IP) or 14h after oral dosing. Estradiol-propionate (EP) was used as a positive control at 0.3 and 1mg/kg bodyweight (bw), DMSO as solvent control. The food-associated estrogenic compounds tested at non-toxic doses were bisphenol A (BPA) and nonylphenol (NP) (both at 10 and 50mg/kgbw), dichlorodiphenyldichloroethylene (p,p'-DDE; at 5 and 25mg/kgbw), quercetin (at 1.66 and 16.6mg/kgbw), di-isoheptyl phthalate (DIHP), di-(2-ethylhexyl) phthalate (DEHP) and di-(2-ethylhexyl) adipate (DEHA) all at 30 and 100mg/kgbw. In general IP dosing resulted in higher luc inductions than oral dosing. EP induced luc activity in the liver in a statistically significant dose-related way with the highest induction of all compounds tested which was 20,000 times higher than the induction by the DMSO-control. NP, DDE, DEHA and DIHP did not induce luc activity in any of the tissues tested. BPA induced luc in the liver up to 420 times via both exposure routes. BPA, DEHP and quercetin induced luc activity in the liver after oral exposure. BPA (50mg/kgbw IP) also induced luc activity in the testis, kidneys and tibia. The current study reveals that biomarker-responses in ER-luc male mice occur after a single oral exposure to food-associated estrogenic model compounds at exposure levels 10 to 10(4) times higher than the established TDI's for some of these compounds. Given the facts that (i) the present study did not include chronic exposure and that (ii) simultaneous exposure to multiple estrogenic compounds may be a realistic exposure scenario, it remains to be seen whether this margin is sufficiently high.


Subject(s)
Endocrine Disruptors/toxicity , Estrogens, Non-Steroidal/toxicity , Food Additives/toxicity , Gene Expression Regulation, Enzymologic/drug effects , Liver/drug effects , Luciferases/genetics , Receptors, Estrogen/metabolism , Administration, Oral , Animals , Benzhydryl Compounds , Diethylhexyl Phthalate/toxicity , Dose-Response Relationship, Drug , Food Contamination , Genes, Reporter/drug effects , Injections, Intraperitoneal , Liver/enzymology , Male , Mice , Mice, Transgenic , Phenols/toxicity , Quercetin/toxicity
6.
Mol Cell Endocrinol ; 273(1-2): 42-50, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17590503

ABSTRACT

Inhibition of NF-kappaB transcriptional activity by steroid receptors is the basis for the antiinflammatory actions of steroid hormones and the molecular mechanism underlying this cross-talk is thought to involve direct protein-protein interactions. In this study, we show that estrogen receptor (ER)alpha and NF-kappaB interact in vivo by using fluorescence resonance energy transfer (FRET) and co-immunoprecipitation. U2-OS cells were used to study direct interactions between fluorescent fusion proteins of ERalpha and the NF-kappaB subunits p50 and p65. Interactions were observed only in the nucleus and maximal FRET signal was detected when ERalpha is co-expressed with both NF-kappaB subunits and cells were stimulated with estrogen. This is in agreement with the induction of nuclear co-localization of the proteins under this condition. Moreover, in a U2-OS clone stably expressing ERalpha, interaction with NF-kappaB was confirmed. A p65 deletion mutant lacking the Rel homology domain was strongly impaired in its interaction with ERalpha showing the importance of this domain. Taken together, these findings provide a strong basis for the direct protein-protein interaction model for cross-talk between ERalpha and NF-kappaB.


Subject(s)
Cell Nucleus/metabolism , Estrogen Receptor alpha/metabolism , NF-kappa B p50 Subunit/metabolism , Transcription Factor RelA/metabolism , Cell Line, Tumor , Cell Survival , Clone Cells , Fluorescence Resonance Energy Transfer , Humans , Immunoprecipitation , Luminescent Proteins/metabolism , Protein Binding , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Transcription Factor RelA/chemistry , Transcription Factor RelA/genetics , Transcription, Genetic
7.
J Agric Food Chem ; 54(12): 4407-16, 2006 Jun 14.
Article in English | MEDLINE | ID: mdl-16756374

ABSTRACT

This study presents the estrogenic potency of 21 food-packaging-associated compounds determined for the first time, using two transfected U2-OS (human osteoblasts devoid of endogenous estrogen receptors) estrogen receptor (ER) alpha and beta cell lines. Six plasticizers and three antioxidants were slightly estrogenic in the ERalpha cells. The model compounds bisphenol A and nonylphenol, one plasticizer [tris(2-ethylhexyl)trimellitate (TEHTM)], and two antioxidants (propyl gallate and butylated hydroxyanisole) were estrogenic in both ERalpha and ERbeta cells. Compared to estradiol (E2), these compounds appeared to be relatively more estrogenic in the ERbeta cells than in the ERalpha cells. Three sorbitol-based plasticizers activated neither ERalpha nor ERbeta and may be good replacements of existing plasticizers. All responses were additive with the response of E2. This indicates that they may contribute to the total effects of the pool of estrogenic compounds humans are exposed to. The estrogenic potencies of these compounds, together with the suggested beneficial effect of ERbeta-mediated responses and adverse ERalpha-mediated effects, support the importance of detecting characteristics for ERalpha and ERbeta response separately in independent models, as done in the present study.


Subject(s)
Antioxidants/pharmacology , Estrogen Receptor alpha/drug effects , Estrogen Receptor beta/drug effects , Estrogens/pharmacology , Food Packaging , Plasticizers/pharmacology , Cell Line , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Genes, Reporter/genetics , Humans , Osteoblasts , Transfection
8.
J Biol Chem ; 281(29): 20055-67, 2006 Jul 21.
Article in English | MEDLINE | ID: mdl-16707494

ABSTRACT

The purpose of physiological cell death is the noninflammatory clearance of cells that have become inappropriate or nonfunctional. Consistent with this function, the recognition of apoptotic cells by professional phagocytes, including macrophages and dendritic cells, triggers a set of potent anti-inflammatory responses manifest on multiple levels. The immediate-early inhibition of proinflammatory cytokine gene transcription in the phagocyte is a proximate consequence of recognition of the apoptotic corpse, independent of subsequent engulfment and soluble factor involvement. Here, we show that recognition is linked to a characteristic signature of responses, including MAPK signaling events and the ablation of proinflammatory transcription and cytokine secretion. Specific recognition and response occurs without regard to the origin (species, tissue type, or suicidal stimulus) of the apoptotic cell and does not involve Toll-like receptor signaling. These features mark this as an innate immunity fundamentally distinct from the discrimination of "self" versus "other" considered to be the hallmark of conventional immunity. This profound unconventional innate immune discrimination of effete from live cells is as ubiquitous as apoptotic cell death itself, manifest by professional and nonprofessional phagocytes and nonphagocytic cell types alike. Innate apoptotic immunity provides an intrinsic anti-inflammatory circuit that attenuates proinflammatory responses dynamically and may act systemically as a powerful physiological regulator of immunity.


Subject(s)
Apoptosis/physiology , Immunity, Innate , Macrophages/immunology , Animals , Cell Death , Cell Line , Dendritic Cells/immunology , HeLa Cells , Humans , Jurkat Cells , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Phagocytes/immunology , Transcription, Genetic
9.
Mol Nutr Food Res ; 49(8): 763-71, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15937998

ABSTRACT

Quercetin causes biphasic modulation of the proliferation of specific colon and mammary cancer cells. In this study, the possible involvement of the estrogen receptor (ER) in the stimulation of cell proliferation by quercetin was investigated. For this purpose, the effect of quercetin on cell proliferation was tested in ER-positive MCF-7 and T47D cells, and in ER-negative HCC-38 and MDA-MB231 cells. Quercetin stimulated proliferation of ER-positive cells only, suggesting this effect to be ER-dependent. In support of these results, quercetin induced ER-ERE-mediated gene expression in a reporter gene assay using U2-OS cells transfected with either ERalpha or ERbeta, with 10(5)-10(6) times lower affinity than 17beta-estradiol (E2) and 10(2)-10(3 )times lower affinity than genistein. Quercetin activated the ERbeta to a 4.5-fold higher level than E2, whereas the maximum induction level of ERalpha by quercetin was only 1.7 fold that of E2. These results point at the relatively high capacity of quercetin to stimulate supposed 'beneficial' ERbeta responses as compared to the stimulation of ERalpha, the receptor possibly involved in adverse cell proliferative effects. Altogether, the results of this study reveal that physiologically relevant concentrations of quercetin can exert phytoestrogen-like activity similar to that observed for the isoflavonoid genistein.


Subject(s)
Cell Division/drug effects , Quercetin/pharmacology , Receptors, Estrogen/physiology , Breast Neoplasms , Cell Line, Tumor , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/physiology , Estrogen Receptor beta/genetics , Estrogen Receptor beta/physiology , Gene Expression/drug effects , Genistein/pharmacology , Humans , Luciferases/metabolism , Receptors, Estrogen/analysis , Transfection
10.
Toxicol Lett ; 156(2): 261-75, 2005 Apr 10.
Article in English | MEDLINE | ID: mdl-15737489

ABSTRACT

Molecules derived from tetralin, indane and isochroman are often used in the synthesis of fragrance materials. The two polycyclic musk fragrances AHTN (6-acetyl-1,1,2,4,4,7-hexamethyltetralin), HHCB (1,2,4,6,7,8-hexahydro-4,6,6,7,8,8-hexamethylcyclopenta-gamma-2-benzopyran) and ADBI (4-acetyl-1,1-dimethyl-6-tert-butylindane) are derived from tetralin, isochroman and indane, respectively. In previous studies, AHTN and HHCB have been shown to antagonize estrogen receptors (ERs), both in vitro and in vivo. Here, we used two newly developed reporter gene assays, to examine the agonistic and antagonistic properties of several indane, tetralin and isochroman derivatives towards the human androgen receptor (AR) and aryl hydrocarbon receptor (AhR). Additionally, we also assessed (anti)estrogenicity of these compounds. A number of compounds showed weak estrogenic activity towards the human ER alpha. Several compounds showed (anti)estrogenic effects, starting at a concentration of 0.1 microM. Surprisingly, almost all compounds were found to be AR antagonists, starting at 0.1 microM. None of the compounds tested, showed either agonism or antagonism towards the AhR. Non-specific effects via crosstalk of the AhR and the ER or AR can therefore be ruled out. As far as we are aware, molecules derived from indane, tetralin and isochroman showing direct interaction with the ER and AR have not been reported previously.


Subject(s)
Androgens , Chromans/pharmacology , Indans/pharmacology , Receptors, Aryl Hydrocarbon/agonists , Receptors, Estrogen/agonists , Tetrahydronaphthalenes/pharmacology , Androgen Receptor Antagonists , Animals , Cell Line , Dose-Response Relationship, Drug , Genes, Reporter , Humans , Perfume , Rats , Receptors, Androgen/genetics , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/genetics , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/genetics
11.
Environ Sci Technol ; 38(23): 6263-70, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15597880

ABSTRACT

In vitro assays and computer models are promising alternatives for in vivo animal testing, but the power of these alternative methods to predict in vivo risk is still very limited. One step forward is to make the outcome of in vitro assays (such as median effect concentrations (EC50 values)) independent of assay conditions such as protein content. Here we show that measured free concentrations of chemicals in the in vitro assay medium result in system-independent EC50 values. We introduce a very simple method to measure free concentrations in miniature test systems using negligible depletion solid-phase microextraction. The generated data are much more suitable for extrapolation to in vivo, provide unbiased input for computational methods (for example, quantitative structure-activity relationships), and can shed an entirely different light on the activity of environmental contaminants.


Subject(s)
Environmental Monitoring/methods , Environmental Pollutants/analysis , Models, Biological , Animals , Cattle , Cell Survival , Dose-Response Relationship, Drug , Drug-Related Side Effects and Adverse Reactions , Environmental Monitoring/instrumentation , Environmental Pollutants/toxicity , Estradiol/blood , Forecasting , Phenols/blood , Proteins/analysis , Risk Assessment , Toxicity Tests
12.
Environ Health Perspect ; 112(15): 1544-9, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15531440

ABSTRACT

Environmental estrogens are of particular concern when exposure occurs during embryonic development. Although there are good models to study estrogenic activity of chemicals in adult animals, developmental exposure is much more difficult to test. The weak estrogenic activity of the environmental estrogen bisphenol A (BPA) in embryos is controversial. We have recently generated transgenic mice that carry a reporter construct with estrogen-responsive elements coupled to luciferase. We show that, using this in vivo model in combination with the IVIS imaging system, activation of estrogen receptors (ERs) by maternally applied BPA and other estrogens can be detected in living embryos in utero. Eight hours after exposure to 1 mg/kg BPA, ER transactivation could be significantly induced in the embryos. This was more potent than would be estimated from in vitro assays, although its intrinsic activity is still lower than that of diethylstilbestrol and 17beta-estradiol dipropionate. On the basis of these results, we conclude that the estrogenic potency of BPA estimated using in vitro assays might underestimate its estrogenic potential in embryos.


Subject(s)
Estrogens, Non-Steroidal/toxicity , Gene Expression Regulation, Developmental , Maternal-Fetal Exchange , Mice, Transgenic/embryology , Phenols/toxicity , Receptors, Estrogen/drug effects , Receptors, Estrogen/physiology , Animals , Benzhydryl Compounds , Environmental Pollutants/toxicity , Estrogens/toxicity , Female , Genes, Reporter , Mice , Pregnancy , Reproducibility of Results
13.
Mol Endocrinol ; 18(12): 2908-23, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15345745

ABSTRACT

CRH-binding protein (CRH-BP) regulates activation of the hypothalamic-pituitary-adrenal (HPA) axis by binding and inhibiting CRH. We investigated for the first time transcriptional regulation of the human CRH-BP promoter using transient transfections. Estrogen receptors (ERs) contributed to ligand-independent constitutive activation of the promoter, whereas in the presence of estradiol ERalpha induced and ERbeta repressed promoter activity in a dose-dependent manner. TNFalpha inhibited promoter induction by ERalpha in the absence and presence of estradiol. Three ERE half-sites in the CRH-BP promoter bound ERalpha and ERbeta in an EMSA, and disruption of ERE half-sites by site-directed mutagenesis abolished ligand-independent induction by ERalpha and ERbeta and promoter enhancement by estradiol-activated ERalpha. Repression by estradiol/ERbeta was unaffected by disruption of ERE half-sites, activating protein 1, cAMP response element, GATA, or nuclear factor kappaB sites, and reversed to promoter induction by estrogen antagonists, tamoxifen and ICI 182,780, suggesting corepressor involvement. In hypothalamic GT1-7 cells, Western blotting demonstrated rapid induction of endogenous CRH-BP expression by estradiol-bound ER, which was inhibited by TNFalpha. We propose a model in which ERs maintain basal CRH-BP expression in pituitary and neurosecretory cells, whereas in the presence of ERalpha estrogen enhances CRH-BP transcription, causing down-regulation of the HPA axis, and nuclear factor kappaB-activating cytokines activate the HPA axis by inhibiting ERalpha.


Subject(s)
Carrier Proteins/genetics , Estradiol/physiology , Estrogen Receptor alpha/physiology , Estrogen Receptor beta/physiology , Promoter Regions, Genetic/genetics , Transcriptional Activation , Animals , Conserved Sequence/genetics , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Humans , Mutation/genetics , Neurosecretion/genetics , Neurosecretion/physiology , Pituitary Gland/metabolism , Promoter Regions, Genetic/drug effects , Response Elements/drug effects , Response Elements/genetics , Tamoxifen/pharmacology , Transcription, Genetic , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/physiology
14.
Int Immunol ; 16(6): 831-41, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15126418

ABSTRACT

In this study, we investigated the localization and functional significance of p53 tumor suppressor-like molecules, p63 and p73, in human thymic epithelial cells (TECs). Immunohistochemical studies showed particular distribution profiles of p63 and p73 in thymic epithelium, in which cortical TECs preferentially expressed p63 in their nuclei whereas subcapsular and medullary TECs expressed both p63 and p73 in their nuclei. The wide distribution of p63 in TECs was further suggested by studies using TECs of primary culture. In vitro studies using two human TEC lines demonstrated that p63 was capable of up-regulating intercellular adhesion molecule-1 (ICAM-1) and enhancing the production of IL-6 and IL-8. Moreover, in vitro studies also indicated that p73, but not p63, had the capacity to induce granulocyte macrophage colony stimulating factor (GM-CSF) and granulocyte colony stimulating factor (G-CSF) in the TEC lines. These findings suggest that p63 would regulate the cell adhesive property through ICAM-1/LFA-1 interaction and the production of IL-6 and IL-8, probably in all TEC subtypes. p73 in subcapslar and medullary TECs was suggested to play a role in the regulation of the production of GM-CSF and G-CSF, which might stimulate other stromal cells such as dendritic cells, macrophages and endothelial cells around these regions.


Subject(s)
DNA-Binding Proteins/physiology , Nuclear Proteins/physiology , Phosphoproteins/physiology , Thymus Gland/metabolism , Trans-Activators/physiology , Transcription Factors/physiology , Cell Line , Child, Preschool , Cytokines/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epithelial Cells/chemistry , Epithelial Cells/metabolism , Genes, Tumor Suppressor , Humans , Infant , Infant, Newborn , Intercellular Adhesion Molecule-1/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Promoter Regions, Genetic/genetics , RNA, Messenger/analysis , Thymus Gland/chemistry , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection , Tumor Protein p73 , Tumor Suppressor Protein p53/physiology , Tumor Suppressor Proteins , Up-Regulation
15.
Exp Cell Res ; 284(2): 264-73, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12651158

ABSTRACT

Progestins are frequently used in the treatment of advanced breast and endometrial cancer. The human breast carcinoma cell line T47D shows a biphasic response to progestins. Short-term progestin treatment leads to enhanced DNA synthesis, while this line is growth inhibited upon prolonged exposure. An important protein involved in growth regulation by progestins in this cell is the CDK inhibitor p21(Cip1,Waf1). We show that after 1 day of progestin treatment in T47D cells, the p21 promoter-proximal region containing Sp1 binding sites is crucial in the induction by progestins. However, after 3 days the activity of the promoter-distal region becomes predominant in T47D cells or the endometrial carcinoma cell line ECC1. This is dependent upon two domains within this region that contain p53 response elements. In ECC1 and T47D cells 3-day progestin treatment induces a reporter containing a p53 response element, but not a mutated version. This induction is due to activation of p53 by progestin, which may be caused by nuclear translocation of p53. These data indicate that upon prolonged exposure, progestins activate p53, in human breast and endometrial tumor cells, which up-regulates the p21(Cip1,Waf1) promoter. This may be an important mechanism involved in progestin-inhibited cellular proliferation in these cells.


Subject(s)
Breast Neoplasms/genetics , Carcinoma/genetics , Cyclins/genetics , Endometrial Neoplasms/genetics , Progestins/pharmacology , Promoter Regions, Genetic/genetics , Tumor Suppressor Protein p53/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Carcinoma/drug therapy , Carcinoma/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/metabolism , Cyclins/drug effects , Cyclins/metabolism , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Progestins/therapeutic use , Promoter Regions, Genetic/drug effects , Response Elements/drug effects , Response Elements/genetics , Tumor Cells, Cultured , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...