Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters











Publication year range
1.
Clin Transl Oncol ; 24(1): 127-144, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34370182

ABSTRACT

Metaplastic breast carcinoma (MBC) is a rare breast cancer subtype with rapid growth, high rates of metastasis, recurrence and drug resistance, and diverse molecular and histological heterogeneity. Patient-derived xenografts (PDXs) provide a translational tool and physiologically relevant system to evaluate tumor biology of rare subtypes. Here, we provide an in-depth comprehensive characterization of a new PDX model for MBC, TU-BcX-4IC. TU-BcX-4IC is a clinically aggressive tumor exhibiting rapid growth in vivo, spontaneous metastases, and elevated levels of cell-free DNA and circulating tumor cell DNA. Relative chemosensitivity of primary cells derived from TU-BcX-4IC was performed using the National Cancer Institute (NCI) oncology drug set, crystal violet staining, and cytotoxic live/dead immunofluorescence stains in adherent and organoid culture conditions. We employed novel spheroid/organoid incubation methods (Pu·MA system) to demonstrate that TU-BcX-4IC is resistant to paclitaxel. An innovative physiologically relevant system using human adipose tissue was used to evaluate presence of cancer stem cell-like populations ex vivo. Tissue decellularization, cryogenic-scanning electron microscopy imaging and rheometry revealed consistent matrix architecture and stiffness were consistent despite serial transplantation. Matrix-associated gene pathways were essentially unchanged with serial passages, as determined by qPCR and RNA sequencing, suggesting utility of decellularized PDXs for in vitro screens. We determined type V collagen to be present throughout all serial passage of TU-BcX-4IC tumor, suggesting it is required for tumor maintenance and is a potential viable target for MBC. In this study we introduce an innovative and translational model system to study cell-matrix interactions in rare cancer types using higher passage PDX tissue.


Subject(s)
Antineoplastic Agents/therapeutic use , Models, Biological , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Animals , Disease Models, Animal , Heterografts , Humans , Xenograft Model Antitumor Assays
2.
Adipocyte ; 7(2): 96-105, 2018.
Article in English | MEDLINE | ID: mdl-29411671

ABSTRACT

Stromal/stem cell differentiation is controlled by a vast array of regulatory mechanisms. Included within these are methods of mRNA gene regulation that occur at the level of epigenetic, transcriptional, and/or posttranscriptional modifications. Current studies that evaluate the posttranscriptional regulation of mRNA demonstrate microRNAs (miRNAs) as key mediators of stem cell differentiation through the inhibition of mRNA translation. miRNA expression is enhanced during both adipogenic and osteogenic differentiation; however, the mechanism by which miRNA expression is altered during stem cell differentiation is less understood. Here we demonstrate for the first time that adipose-derived stromal/stem cells (ASCs) induced to an adipogenic or osteogenic lineage have differences in strand preference (-3p and -5p) for miRNAs originating from the same primary transcript. Furthermore, evaluation of miRNA expression in ASCs demonstrates alterations in both miRNA strand preference and 5'seed site heterogeneity. Additionally, we show that during stem cell differentiation there are alterations in expression of genes associated with the miRNA biogenesis pathway. Quantitative RT-PCR demonstrated changes in the Argonautes (AGO1-4), Drosha, and Dicer at intervals of ASC adipogenic and osteogenic differentiation compared to untreated ASCs. Specifically, we demonstrated altered expression of the AGOs occurring during both adipogenesis and osteogenesis, with osteogenesis increasing AGO1-4 expression and adipogenesis decreasing AGO1 gene and protein expression. These data demonstrate changes to components of the miRNA biogenesis pathway during stromal/stem cell differentiation. Identifying regulatory mechanisms for miRNA processing during ASC differentiation may lead to novel mechanisms for the manipulation of lineage differentiation of the ASC through the global regulation of miRNA as opposed to singular regulatory mechanisms.


Subject(s)
Adipose Tissue/cytology , Cell Differentiation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , MicroRNAs/biosynthesis , Cells, Cultured , Humans
3.
Oncogenesis ; 4: e168, 2015 Oct 05.
Article in English | MEDLINE | ID: mdl-26436950

ABSTRACT

Liver kinase B1 (LKB1), also known as serine/threonine kinase 11 (STK11), has been identified as a tumor suppressor in many cancers including breast. Low LKB1 expression has been associated with poor prognosis of breast cancer patients, and we report here a significant association between loss of LKB1 expression and reduced patient survival specifically in the basal subtype of breast cancer. Owing to the aggressive nature of the basal subtype as evidenced by high incidences of metastasis, the purpose of this study was to determine if LKB1 expression could regulate the invasive and metastatic properties of this specific breast cancer subtype. Induction of LKB1 expression in basal-like breast cancer (BLBC)/triple-negative breast cancer cell lines, MDA-MB-231 and BT-549, inhibited invasiveness in vitro and lung metastatic burden in an orthotopic xenograft model. Further analysis of BLBC cells overexpressing LKB1 by unbiased whole transcriptomics (RNA-sequencing) revealed striking regulation of metastasis-associated pathways, including cell adhesion, extracellular matrix remodeling, and epithelial-to-mesenchymal transition (EMT). In addition, LKB1 overexpression inhibited EMT-associated genes (CDH2, Vimentin, Twist) and induced the epithelial cell marker CDH1, indicating reversal of the EMT phenotype in the MDA-MB-231 cells. We further demonstrated marked inhibition of matrix metalloproteinase 1 expression and activity via regulation of c-Jun through inhibition of p38 signaling in LKB1-expressing cells. Taken together, these data support future development of LKB1 inducing therapeutics for the suppression of invasion and metastasis of BLBC.

4.
Oncogene ; 30(9): 1082-97, 2011 Mar 03.
Article in English | MEDLINE | ID: mdl-21057537

ABSTRACT

Fulvestrant is a selective estrogen receptor downregulator (SERD) and highly effective antagonist to hormone-sensitive breast cancers following failure of previous tamoxifen or aromatase inhibitor therapies. However, after prolonged fulvestrant therapy, acquired resistance eventually occurs in the majority of breast cancer patients, due to poorly understood mechanisms. To examine a possible role(s) of aberrantly expressed microRNAs (miRNAs) in acquired fulvestrant resistance, we compared antiestrogen-resistant and -sensitive breast cancer cells, revealing the overexpression of miR-221/222 in the SERD-resistant cell lines. Fulvestrant treatment of estradiol (E2)- and fulvestrant-sensitive MCF7 cells resulted in increased expression of endogenous miR-221/222. Ectopic upregulation of miR-221/222 in estrogen receptor-α (ERα)-positive cell lines counteracted the effects of E2 depletion or fulvestrant-induced cell death, thus also conferring hormone-independent growth and fulvestrant resistance. In cells with acquired resistance to fulvestrant, miR-221/222 expression was essential for cell growth and cell cycle progression. To identify possible miR-221/222 targets, miR-221- or miR-222- induced alterations in global gene expression profiles and target gene expression at distinct time points were determined, revealing that miR-221/222 overexpression resulted in deregulation of multiple oncogenic signaling pathways previously associated with drug resistance. Activation of ß-catenin by miR-221/222 contributed to estrogen-independent growth and fulvestrant resistance, whereas TGF-ß-mediated growth inhibition was repressed by the two miRNAs. This first in-depth investigation into the role of miR-221/222 in acquired fulvestrant resistance, a clinically important problem, demonstrates that these two 'oncomirs' may represent promising therapeutic targets for treating hormone-independent, SERD-resistant breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Drug Resistance, Neoplasm , Estradiol/analogs & derivatives , MicroRNAs/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/metabolism , Female , Fulvestrant , Gene Expression Regulation, Neoplastic/drug effects , Humans , MicroRNAs/metabolism , Oligonucleotide Array Sequence Analysis , Selective Estrogen Receptor Modulators/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Transforming Growth Factor beta/metabolism , Up-Regulation , beta Catenin/genetics
5.
Radiat Prot Dosimetry ; 144(1-4): 85-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21062805

ABSTRACT

The systematic quality assurance (QA) and control of testing methods in incorporation monitoring consists of continual measures for internal QA and additional measures such as external laboratory controls. This includes among other aspects accuracy, precision and descriptions of the methods as well as the representation and timely availability of analytic results of measurements and internal dose assessment. At the officially recognised incorporation measurement office at Jülich, QA is performed for direct measurements (whole-body counter), indirect measurements with radiochemical testing methods of excretion samples and internal dose assessment.


Subject(s)
Laboratories/standards , Quality Control , Radiation Monitoring/standards , Radiation Protection/standards , Radiometry/standards , Cesium Radioisotopes/analysis , Germany , Humans , Mass Spectrometry/methods , Radiation Monitoring/methods , Radiation Protection/methods , Radiometry/methods , Reproducibility of Results , Uranium/analysis , Whole-Body Counting
6.
Theor Appl Genet ; 108(6): 1064-70, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15067392

ABSTRACT

A major constraint to the application of biotechnology to the improvement of the allotetraploid peanut, or groundnut ( Arachis hypogaea L.), has been the paucity of polymorphism among germplasm lines using biochemical (seed proteins, isozymes) and DNA markers (RFLPs and RAPDs). Six sequence-tagged microsatellite (STMS) markers were previously available that revealed polymorphism in cultivated peanut. Here, we identify and characterize 110 STMS markers that reveal genetic variation in a diverse array of 24 peanut landraces. The simple-sequence repeats (SSRs) were identified with a probe of two 27648-clone genomic libraries: one constructed using PstI and the other using Sau3AI/ BamHI. The most frequent, repeat motifs identified were ATT and GA, which represented 29% and 28%, respectively, of all SSRs identified. These were followed by AT, CTT, and GT. Of the amplifiable primers, 81% of ATT and 70.8% of GA repeats were polymorphic in the cultivated peanut test array. The repeat motif AT showed the maximum number of alleles per locus (5.7). Motifs ATT, GT, and GA had a mean number of alleles per locus of 4.8, 3.8, and 3.6, respectively. The high mean number of alleles per polymorphic locus, combined with their relative frequency in the genome and amenability to probing, make ATT and GA the most useful and appropriate motifs to target to generate further SSR markers for peanut.


Subject(s)
Arachis/genetics , Genetic Variation , Genomic Library , Microsatellite Repeats/genetics , Agriculture , DNA Primers , Gene Frequency , Species Specificity
7.
J Steroid Biochem Mol Biol ; 78(5): 409-18, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11738551

ABSTRACT

In oestrogen receptor (ER)-positive breast carcinoma cells, 17beta-oestradiol suppresses a dose-dependent induction of cell death by tumour necrosis factor alpha (TNF). The ability of oestrogens to promote cell survival in ER-positive breast carcinoma cells is linked to a coordinate increase in Bcl-2 expression, an effect that is blocked with the pure anti-oestrogen ICI 182,780. The role of Bcl-2 in MCF-7 cell survival was confirmed by stable overexpression of Bcl-2 which resulted in suppression of apoptosis induced by doxorubicin (DOX), paclitaxel (TAX) and TNF as compared to vector-control cells. The pure anti-oestrogen ICI 182,780 in combination with TNF, DOX or TAX potentiated apoptosis in vector-transfected cells. Interestingly, pre-treatment with ICI 182,780 markedly enhanced chemotherapeutic drug- or TNF-induced apoptosis in Bcl-2 expressing cells, an effect that was correlated with ICI 182,780 induced activation of c-Jun N-terminal kinase. Our results suggest that the effects of oestrogens/anti-oestrogens on the regulation of apoptosis may involve coordinate activation of signalling events and Bcl-2 expression.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Estradiol/analogs & derivatives , Estradiol/pharmacology , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/pathology , Tumor Necrosis Factor-alpha/pharmacology , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Survival/drug effects , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Drug Interactions , Estradiol/administration & dosage , Estrogen Receptor Modulators/administration & dosage , Estrogen Receptor Modulators/pharmacology , Female , Fulvestrant , Genes, bcl-2 , Humans , Mitogen-Activated Protein Kinases/metabolism , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/metabolism , Paclitaxel/administration & dosage , Paclitaxel/pharmacology , Signal Transduction , Transfection , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/administration & dosage
8.
Mol Carcinog ; 32(4): 213-9, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11746833

ABSTRACT

Mouse abdominal B-like Hoxa genes are expressed and functionally required in the developing reproductive tracts. Mice lacking either Hoxa-10 or Hoxa-11, two of the AbdB Hoxa genes, exhibit abnormal uterine development similar to that induced by in utero diethylstilbestrol (DES) exposure. Indeed, uterine Hoxa-10 and Hoxa-11 expression is potently repressed by perinatal DES exposure, providing a potential molecular mechanism for DES-induced reproductive tract malformations. We have shown previously that DES can permanently alter uterine lactoferrin gene expression through modulation of the lactoferrin promoter methylation pattern. Here we ask whether a similar mechanism also functions to deregulate uterine Hoxa-10 or Hoxa-11 expression during neonatal DES exposure. We mapped the Hoxa-10 promoter by cloning a 1.485 kb DNA fragment 5' of the Hoxa-10 exon1a. A 5' rapid amplification of cDNA ends (RACE) experiment revealed a transcription start site for the a10-1 transcript. Functional analysis of the proximal 200-bp sequences demonstrated significant promoter activity, confirming the location of the Hoxa-10 promoter. Moreover, methylation assays performed on eight CpGs in Hoxa-10 and 19 CpGs in Hoxa-11 proximal promoters demonstrated that all these CpGs were highly unmethylated in both control and DES-dosed mice from postnatal day 5 to day 30. Significant methylation around Hoxa-10 and Hoxa-11 promoters was only observed in DES-induced uterine carcinomas in 18-mo-old mice. Our results suggest that DES-induced downregulations of Hoxa-10 or Hoxa-11 gene expression are not associated with methylation changes in their proximal promoters and that gene imprinting by developmental DES exposure may be a gene-specific phenomenon.


Subject(s)
DNA Methylation/drug effects , DNA-Binding Proteins/genetics , Diethylstilbestrol/pharmacology , Estrogens, Non-Steroidal/pharmacology , Homeodomain Proteins/genetics , Uterus/physiology , Animals , Animals, Newborn , Base Sequence , Female , Gene Expression Regulation/drug effects , Homeobox A10 Proteins , Humans , Mice , Molecular Sequence Data , Promoter Regions, Genetic/genetics
9.
Genetics ; 159(2): 823-37, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11606556

ABSTRACT

Polyploidy creates severe genetic bottlenecks, contributing to the genetic vulnerability of leading crops. Cultivated peanut is thought to be of monophyletic origin, harboring relatively little genetic diversity. To introduce variability from diploid wild species into tetraploid cultivated Arachis hypogaea, a synthetic amphidiploid [[A. batizocoi K9484 x (A. cardenasii GKP10017 x A. diogoi GKP10602)](4x)] was used as donor parent to generate a backcross population of 78 progeny. Three hundred seventy RFLP loci were mapped onto 23 linkage groups, spanning 2210 cM. Chromatin derived from the two A-genome diploid ancestors (A. cardenasii and A. diogoi) comprised mosaic chromosomes, reflecting crossing over in the diploid A-genome interspecific F(1) hybrid. Recombination between chromosomes in the tetraploid progeny was similar to chromosome pairing reported for A. hypogaea, with recombination generally between chromosomes of the same subgenomic affinity. Segregation distortion was observed for 25% of the markers, distributed over 20 linkage groups. Unexpectedly, 68% of the markers deviating from expected segregation showed an excess of the synthetic parent allele. Genetic consequences, relationship to species origins, and significance for comparative genetics are discussed.


Subject(s)
Arachis/genetics , Chromatin/genetics , Diploidy , Genetic Linkage , Polymorphism, Restriction Fragment Length , Recombination, Genetic
10.
Pediatr Clin North Am ; 48(5): 1223-40, x, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11579671

ABSTRACT

What do pseudohermaphroditic polar bears and girls with premature breast development have in common? Hormones. Sexual differentiation and the initiation of secondary sexual characteristics, such as breast growth, are both under the control of the sex hormones estrogen and androgen. Abnormal differentiation of the internal or external genitalia in bears and early onset of breast development in girls also may have a common element--exposure to environmental hormones.


Subject(s)
Disorders of Sex Development/physiopathology , Endocrine System Diseases/etiology , Endocrine System Diseases/physiopathology , Environmental Exposure/adverse effects , Gonadal Steroid Hormones/adverse effects , Pediatrics , Puberty/physiology , Sex Differentiation/physiology , Adolescent , Alligators and Crocodiles , Animals , Birds , Child , Environmental Health , Female , Fishes , Humans , Male , Seals, Earless , Ursidae
11.
Nature ; 413(6852): 128-9, 2001 Sep 13.
Article in English | MEDLINE | ID: mdl-11557969

ABSTRACT

Nitrogen fixation is a symbiotic process initiated by chemical signals from legumes that are recognized by soil bacteria. Here we show that some endocrine-disrupting chemicals (EDCs), so called because of their effect on hormone-signalling pathways in animal cells, also interfere with the symbiotic signalling that leads to nitrogen fixation. Our results raise the possibility that these phytochemically activated pathways may have features in common with hormonal signalling in vertebrates, thereby extending the biological and ecological impact of EDCs.


Subject(s)
Hormones , Nitrogen Fixation , Pesticides/pharmacology , Phenols/pharmacology , Medicago sativa/microbiology , Medicago sativa/physiology , Pesticides/chemistry , Phenols/chemistry , Rhizobiaceae/physiology , Signal Transduction/drug effects , Symbiosis
12.
Surgery ; 130(2): 143-50, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11490342

ABSTRACT

BACKGROUND: Nuclear factor-kappa B (NF-kappa B) is a known survival pathway, and it may explain differential sensitivity to tumor necrosis factor-alpha (TNF-alpha) and chemotherapeutic-induced apoptosis in apoptotically sensitive (APO+) and apoptotically resistant (APO-) Michigan Cancer Foundation-7 breast cancer cells. METHODS: Crystal violet viability and luciferase reporter gene assays were used to determine the inhibitory concentration of viability at 50% (IC(50)) and the inhibitory concentration of activity at 50% (EC(50)) values in APO- and APO+ cells with the selective NF-kappa B inhibitor, BAY 11-7082 (BAY). The apoptotic reporter assay was used to determine the effects of the transfection of the inhibitory kappa B-dominant negative (I kappa B-DN) construct in conjunction with TNF, paclitaxel, or doxorubicin treatments in these cells. RESULTS: The concentrations at which 50% of cell viability is inhibited (IC(50)) and at which 50% of NF-kappa B activity is inhibited (EC(50)) for BAY in APO- and APO+ cells were 95.24 micromol/L and 1.53 micromol/L, respectively, and 7.62 micromol/L and 2.64 micromol/L, respectively. The IC(50) and the EC(50) values were equivalent for the APO+ cells (P =.665), but not for the APO- cells (P =.025). I kappa B-DN--transfection alone, or with TNF, doxorubicin, or paclitaxel treatments resulted in cell death of both APO- and APO+ cells as compared with vector-control; however, greater cytotoxicity was seen in the APO+ cells. Direct comparison of the APO+ cells versus the APO- cells revealed that these differences were significant (P =.05). CONCLUSIONS: Pharmacologic or molecular inhibition of the NF-kappa B pathway blocked cell survival in MCF-7 APO+ cells, while only molecular inhibition induced cytotoxicity in the APO- cells. Selective manipulation of the NF-kappa B pathway in combination with standard chemotherapeutic agents may lead to an increased potency and efficacy of these agents.


Subject(s)
Breast Neoplasms , Drug Resistance, Neoplasm , NF-kappa B/genetics , NF-kappa B/metabolism , Nitriles , Organic Chemicals , Sulfones , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Doxorubicin/pharmacology , Female , Gene Expression/drug effects , Genes, Reporter , Humans , Inhibitory Concentration 50 , Luciferases/genetics , NF-kappa B/antagonists & inhibitors , Paclitaxel/pharmacology , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects
13.
APMIS ; 109(4): 263-72, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11469497

ABSTRACT

For three decades, we have known that estrogens alter the development of the mammalian reproductive system in predictable ways. In mice exposed prenatally to diethylstilbestrol (DES) or other estrogens, the male offspring exhibit structural malformations including cryptorchidism, epididymal cysts and retained Mullerian ducts. The estrogen-associated alterations in the genital tract phenotype can be usefully considered as a model called Developmental Estrogenization Syndrome. While estrogen treatment during critical periods of morphogenesis of the male reproductive system has been associated with these changes, the mechanisms at the molecular level are still being discovered. Parallel findings on the hormones involved in Mullerian duct regression and testicular descent have helped guide research on the mechanisms of developmental estrogenization of the male. Cellular localization of molecular signals associated with key steps in genital tract development, use of mice with gene disruption, and knowledge of the mechanisms underlying persistent changes in gene expression are beginning to provide a blue print for both the physiological role and pathological effects of estrogens in reproductive tract development. Since many of the same biological principles underlie genital tract morphogenesis in mammals, one may expect some of the same changes in males of other species exposed to estrogen during the appropriate developmental periods.


Subject(s)
Abnormalities, Drug-Induced/etiology , Diethylstilbestrol/toxicity , Estrogens, Non-Steroidal/toxicity , Genitalia, Male/abnormalities , Abnormalities, Drug-Induced/embryology , Animals , DNA Methylation/drug effects , Female , Genes, Reporter/drug effects , Genomic Imprinting , Humans , Male , Maternal-Fetal Exchange , Mice , Phenotype , Pregnancy , Sex Differentiation/drug effects , Syndrome
14.
Mol Cell Endocrinol ; 176(1-2): 67-75, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11369444

ABSTRACT

Leptin is a potential regulator of conceptus development. We have previously suggested that in primate pregnancy, leptin biosynthesis is regulated by estrogen in a tissue-specific manner. Therefore, the objective of the current study was to determine the mechanism of estrogen action on LEP promoter activation in divergent cell types. The effects of estrogen were investigated in estrogen receptor (ER)-positive MCF-7 breast cancer cells and in ER-negative JEG-3 choriocarcinoma cells. Cells were transfected with a leptin-luciferase or an estrogen responsive element (ERE)-luciferase reporter construct, in conjunction with ERalpha, ERbeta, or empty vector expression plasmids. Cells were treated with estradiol and/or the specific estrogen antagonists, ICI-182,780 or 4-hydroxytamoxifen. In MCF-7 cells, estradiol stimulated (P<0.05) ERE-luciferase activity and was inhibited by ICI-182,780, but did not stimulate leptin-luciferase activity. However, leptin-luciferase was stimulated by estradiol (P<0.05) and inhibited by antiestrogens in JEG-3 cells that were co-transfected with ERalpha. Both antiestrogens stimulated leptin-luciferase activity (P<0.05) in JEG-3 cells co-transfected with ERbeta. Results suggested that LEP promoter activation may depend upon co-activators present in leptin-producing cells and may be inhibited by repressors present in non-leptin producing cells. Divergent effects of estrogen may be owed to differences in the type of ER (alpha or beta) expressed in target tissues.


Subject(s)
Breast Neoplasms/metabolism , Choriocarcinoma/metabolism , Estradiol/pharmacology , Leptin/genetics , Promoter Regions, Genetic/genetics , Receptors, Estrogen/metabolism , Transcriptional Activation/drug effects , Estradiol/analogs & derivatives , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha , Estrogen Receptor beta , Female , Fulvestrant , Genes, Reporter/genetics , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Estrogen/genetics , Response Elements/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Tumor Cells, Cultured
15.
Toxicol Lett ; 120(1-3): 161-4, 2001 Mar 31.
Article in English | MEDLINE | ID: mdl-11323173

ABSTRACT

Gene imprinting is an epigenetic mechanism for accomplishing persistent change in gene expression. In this brief paper, we explore the mechanisms for imprinting genes and present data showing that the synthetic estrogen, diethylstilbestrol (DES) can developmentally imprint genes by changing the pattern of DNA methylation. We further discuss the implications of this and other findings for non-mutagenic aspects of developmental toxicology, and suggest ways to use this concept in modifying in vitro screening for developmental toxicants.


Subject(s)
Fetus/drug effects , Genomic Imprinting , Toxicology , Animals , DNA Methylation/drug effects , Diethylstilbestrol/toxicity , Estrogens/toxicity , Humans
16.
J Clin Endocrinol Metab ; 86(4): 1750-8, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11297613

ABSTRACT

The flavonoid family of phytochemicals, particularly those derived from soy, has received attention regarding their estrogenic activity as well as their effects on human health and disease. In addition to these flavonoids other phytochemicals, including phytostilbene, enterolactone, and lignans, possess endocrine activity. The types and amounts of these compounds in soy and other plants are controlled by both constitutive expression and stress-induced biosynthesis. The health benefits of soy-based foods may, therefore, be dependent upon the amounts of the various hormonally active phytochemicals within these foods. The aim was to identify unique soy phytochemicals that had not been previously assessed for estrogenic or antiestrogenic activity. Here we describe increased biosynthesis of the isoflavonoid phytoalexin compounds, glyceollins, in soy plants grown under stressed conditions. In contrast to the observed estrogenic effects of coumestrol, daidzein, and genistein, we observed a marked antiestrogenic effect of glyceollins on ER signaling, which correlated with a comparable suppression of 17 beta-estradiol-induced proliferation in MCF-7 cells. Further evaluation revealed greater antagonism toward ER alpha than ER beta in transiently transfected HEK 293 cells. Competition binding assays revealed a greater affinity of glyceollins for ER alpha vs. ER beta, which correlated to greater suppression of ER alpha signaling with higher concentrations of glyceollins. In conclusion, we describe the phytoalexin compounds known as glyceollins, which exhibit unique antagonistic effects on ER in both HEK 293 and MCF-7 cells. The glyceollins as well as other phytoalexin compounds may represent an important component of the health effects of soy-based foods.


Subject(s)
Benzopyrans/pharmacology , Glycine max/chemistry , Hormone Antagonists/pharmacology , Plant Extracts/pharmacology , Receptors, Estrogen/physiology , Benzopyrans/metabolism , Binding, Competitive , Cell Division/drug effects , Cell Line/cytology , Cell Line/drug effects , Estradiol/pharmacology , Estrogen Receptor alpha , Estrogen Receptor beta , Humans , Osmolar Concentration , Pterocarpans , Receptors, Estrogen/metabolism , Signal Transduction/drug effects
17.
ScientificWorldJournal ; 1: 653-5, 2001 Nov 13.
Article in English | MEDLINE | ID: mdl-12805768

ABSTRACT

Endocrine disrupting chemicals (EDCs) include organochlorine pesticides, plastics manufacturing by-products, and certain herbicides. These chemicals have been shown to disrupt hormonal signaling in exposed wildlife, lab animals, and mammalian cell culture by binding to estrogen receptors (ER-+/- and ER-) and affecting the expression of estrogen responsive genes. Additionally, certain plant chemicals, termed phytoestrogens, are also able to bind to estrogen receptors and modulate gene expression, and as such also may be considered EDCs. One example of phytoestrogen action is genistein, a phytochemical produced by soybeans, binding estrogen receptors, and changing expression of estrogen responsive genes which certain studies have linked to a lower incidence of hormonally related cancers in Japanese populations. Why would plants make compounds that are able to act as estrogens in the human body? Obviously, soybeans do not intentionally produce phytoestrogens to prevent breast cancer in Japanese women.


Subject(s)
Endocrine System/drug effects , Gene Expression Regulation, Bacterial/drug effects , Gene Expression Regulation, Bacterial/genetics , Herbicides/pharmacology , Hydrocarbons, Chlorinated , Insecticides/pharmacology , Symbiosis/drug effects , Symbiosis/genetics , Animals , Gene Expression Regulation, Bacterial/physiology , Humans , Isoflavones/antagonists & inhibitors , Isoflavones/physiology , Phytoestrogens , Plant Preparations/antagonists & inhibitors , Plants/chemistry , Symbiosis/physiology , Transcriptional Activation
19.
J Biol Chem ; 275(36): 27694-702, 2000 Sep 08.
Article in English | MEDLINE | ID: mdl-10874044

ABSTRACT

The mouse heme oxygenase-1 (HO-1) gene, ho-1, contains two inducible enhancers, E1 and E2. Of several cell lines tested, induction of an E1/luciferase fusion construct, pE1-luc, by CdCl(2) is most pronounced in MCF-7 cells. In these cells, E1, but not E2, is necessary and sufficient for ho-1 gene activation. Exposure of MCF-7 cells to 10 micrometer CdCl(2) stimulates phosphorylation of ERK, JNK, and p38 mitogen-activated protein kinases, implicating one or more of these signaling pathways in ho-1 gene induction. SB203580, an inhibitor of p38, diminishes cadmium-stimulated pE1-luc expression and HO-1 mRNA levels by up to 70-80%. PD098059, an ERK pathway inhibitor, does not affect HO-1 mRNA induction at the highest concentration (40 micrometer) tested. Similarly, co-expression of a dominant-negative mutant of p38alpha, but not of ERK1, ERK2, JNK1, or JNK2, reduces basal and cadmium-induced pE1-luc activity. E1 contains binding sites for the activator protein-1 (Fos/Jun), Cap'n'Collar/basic leucine zipper (CNC-bZIP), and CCAAT/enhancer-binding protein (C/EBP) families of transcription factors. A dominant-negative mutant of Nrf2 (a CNC-bZIP member), but not of c-Jun or C/EBPbeta, inhibits pE1-luc activation by cadmium. Induction of the endogenous ho-1 gene is also inhibited by the Nrf2 mutant. Mutations of E1 that inhibit cadmium inducibility also suppress the trans-activation and DNA binding activities of Nrf2, and SB203580, but not PD098059, attenuates Nrf2-mediated trans-activation of pE1-luc. Taken together, these results indicate that cadmium induces ho-1 gene expression via sequential activation of the p38 kinase pathway and Nrf2.


Subject(s)
Cadmium/pharmacology , DNA-Binding Proteins/metabolism , Epithelial Cells/enzymology , Gene Expression Regulation, Enzymologic/drug effects , Heme Oxygenase (Decyclizing)/genetics , Mitogen-Activated Protein Kinases/metabolism , Trans-Activators/metabolism , Transcription, Genetic/drug effects , Animals , Base Sequence , Breast Neoplasms , Cell Line , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Female , Flavonoids/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , HeLa Cells , Heme Oxygenase-1 , Humans , L Cells , Membrane Proteins , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase 9 , Molecular Sequence Data , NF-E2-Related Factor 2 , Transcription Factors/metabolism , Transcriptional Activation , Tumor Cells, Cultured , p38 Mitogen-Activated Protein Kinases
20.
Int J Oncol ; 16(6): 1179-87, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10811993

ABSTRACT

Widespread use of MCF-7 human breast cancer cells as a model system for breast cancer has lead to variations in these cells between different laboratories. Although several reports have addressed these differences in terms of proliferation and estrogenic response, differences in sensitivity to apoptosis have just begun to be described. Based on the possible differences in apoptotic sensitivity that may arise due to the existence of MCF-7 cell variants, we determined the relative sensitivity of MCF-7 cell variants from three established laboratories (designated M, L and N) to known inducers of apoptosis. Consistent with our previous studies we demonstrate that differences exist among these variants in regards to tumor necrosis factor alpha (TNF)-induced cell death and inhibition of proliferation in a dose-dependent manner. To establish if the difference in apoptotic susceptibility was specific to TNF, the three MCF-7 cell variants were tested for their response to other known inducers of apoptosis: okadaic acid, staurosporine and 4-hydroxy-tamoxifen. Viability and DNA fragmentation analysis revealed a similar pattern of resistance to apoptosis by all agents in the MCF-7 M variant. The MCF-7 L variant was resistant to okadaic acid and 4-hydroxy-tamoxifen but not staurosporine. In contrast, MCF-7 N cells were sensitive to induction of apoptosis by all agents. The role of both protein kinase C (PKC) and estrogen signaling in the regulation of cell survival prompted investigation of these pathways as a mechanism for differential sensitivity of MCF-7 cell variants to apoptosis. While both estrogen receptor alpha (ERalpha) and ERbeta were expressed in MCF-7 M and N cells, the absence of ERbeta in MCF-7 L cells correlated with decreased estrogen responsiveness of the L variant. Variations in estrogenic responsiveness and PKC isoform expression may account for the enhanced susceptibility of both the L and N variants to staurosporine.


Subject(s)
Breast Neoplasms/metabolism , Protein Kinase C/metabolism , Receptors, Estrogen/metabolism , Tumor Cells, Cultured/metabolism , Apoptosis/drug effects , Apoptosis/physiology , Breast Neoplasms/physiopathology , Estradiol/pharmacology , Estrogen Receptor alpha , Estrogen Receptor beta , Female , Humans , Protein Isoforms/pharmacology , Receptors, Estrogen/drug effects , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/physiology
SELECTION OF CITATIONS
SEARCH DETAIL