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










Publication year range
1.
Clin Proteomics ; 15: 30, 2018.
Article in English | MEDLINE | ID: mdl-30250408

ABSTRACT

In recent years, there has been an emphasis on personalizing breast cancer treatment in order to avoid the debilitating side effects caused by broad-spectrum chemotherapeutic drug treatment. Development of personalized medicine requires the identification of proteins that are expressed by individual tumors. Herein, we reveal the identity of plasma membrane proteins that are overexpressed in estrogen receptor α-positive, HER2-positive, and triple negative breast cancer cells. The proteins we identified are involved in maintaining protein structure, intracellular homeostasis, and cellular architecture; enhancing cell proliferation and invasion; and influencing cell migration. These proteins may be useful for breast cancer detection and/or treatment.

2.
PLoS One ; 11(6): e0158296, 2016.
Article in English | MEDLINE | ID: mdl-27355404

ABSTRACT

Cancer cells secrete factors that influence adjacent cell behavior and can lead to enhanced proliferation and metastasis. To better understand the role of these factors in oncogenesis and disease progression, estrogen and progesterone receptor positive MCF-7 cells, triple negative breast cancer MDA-MB-231, DT22, and DT28 cells, and MCF-10A non-transformed mammary epithelial cells were grown in 3D cultures. A special emphasis was placed on triple negative breast cancer since these tumors are highly aggressive and no targeted treatments are currently available. The breast cancer cells secreted factors of variable potency that stimulated proliferation of the relatively quiescent MCF-10A cells. The conditioned medium from each cell line was subjected to mass spectrometry analysis and a variety of secreted proteins were identified including glycolytic enzymes, proteases, protease inhibitors, extracellular matrix proteins, and insulin-like growth factor binding proteins. An investigation of the secretome from each cell line yielded clues about strategies used for breast cancer proliferation and metastasis. Some of the proteins we identified may be useful in the development of a serum-based test for breast cancer detection, diagnosis, prognosis, and monitoring.


Subject(s)
Biomarkers, Tumor/blood , Breast Neoplasms/blood , Breast Neoplasms/diagnosis , Breast/pathology , Cell Culture Techniques , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic , Early Detection of Cancer/methods , Female , Glycolysis , Humans , Neoplasm Metastasis , Prognosis
3.
Mol Cell Endocrinol ; 426: 11-21, 2016 May 05.
Article in English | MEDLINE | ID: mdl-26872614

ABSTRACT

Although substantial evidence has demonstrated that parity and 17ß-estradiol (E2) reduce mammary carcinogenesis, it is not clear how this protection is conferred. Thus, we examined the effects of parity and E2 treatment in the mammary glands of ovariectomized 15 week-old virgin mice, 15 week-old primiparous mice, and 9 month-old retired breeders. E2 treatment significantly increased lipid peroxidation, protein carbonylation, and protein nitrosylation in the virgin mice, but not in the age-matched primiparous mice or retired breeders. Mammary gland expression of the oxidative stress response protein Cu/Zn superoxide dismutase was consistently reduced in all of the E2-treated mice regardless of parity. Expression of the oxidative stress and DNA repair protein apurinic endonuclease (Ape1) was significantly increased only in the mammary glands of the E2-treated retired breeders. These findings suggest that E2 and parity help to reduce mammary oncogenesis by maintaining the structure and function of proteins, lipids, and DNA.


Subject(s)
Estradiol/pharmacology , Mammary Glands, Animal/metabolism , Oxidative Stress , Animals , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Female , Gene Expression , Lipid Peroxidation , Mammary Glands, Animal/cytology , Mice, Inbred C57BL , Parity , Pregnancy , Protective Factors , Protein Carbonylation
4.
PLoS One ; 9(11): e111975, 2014.
Article in English | MEDLINE | ID: mdl-25372139

ABSTRACT

17ß-estradiol (E2) plays critical roles in a number of target tissues including the mammary gland, reproductive tract, bone, and brain. Although it is clear that E2 reduces inflammation and ischemia-induced damage in the cerebral cortex, the molecular mechanisms mediating the effects of E2 in this brain region are lacking. Thus, we examined the cortical transcriptome using a mouse model system. Female adult mice were ovariectomized and implanted with silastic tubing containing oil or E2. After 7 days, the cerebral cortices were dissected and RNA was isolated and analyzed using RNA-sequencing. Analysis of the transcriptomes of control and E2-treated animals revealed that E2 treatment significantly altered the transcript levels of 88 genes. These genes were associated with long term synaptic potentiation, myelination, phosphoprotein phosphatase activity, mitogen activated protein kinase, and phosphatidylinositol 3-kinase signaling. E2 also altered the expression of genes linked to lipid synthesis and metabolism, vasoconstriction and vasodilation, cell-cell communication, and histone modification. These results demonstrate the far-reaching and diverse effects of E2 in the cerebral cortex and provide valuable insight to begin to understand cortical processes that may fluctuate in a dynamic hormonal environment.


Subject(s)
Cerebral Cortex/metabolism , Estradiol/metabolism , Gene Expression Regulation , Animals , Cerebral Cortex/blood supply , Cerebral Cortex/drug effects , Cluster Analysis , Estradiol/pharmacology , Female , Gene Expression Profiling , Gene Expression Regulation/drug effects , Gene Regulatory Networks , Mice , Microvessels/drug effects , Microvessels/metabolism , Reproducibility of Results , Signal Transduction
5.
PLoS One ; 9(7): e102341, 2014.
Article in English | MEDLINE | ID: mdl-25029196

ABSTRACT

The use of broad spectrum chemotherapeutic agents to treat breast cancer results in substantial and debilitating side effects, necessitating the development of targeted therapies to limit tumor proliferation and prevent metastasis. In recent years, the list of approved targeted therapies has expanded, and it includes both monoclonal antibodies and small molecule inhibitors that interfere with key proteins involved in the uncontrolled growth and migration of cancer cells. The targeting of plasma membrane proteins has been most successful to date, and this is reflected in the large representation of these proteins as targets of newer therapies. In view of these facts, experiments were designed to investigate the plasma membrane proteome of a variety of human breast cancer cell lines representing hormone-responsive, ErbB2 over-expressing and triple negative cell types, as well as a benign control. Plasma membranes were isolated by using an aqueous two-phase system, and the resulting proteins were subjected to mass spectrometry analysis. Overall, each of the cell lines expressed some unique proteins, and a number of proteins were expressed in multiple cell lines, but in patterns that did not always follow traditional clinical definitions of breast cancer type. From our data, it can be deduced that most cancer cells possess multiple strategies to promote uncontrolled growth, reflected in aberrant expression of tyrosine kinases, cellular adhesion molecules, and structural proteins. Our data set provides a very rich and complex picture of plasma membrane proteins present on breast cancer cells, and the sorting and categorizing of this data provides interesting insights into the biology, classification, and potential treatment of this prevalent and debilitating disease.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/drug therapy , Drug Delivery Systems/methods , Membrane Proteins/metabolism , Blotting, Western , Breast Neoplasms/metabolism , Cell Line, Tumor , DNA Primers/genetics , Female , Humans , Mass Spectrometry , Membrane Proteins/genetics , Microscopy, Fluorescence , Proteomics , Reverse Transcriptase Polymerase Chain Reaction
6.
J Steroid Biochem Mol Biol ; 127(3-5): 382-9, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21704159

ABSTRACT

A number of studies have demonstrated that 17ß-estradiol (E(2)) protects the brain from ischemia and yet the mechanism by which this hormone brings about its protective effect is unclear. Interestingly, like E(2), overexpression of the oxidative stress response protein Cu/Zn superoxide dismutase (SOD1), which plays a critical role in regulating reactive oxygen species, also protects the brain from ischemia. Because we previously showed that E(2) treatment of cultured mammary cells increases SOD1 expression, we hypothesized that E(2) might increase SOD1 expression in the brain and that this E(2)-mediated increase in SOD1 expression might help to protect the brain from ischemia. We now show that SOD1 is expressed in cortical neurons, that SOD1 expression is increased by exposure of brain slice cultures to E(2), and that the E(2)-mediated increase in SOD1 expression is further augmented by exposure of brain slice cultures to increased superoxide levels or oxygen and glucose deprivation. Importantly, when cortical neurons are exposed to increased superoxide levels and markers of protein and DNA damage, nitrotyrosine and 8-oxoguanine, respectively, are measured, both protein and DNA damage are reduced. In fact, E(2) reduces nitrotyrosine and 8-oxoguanine levels in brain slice cultures regardless of whether they have or have not been exposed to increased superoxide levels. Likewise, when brain slice cultures are treated with E(2) and deprived of oxygen and glucose, 8-oxoguanine levels are reduced. Taken together, these studies provide a critical link between E(2) treatment, SOD1 expression, and neuroprotection and help to define a mechanism through which E(2)-mediated neuroprotection may be conferred.


Subject(s)
Brain/drug effects , Estradiol/pharmacology , Ischemia/prevention & control , Neurons/pathology , Superoxide Dismutase/metabolism , Animals , Brain/enzymology , Brain/metabolism , Brain/pathology , DNA Damage , Estrogen Receptor alpha/metabolism , Immunohistochemistry , Mice , Mice, Inbred C57BL , Oxidative Stress , Reactive Oxygen Species/metabolism
7.
Trends Endocrinol Metab ; 22(4): 124-9, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21371903

ABSTRACT

Estrogen receptor α (ERα) is a ligand-activated transcription factor that, upon binding hormone, interacts with specific recognition sequences in DNA. An extensive body of literature has documented the association of individual regulatory proteins with ERα. It has recently become apparent that, instead of simply recruiting individual proteins, ERα recruits interconnected networks of proteins with discrete activities that play crucial roles in maintaining the structure and function of the receptor, stabilizing the receptor-DNA interaction, influencing estrogen-responsive gene expression, and repairing misfolded proteins and damaged DNA. Together these studies suggest that the DNA-bound ERα serves as a nucleating factor for the recruitment of protein complexes involved in key processes including the oxidative stress response, DNA repair, and transcription regulation.


Subject(s)
Estrogen Receptor alpha/metabolism , Animals , DNA Repair/genetics , DNA Repair/physiology , Estrogen Receptor alpha/genetics , Humans , Models, Biological , Oxidative Stress/genetics , Oxidative Stress/physiology
8.
Mol Endocrinol ; 24(2): 346-58, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19952285

ABSTRACT

Estrogen receptor alpha (ERalpha) binds to specific target DNA sequences, estrogen response elements (EREs), to regulate estrogen-responsive gene expression. The progesterone receptor (PR) gene has been used extensively as a marker of estrogen responsiveness. Although we previously identified cis elements within 1 kb of the PR-B transcription start site that are associated with ERalpha and help to confer estrogen responsiveness, the identification of ERalpha binding sites far removed from the transcription start site suggested that long-range regulation of this gene may occur. We now show that eight regions of the PR gene from 311 kb upstream to 4 kb downstream of the PR-B transcription start site interact with ERalpha and that coactivator proteins and acetylated histones are selectively associated with these gene regions. Specific PR gene regions confer estrogen responsiveness to a heterologous reporter plasmid, and mutation of EREs within these regions diminishes estrogen-induced transactivation. Importantly, chromosome conformation capture assays reveal ERalpha- and ligand-dependent interactions between proximal and distal PR gene regions. Taken together, our studies suggest that distal regions of the PR gene participate in the dynamic regulation of this gene and that the coordinated action of proximal and distal PR gene regions allows cells to respond to changes in hormone levels with extraordinary versatility and sensitivity.


Subject(s)
DNA-Binding Proteins/metabolism , Estrogen Receptor alpha/metabolism , Estrogens/pharmacology , Gene Expression Regulation , Receptors, Progesterone/genetics , Response Elements/genetics , Acetylation/drug effects , Cell Line, Tumor , Chromatin/chemistry , Computational Biology/methods , Estradiol/pharmacology , Forkhead Transcription Factors/metabolism , Histones/metabolism , Humans , Nuclear Receptor Coactivator 3/metabolism , Nucleic Acid Conformation , RNA, Messenger/metabolism , Receptors, Progesterone/metabolism , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism , TRPC6 Cation Channel , Time Factors , p300-CBP Transcription Factors/metabolism
9.
Methods Mol Biol ; 590: 209-21, 2009.
Article in English | MEDLINE | ID: mdl-19763506

ABSTRACT

Regulating gene expression is a complex process requiring the interaction of multiple transcription factors with their cognate recognition sequences. While these DNA-bound transcription factors are the primary drivers of gene expression, the capacity of a transcription factor to alter gene expression is tempered by its association with a host of coregulatory proteins that are recruited to the DNA-bound transcription factor. We have developed a novel approach to isolate large complexes of proteins associated with the DNA-bound estrogen receptor alpha (ERalpha) using an agarose-based electrophoretic mobility shift assay (EMSA). This method should be readily adapted to a variety of cultured cell lines, DNA sequences, and transcription factors and has the potential to provide valuable information about a wide variety of regulatory proteins involved in influencing gene expression.


Subject(s)
DNA/metabolism , Estrogen Receptor alpha/metabolism , Proteins/isolation & purification , Base Sequence , DNA Primers , Electrophoresis, Agar Gel , Electrophoretic Mobility Shift Assay , Estrogen Receptor alpha/genetics , HeLa Cells , Humans , Proteins/metabolism , Transcription, Genetic
10.
J Mol Endocrinol ; 43(6): 251-61, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19620238

ABSTRACT

Accumulation of reactive oxygen species (ROS) in cells damages resident proteins, lipids, and DNA. In order to overcome the oxidative stress that occurs with ROS accumulation, cells must balance free radical production with an increase in the level of antioxidant enzymes that convert free radicals to less harmful species. We identified two antioxidant enzymes, thioredoxin (Trx) and Trx reductase (TrxR), in a complex associated with the DNA-bound estrogen receptor alpha (ERalpha). Western analysis and immunocytochemistry were used to demonstrate that Trx and TrxR are expressed in the cytoplasm and in the nuclei of MCF-7 human breast cancer cells. More importantly, endogenously expressed ERalpha, Trx, and TrxR interact and ERalpha and TrxR associate with the native, estrogen-responsive pS2 and progesterone receptor genes in MCF-7 cells. RNA interference assays demonstrated that Trx and TrxR differentially influence estrogen-responsive gene expression and that together, 17beta-estradiol, Trx, and TrxR alter hydrogen peroxide (H(2)O(2)) levels in MCF-7 cells. Our findings suggest that Trx and TrxR are multifunctional proteins that, in addition to modulating H(2)O(2) levels and transcription factor activity, aid ERalpha in regulating the expression of estrogen-responsive genes in target cells.


Subject(s)
Estrogen Receptor alpha/physiology , Gene Expression , Thioredoxin-Disulfide Reductase/physiology , Thioredoxins/physiology , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Ethanol/pharmacology , Gene Expression/drug effects , Gene Expression/genetics , Humans , Hydrogen Peroxide/metabolism , Immunohistochemistry , Immunoprecipitation , Oxidative Stress/drug effects , Oxidative Stress/genetics , Protein Binding , RNA Interference , Thioredoxin-Disulfide Reductase/genetics , Thioredoxin-Disulfide Reductase/metabolism , Thioredoxins/genetics , Thioredoxins/metabolism
11.
Mol Endocrinol ; 23(9): 1346-59, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19460860

ABSTRACT

Apurinic/apyrimidinic endonuclease 1 or redox factor-1 (Ape1/Ref-1) is a pleiotropic cellular protein involved in DNA repair and, through its redox activity, enhances the binding of a select group of transcription factors to their cognate recognition sequences in DNA. Thus, we were intrigued when we identified Ape1/Ref-1 and a number of DNA repair and oxidative stress proteins in a complex associated with the DNA-bound estrogen receptor alpha (ERalpha). Because Ape1/Ref-1 interacts with a number of transcription factors and influences their activity, we determined whether it might also influence ERalpha activity. We found that endogenously expressed Ape1/Ref-1 and ERalpha from MCF-7 human breast cancer cells interact and that Ape1/Ref-1 enhances the interaction of ERalpha with estrogen-response elements (EREs) in DNA. More importantly, Ape1/Ref-1 alters expression of the endogenous, estrogen-responsive progesterone receptor and pS2 genes in MCF-7 cells and associates with ERE-containing regions of these genes in native chromatin. Interestingly, knocking down Ape1/Ref-1 expression or inhibiting its redox activity with the small molecule inhibitor E3330 enhances estrogen responsiveness of the progesterone receptor and pS2 genes but does not alter the expression of the constitutively active 36B4 gene. Additionally, the reduced form of Ape1/Ref-1 increases and E3330 limits ERalpha-ERE complex formation in vitro and in native chromatin. Our studies demonstrate that Ape1/Ref-1 mediates its gene-specific effects, in part, by associating with endogenous, estrogen-responsive genes and that the redox activity of Ape1/Ref-1 is instrumental in altering estrogen-responsive gene expression.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Estrogens/metabolism , Gene Expression Regulation, Neoplastic , Receptors, Estrogen/metabolism , Breast/metabolism , Cell Line, Tumor , Chromatin/chemistry , DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , Female , Humans , Models, Biological , Oxidation-Reduction , Oxidative Stress , Response Elements , Transcriptional Activation
12.
BMC Mol Biol ; 9: 97, 2008 Oct 30.
Article in English | MEDLINE | ID: mdl-18973695

ABSTRACT

BACKGROUND: DNA-bound transcription factors recruit an array of coregulatory proteins that influence gene expression. We previously demonstrated that DNA functions as an allosteric modulator of estrogen receptor alpha (ERalpha) conformation, alters the recruitment of regulatory proteins, and influences estrogen-responsive gene expression and reasoned that it would be useful to develop a method of isolating proteins associated with the DNA-bound ERalpha using full-length receptor and endogenously-expressed nuclear proteins. RESULTS: We have developed a novel approach to isolate large complexes of proteins associated with the DNA-bound ERalpha. Purified ERalpha and HeLa nuclear extracts were combined with oligos containing ERalpha binding sites and fractionated on agarose gels. The protein-DNA complexes were isolated and mass spectrometry analysis was used to identify proteins associated with the DNA-bound receptor. Rather than simply identifying individual proteins that interact with ERalpha, we identified interconnected networks of proteins with a variety of enzymatic and catalytic activities that interact not only with ERalpha, but also with each other. Characterization of a number of these proteins has demonstrated that, in addition to their previously identified functions, they also influence ERalpha activity and expression of estrogen-responsive genes. CONCLUSION: The agarose gel fractionation method we have developed would be useful in identifying proteins that interact with DNA-bound transcription factors and should be easily adapted for use with a variety of cultured cell lines, DNA sequences, and transcription factors.


Subject(s)
DNA/metabolism , Electrophoresis, Agar Gel/methods , Estrogen Receptor alpha/metabolism , Proteins/isolation & purification , Proteins/metabolism , Cell Line, Tumor , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/isolation & purification , DNA-Binding Proteins/metabolism , Estrogen Receptor alpha/isolation & purification , Gene Expression Regulation , HeLa Cells , Humans , Protein Binding , Proteins/chemistry
13.
Mol Cell Endocrinol ; 291(1-2): 79-86, 2008 Sep 10.
Article in English | MEDLINE | ID: mdl-18577416

ABSTRACT

The differential recruitment of coregulatory proteins to the DNA-bound estrogen receptor alpha (ERalpha) plays a critical role in mediating estrogen-responsive gene expression. We previously isolated and identified retinoblastoma-associated proteins 46 (RbAp46) and 48 (RbAp48), which are associated with chromatin remodeling, histone deacetylation, and transcription repression, as proteins associated with the DNA-bound ERalpha. We now demonstrate that RbAp46 and RbAp48 interact with ERalphain vitro and in vivo, associate with ERalpha at endogenous, estrogen-responsive genes, and alter expression of endogenous, ERalpha-activated and -repressed genes in MCF-7 breast cancer cells. Our findings reveal that RbAp48 limits expression of estrogen-responsive genes and that RbAp46 modulates estrogen responsiveness in a gene-specific manner. The ability of RbAp46 and RbAp48 to interact with ERalpha and influence its activity reveals yet another role for these multifunctional proteins in regulating gene expression.


Subject(s)
Carrier Proteins/metabolism , Estrogen Receptor alpha/metabolism , Gene Expression Regulation , Nuclear Proteins/metabolism , Breast Neoplasms/metabolism , Carrier Proteins/genetics , Cell Line, Tumor , Estrogen Receptor alpha/genetics , Estrogens/metabolism , Female , Humans , Nuclear Proteins/genetics , Retinoblastoma-Binding Protein 4 , Retinoblastoma-Binding Protein 7 , Transcription, Genetic
14.
Mol Endocrinol ; 22(5): 1113-24, 2008 May.
Article in English | MEDLINE | ID: mdl-18258688

ABSTRACT

The effects of estrogen on gene expression in mammary cells are mediated by interaction of the estrogen receptor (ER) with estrogen response elements in target DNA. Whereas the ER is the primary initiator of transcription, the recruitment of coregulatory proteins to the DNA-bound receptor influences estrogen responsiveness. To better understand how estrogen alters gene expression, we identified proteins associated with the DNA-bound ERalpha. Surprisingly, the antioxidant enzyme Cu/Zn superoxide dismutase (SOD1), which is known primarily as a scavenger of superoxide, was associated with the DNA-bound receptor. We have now demonstrated that SOD1 interacts with ERalpha from MCF-7 cell nuclear extracts and with purified ERalpha and that SOD1 enhances binding of ERalpha to estrogen response element-containing DNA. Although SOD1 decreases transcription of an estrogen-responsive reporter plasmid in transiently transfected U2 osteosarcoma cells, RNA interference assays demonstrate that SOD1 is required for effective estrogen responsiveness of the endogenous pS2, progesterone receptor, cyclin D1, and Cathepsin D genes in MCF-7 breast cancer cells. Furthermore, ERalpha and SOD1 are associated with regions of the pS2 and progesterone receptor genes involved in conferring estrogen-responsive gene expression. Interestingly, when MCF-7 cells are exposed to 17beta-estradiol and superoxide generated by addition of potassium superoxide (KO2) to the cell medium, SOD1 levels are increased and tyrosine nitration, which is an indicator of oxidative stress-induced protein damage, is significantly diminished. Our studies have identified a new role for SOD1 in regulating estrogen-responsive gene expression and suggest that the 17beta-estradiol- and KO2-induced increase in SOD1 may play a role in the survival of breast cancer cells and the progression of mammary tumors.


Subject(s)
Estrogens/pharmacology , Oxidative Stress/drug effects , Superoxide Dismutase/metabolism , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Estrogen Receptor alpha/metabolism , Fluorescent Antibody Technique , Humans , Protein Binding , RNA Interference , RNA, Small Interfering/genetics , Superoxide Dismutase/genetics , Superoxides/pharmacology , Transcription, Genetic/drug effects
15.
Nucleic Acids Res ; 35(15): 5028-38, 2007.
Article in English | MEDLINE | ID: mdl-17636311

ABSTRACT

The ability of estrogen receptor alpha (ERalpha) to modulate gene expression is influenced by the recruitment of a host of co-regulatory proteins to target genes. To further understand how estrogen-responsive genes are regulated, we have isolated and identified proteins associated with ERalpha when it is bound to DNA containing the consensus estrogen response element (ERE). One of the proteins identified in this complex, proliferating cell nuclear antigen (PCNA), is required for DNA replication and repair. We show that PCNA interacts with ERalpha in the absence and in the presence of DNA, enhances the interaction of ERalpha with ERE-containing DNA, and associates with endogenous estrogen-responsive genes. Interestingly, rather than altering hormone responsiveness of endogenous, estrogen-responsive genes, PCNA increases the basal expression of these genes. Our studies suggest that in addition to serving as a platform for the recruitment of DNA replication and repair proteins, PCNA may serve as a platform for transcription factors involved in regulating gene expression.


Subject(s)
Estrogen Receptor alpha/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Cell Line, Tumor , Estradiol/pharmacology , Humans , Proliferating Cell Nuclear Antigen/isolation & purification , Response Elements , Transcription, Genetic/drug effects
16.
Mol Endocrinol ; 21(7): 1569-80, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17488975

ABSTRACT

The ligand-occupied estrogen receptor alpha (ERalpha) initiates changes in gene expression through its interaction with target DNA. The capacity of ERalpha to modulate gene expression is influenced by the association of the receptor with a variety of coregulatory proteins. To further understand the role of these coregulatory proteins in ERalpha-mediated transcription, we have isolated and identified proteins associated with ERalpha when it is bound to the consensus estrogen response element. One of the proteins identified in this complex, flap endonuclease-1 (FEN-1), is required for DNA replication and repair. We show that FEN-1 interacts directly with ERalpha and enhances the interaction of ERalpha with estrogen response element-containing DNA. More importantly, chromatin immunoprecipitation and RNA interference assays demonstrate that endogenously expressed FEN-1 associates with the native pS2 gene in MCF-7 cells and influences estrogen-responsive gene expression. Interestingly, estrogen differentially regulates expression of FEN-1 in mouse uterine epithelial, stromal, and myometrial cells. Together, our studies help to elucidate the functional consequence of the ERalpha-FEN-1 interaction and increase our understanding of the elaborate regulatory mechanisms that drive estrogen-responsive gene expression and DNA repair.


Subject(s)
DNA Repair , Estrogen Receptor alpha/metabolism , Flap Endonucleases/metabolism , Amino Acid Sequence , Animals , Base Sequence , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , DNA, Complementary/genetics , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Female , Flap Endonucleases/genetics , Gene Expression/drug effects , Humans , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Ovariectomy , RNA Interference , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Uterus/drug effects , Uterus/metabolism
17.
Mol Cell Endocrinol ; 267(1-2): 106-15, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17275994

ABSTRACT

We have used a chromatin immunoprecipitation (ChIP)-based cloning strategy to isolate and identify genes associated with estrogen receptor alpha (ERalpha) in MCF-7 human breast cancer cells. One of the gene regions isolated was a 288bp fragment from the ninth intron of the breast cancer 1 associated ring domain (BARD1) gene. We demonstrated that ERalpha associated with this region of the endogenous BARD 1 gene in MCF-7 cells, that ERalpha bound to three of five ERE half sites located in the 288bp BARD1 region, and that this 288bp BARD1 region conferred estrogen responsiveness to a heterologous promoter. Importantly, treatment of MCF-7 cells with estrogen increased BARD1 mRNA and protein levels. These findings demonstrate that ChIP cloning strategies can be utilized to successfully isolate regulatory regions that are far removed from the transcription start site and assist in identifying cis elements involved in conferring estrogen responsiveness.


Subject(s)
Estrogen Receptor alpha/metabolism , Gene Expression Regulation, Neoplastic , Introns/genetics , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Base Pairing/drug effects , Base Pairing/genetics , Base Sequence , Binding Sites/drug effects , Cell Line, Tumor , Chromatin Immunoprecipitation , Deoxyribonuclease I/metabolism , Estradiol/pharmacology , Estrogens/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Molecular Sequence Data , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Response Elements/drug effects , Response Elements/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism
18.
J Steroid Biochem Mol Biol ; 88(2): 113-22, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15084343

ABSTRACT

The progesterone receptor (PR) gene is regulated by estrogen in normal reproductive tissues and in MCF-7 human breast cancer cells. Although it is generally thought that estrogen responsiveness is mediated by interaction of the ligand-occupied estrogen receptor (ER) with estrogen response elements (EREs) in target genes, the human progesterone receptor (PR) gene lacks a palindromic ERE. Promoter A of the PR gene does, however, contain an ERE half site upstream of two adjacent Sp1 sites from +571 to +595, the +571 ERE/Sp1 site. We have examined the individual contributions of the ERE half site and the two Sp1 sites in regulating estrogen responsiveness. Transient transfection assays demonstrated that both Sp1 sites were critical for estrogen-mediated activation of the PR gene. Interestingly, rather than decreasing transcription, mutations in the ERE half site increased transcription substantially suggesting that this site plays a role in limiting transcription. Chromatin immunoprecipitation assays demonstrated that Sp1 was associated with the +571 ERE/Sp1 site in the endogenous PR gene in the absence and in the presence of estrogen, but that ERalpha was only associated with this region of the PR gene after MCF-7 cells had been treated with estrogen. Our studies provide evidence that effective regulation of transcription through the +571 ERE/Sp1 site requires the binding of ERalpha and Sp1 to their respective cis elements and the appropriate interaction of ERalpha and Sp1 with other coregulatory proteins and transcription factors.


Subject(s)
Estrogens/metabolism , Receptors, Progesterone/genetics , Sp1 Transcription Factor/metabolism , Base Sequence , Binding Sites , Cell Line, Tumor , DNA Primers , Estrogen Receptor alpha , Gene Expression Regulation , Humans , Ligands , Plasmids , Precipitin Tests , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Transcription, Genetic
19.
Mol Endocrinol ; 18(3): 521-32, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14684847

ABSTRACT

The progesterone receptor (PR) gene is activated by estrogen in normal reproductive tissues and in MCF-7 human breast cancer cells. Although it is typically thought that estrogen responsiveness is mediated through estrogen response elements (EREs), the human PR gene lacks a palindromic ERE sequence. We have identified an activating protein-1 (AP-1) site at +745 in the human PR gene that bound purified Fos and Jun and formed a complex with Fos/Jun heterodimers present in MCF-7 nuclear extracts. Surprisingly, mutating the +745 AP-1 site in the context of a 1.5-kb region of the PR gene significantly enhanced estrogen receptor (ER) alpha-mediated transactivation, suggesting that the wild-type +745 AP-1 site plays a role in inhibiting PR gene expression in the presence of hormone. In support of this idea, transient transfection assays demonstrated that increasing levels of Fos and Jun repressed transcription of a reporter plasmid containing the +745 AP-1 site. Fos levels were transiently increased, ERalpha levels were decreased, and Jun was dephosphorylated after MCF-7 cells were treated with estrogen. Chromatin immunoprecipitation assays demonstrated that Jun was associated with the +745 AP-1 site in the endogenous PR gene in the presence and in the absence of estrogen, but that ERalpha and Fos were only associated with the +745 AP-1 site after estrogen treatment of MCF-7 cells. Our studies suggest that the human PR gene is regulated by multiple transcription factors and that the differential binding of these dynamically regulated trans-acting factors influences gene expression.


Subject(s)
Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Receptors, Progesterone/genetics , Transcription Factor AP-1/metabolism , Animals , Base Sequence , Binding Sites , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cells, Cultured , Chromatin/genetics , Chromatin/metabolism , Conserved Sequence , Estradiol/pharmacology , Estrogen Receptor alpha , Estrogens/pharmacology , Gene Expression Regulation , Humans , Mice , Mutation , Phosphorylation , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/isolation & purification , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/isolation & purification , Rabbits , Rats , Receptors, Estrogen/drug effects , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Receptors, Progesterone/drug effects , Receptors, Progesterone/metabolism , Response Elements , Transcription, Genetic
20.
Endocrinology ; 143(12): 4583-91, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12446585

ABSTRACT

The progesterone receptor (PR) gene is activated by estrogen in MCF-7 human breast cancer cells. Although the human PR gene does not contain an estrogen response element (ERE), we have identified a putative activating protein-1 (AP-1) site at +90 in the PR gene that was hypersensitive to deoxyribonuclease I cleavage in genomic Southern analysis, bound purified Fos and Jun, formed a complex with Fos/Jun heterodimers present in MCF-7 nuclear extracts in gel mobility shift assays, and functioned as an estrogen-responsive enhancer in transient cotransfection assays. When the +90 AP-1 site was mutated in the context of the PR gene, estrogen responsiveness was significantly decreased. Purified estrogen receptor (ER) enhanced binding of Fos and Jun to the +90 AP-1 site and bound to an adjacent imperfect ERE half-site. Mutating this ERE half-site diminished the binding of ER, Fos, and Jun and decreased transcription. Chromatin immunoprecipitation assays demonstrated that the ER, Fos, and Jun were present at the +90 AP-1 site in the endogenous PR gene only after treatment of MCF-7 cells with estrogen. These studies suggest that the cooperative interaction of the ER with Fos and Jun proteins helps confer estrogen responsiveness to the endogenous PR gene.


Subject(s)
Breast Neoplasms/metabolism , Estrogens/pharmacology , Receptors, Estrogen/physiology , Receptors, Progesterone/genetics , Response Elements , Transcription Factor AP-1/physiology , Binding Sites , Blotting, Southern , Cell Nucleus/chemistry , DNA/metabolism , Deoxyribonuclease I/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha , Humans , Mutagenesis , Promoter Regions, Genetic , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Transcription Factor AP-1/genetics , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...