Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Oncogene ; 34(28): 3676-87, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25241899

ABSTRACT

Progesterone (P4) has emerged as an important hormone-regulating mammary stem cell (MaSC) populations. In breast cancer, P4 and synthetic analogs increase the number of stem-like cells within luminal estrogen receptor (ER)- and progesterone receptor (PR)-positive breast cancers. These cells gain expression of de-differentiated cell markers CD44 and cytokeratin 5 (CK5), lose luminal markers ER and PR, and are more therapy resistant. We previously described that P4 downregulation of microRNA (miR)-29a contributes to the expansion of CD44(high) and CK5(+) cells. Here we investigated P4 downregulation of miR-141, a member of the miR-200 family of tumor suppressors, in facilitating an increase in stem-like breast cancer cells. miR-141 was the sole member of the miR-200 family P4-downregulated at the mature miRNA level in luminal breast cancer cell lines. Stable inhibition of miR-141 alone increased the CD44(high) population, and potentiated P4-mediated increases in both CD44(high) and CK5(+) cells. Loss of miR-141 enhanced both mammosphere formation and tumor initiation. miR-141 directly targeted both PR and signal transducer and activator of transcription 5A (Stat5a), transcription factors important for MaSC expansion. miR-141 depletion increased PR protein levels, even in cell lines where PR expression is estrogen dependent. Stat5a suppression via small interfering RNA or a small-molecule inhibitor reduced the P4-dependent increase in CK5(+) and CD44(high) cells. These data support a mechanism by which P4-triggered loss of miR-141 facilitates breast cancer cell de-differentiation through deregulation of PR and Stat5a, two transcription factors important for controlling mammary cell fate.


Subject(s)
Breast Neoplasms/genetics , MicroRNAs/genetics , Neoplastic Stem Cells/drug effects , Progesterone/pharmacology , Progestins/pharmacology , STAT5 Transcription Factor/genetics , Tumor Suppressor Proteins/genetics , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Down-Regulation/drug effects , Female , Humans , Hyaluronan Receptors/metabolism , Keratin-5/metabolism , Mice , Neoplasm Transplantation , Neoplastic Stem Cells/pathology , Receptors, Progesterone
2.
Oncogene ; 32(20): 2555-64, 2013 May 16.
Article in English | MEDLINE | ID: mdl-22751119

ABSTRACT

The female hormone progesterone (P4) promotes the expansion of stem-like cancer cells in estrogen receptor (ER)- and progesterone receptor (PR)-positive breast tumors. The expanded tumor cells lose expression of ER and PR, express the tumor-initiating marker CD44, the progenitor marker cytokeratin 5 (CK5) and are more resistant to standard endocrine and chemotherapies. The mechanisms underlying this hormone-stimulated reprogramming have remained largely unknown. In the present study, we investigated the role of microRNAs in progestin-mediated expansion of this dedifferentiated tumor cell population. We demonstrate that P4 rapidly downregulates miR-29 family members, particularly in the CD44(+) cell population. Downregulation of miR-29 members potentiates the expansion of CK5(+) and CD44(+) cells in response to progestins, and results in increased stem-like properties in vitro and in vivo. We demonstrate that miR-29 directly targets Krüppel-like factor 4 (KLF4), a transcription factor required for the reprogramming of differentiated cells to pluripotent stem cells, and for the maintenance of breast cancer stem cells. These results reveal a novel mechanism, whereby progestins increase the stem cell-like population in hormone-responsive breast cancers, by decreasing miR-29 to augment PR-mediated upregulation of KLF4. Elucidating the mechanisms whereby hormones mediate the expansion of stem-like cells furthers our understanding of the progression of hormone-responsive breast cancers.


Subject(s)
Breast Neoplasms/genetics , Cell Differentiation/genetics , Kruppel-Like Transcription Factors/genetics , MicroRNAs/genetics , Progestins/pharmacology , 3' Untranslated Regions , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Differentiation/drug effects , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hyaluronan Receptors/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, SCID , MicroRNAs/metabolism , Progesterone/pharmacology , Up-Regulation , Xenograft Model Antitumor Assays
3.
Br J Cancer ; 95(9): 1220-8, 2006 Nov 06.
Article in English | MEDLINE | ID: mdl-17043687

ABSTRACT

Activation of the type I insulin-like growth factor receptor (IGF-IR) regulates several aspects of the malignant phenotype, including cancer cell proliferation and metastasis. Phosphorylation of adaptor proteins downstream of IGF-IR may couple IGF action to specific cancer phenotypes. In this study, we sought to determine if insulin receptor substrate-1 and -2 (IRS-1 and -2) mediate distinct biological effects in breast cancer cells. Insulin receptor substrate-1 and IRS-2 were expressed in T47D-YA breast cancer cells, which lack IRS-1 and -2 expression, yet retain functional IGF-IR. In the absence of IRS-1 and -2 expression, IGF-IR activation was unable to stimulate proliferation or motility in T47D-YA cells. Expression of IRS-1 resulted in IGF-I-stimulated proliferation, but did not affect motility. In contrast, expression of IRS-2 enhanced IGF-I-stimulated motility, but did not stimulate proliferation. The alphaIR-3, an inhibitor of the IGF-IR, was unable to affect these IGF-stimulated phenotypes unless IRS-1 or -2 was expressed. Thus, IGF-IR alone is unable to regulate important breast cancer cell phenotypes. In these cells, IRS proteins are required for and mediate distinct aspects of IGF-IR-stimulated behaviour. As multiple agents targeting the IGF-IR are currently in early clinical trials, IRS expression should be considered as a potential biomarker for IGF-IR responsiveness.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Phosphoproteins/physiology , Receptor, IGF Type 1/physiology , Antibodies, Monoclonal/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/physiopathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Gene Expression/drug effects , Humans , Insulin Receptor Substrate Proteins , Insulin-Like Growth Factor I/pharmacology , Intracellular Signaling Peptides and Proteins/genetics , Phosphoproteins/genetics , RNA Interference , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/immunology , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/physiology , Transfection
4.
Breast Cancer Res Treat ; 94(2): 171-83, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16175315

ABSTRACT

Progesterone in hormone replacement therapy (HRT) preparations increases, while hysterectomy greatly reduces, the incidence of breast cancer. Cross-talk between the progesterone and growth factor signaling pathways occurs at multiple levels and this maybe a key factor in breast cancer survival and progression. To test this hypothesis, we characterized the effect of progesterone pre-treatment on the sensitization of the epidermal growth factor (EGF) signaling pathway to EGF in the breast cancer cell line ZR-75. For the first time in ZR-75 cells and in agreement with previous work using synthetic progestins, we demonstrate that pre-treatment with the natural ligand progesterone increases EGF receptor (EGFR) levels and subsequent ligand-dependent phosphorylation. Downstream we demonstrate that progesterone alone increases erk-1 + 2 phosphorylation, potentiates EGF-phosphorylated erk-1 + 2 and maintains these levels elevated for 24 h; over 20 h longer than in vehicle treated cells. Additionally, progesterone increased the levels of STAT5, another component of the EGF signaling cascade. Progesterone increased EGF mediated transcription of a c-fos promoter reporter and the nuclear localization of the native c-fos protein. Furthermore, progesterone and EGF both alone and in combination, significantly increase cell proliferation. Several results presented herein demonstrate the conformity between the action of the natural ligand progesterone with that of synthetic progestins such as MPA and R5020 and allows the postulation that the progestin/progesterone-dependent increase of EGF signaling provides a survival advantage to burgeoning cancer cells and may contribute to the breast cancer risk associated with endogenous progesterone and with progestin-containing HRT.


Subject(s)
Breast Neoplasms/metabolism , Epidermal Growth Factor/drug effects , ErbB Receptors/metabolism , Progesterone/pharmacology , Breast Neoplasms/pathology , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic , Humans , Phosphorylation/drug effects , Progesterone/administration & dosage , Signal Transduction/drug effects
5.
Steroids ; 65(10-11): 579-84, 2000.
Article in English | MEDLINE | ID: mdl-11108862

ABSTRACT

The development of tamoxifen resistance and consequent disease progression are common occurrences in breast cancers, often despite the continuing expression of estrogen receptors (ER). Tamoxifen is a mixed antagonist, having both agonist and antagonist properties. We have suggested that the development of tamoxifen resistance is associated with an increase in its agonist-like properties, resulting in loss of antagonist effects or even inappropriate tumor stimulation. Nuclear receptor function is influenced by a family of transcriptional coregulators, that either enhance or suppress transcriptional activity. Using a mixed antagonist-biased two-hybrid screening strategy, we identified two such proteins: the human homolog of the nuclear receptor corepressor, N-CoR, and a novel coactivator, L7/SPA (Switch Protein for Antagonists). In transcriptional studies, N-CoR suppressed the agonist properties of tamoxifen and RU486, and L7/SPA increased agonist effects. We speculated that the relative levels of these coactivators and corepressors may determine the balance of agonist and antagonist properties of mixed antagonists, such as tamoxifen. Using quantitative RT-PCR, we, therefore, measured the levels of transcripts encoding these coregulators, as well as the corepressor SMRT, and the coactivator SRC-1, in a small cohort of tamoxifen-resistant and sensitive breast tumors. The results suggest that tumor sensitivity to mixed antagonists may be governed by a complex set of transcription factors, which we are only now beginning to understand.


Subject(s)
Breast Neoplasms/drug therapy , Receptors, Estrogen/genetics , Tamoxifen/pharmacology , Breast Neoplasms/pathology , Drug Resistance , Female , Humans , Nuclear Proteins/genetics , Nuclear Proteins/pharmacology , Nuclear Receptor Co-Repressor 1 , Receptors, Estrogen/chemistry , Repressor Proteins/genetics , Repressor Proteins/pharmacology , Transcription Factors/genetics , Transcription Factors/pharmacology
6.
J Mol Endocrinol ; 24(2): 165-82, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10750018

ABSTRACT

Ligand-activated progesterone receptors (PR) bind to DNA at specific progesterone response elements by means of a DNA binding domain (DBD(PR)) containing two highly conserved zinc fingers. DNA-bound PRs regulate transcription via interaction with other nuclear proteins and transcription factors. We have now identified four HeLa cell nuclear proteins that copurify with a glutathionine-S-transferase-human DBD(PR )fusion protein. Microsequence and immunoblot analyses identified one of these proteins as the 113 kDa poly(ADP-ribose) polymerase. The three other proteins were identified as subunits of the DNA-dependent protein kinase (DNA-PK) holoenzyme: its DNA binding regulatory heterodimers consisting of Ku70 and Ku86, and the 460 kDa catalytic subunit, DNA-PK(CS). DNA-PK that was 'pulled-down' by DBD(PR) on the affinity resin was able to (1) autophosphorylate Ku70, Ku86, and DNA-PK(CS), (2) transphosphorylate DBD(PR), and (3) phosphorylate a DNA-PK-specific p53 peptide substrate. DNA-PK was also able to associate with the DBD of the yeast activator GAL4. However, neither a PR DBD mutant lacking a structured first zinc finger (DBD(CYS)) nor the core DBD of the estrogen receptor (DBD(ER)) copurified DNA-PK, suggesting the interaction is not non-specific for DBDs. Lastly, we found that DNA-PK copurified with full-length human PR transiently expressed in HeLa cells, suggesting that the human PR/DNA-PK complex can assemble in vivo. These data show that DNA-PK and DBD(PR) interact, that DBD(PR) is a phosphorylation substrate of DNA-PK, and suggest a potential role for DNA-PK in PR-mediated transcription.


Subject(s)
Antigens, Nuclear , DNA Helicases , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Progesterone/metabolism , Saccharomyces cerevisiae Proteins , Amino Acid Sequence , Autoantigens/isolation & purification , Autoantigens/metabolism , Binding Sites , DNA-Activated Protein Kinase , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/isolation & purification , Glutathione Transferase/genetics , HeLa Cells , Humans , Ku Autoantigen , Ligands , Methionine/metabolism , Molecular Sequence Data , Phosphorylation , Poly(ADP-ribose) Polymerases/chemistry , Poly(ADP-ribose) Polymerases/isolation & purification , Protein Biosynthesis , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/isolation & purification , Receptors, Progesterone/chemistry , Receptors, Progesterone/isolation & purification , Recombinant Fusion Proteins/metabolism , Transcription Factors/metabolism
7.
J Steroid Biochem Mol Biol ; 74(5): 255-9, 2000 Nov 30.
Article in English | MEDLINE | ID: mdl-11162933

ABSTRACT

The antiestrogen tamoxifen is an effective treatment for estrogen receptor positive breast cancers, slowing tumor growth and preventing disease recurrence, with relatively few side effects. However, many patients who initially respond to treatment, later become resistant to treatment. Tamoxifen has both agonist and antagonist activities, which are manifested in a tissue-specific pattern. Development of tamoxifen resistance can be characterized by an increase in the partial agonist properties of the antiestrogen in the breast, resulting in loss of growth inhibition and even inappropriate tumor stimulation. Nuclear receptor function is modulated by transcriptional coregulators, which either enhance or repress receptor activity. Using a mixed antagonist-biased two-hybrid screening strategy, we identified two such proteins: the human homolog of the nuclear receptor corepressor, N-CoR, and a novel coactivator, L7/SPA (Switch Protein for Antagonists). In transcriptional studies N-CoR suppressed the agonist properties of tamoxifen and RU486, while L7/SPA increased agonist effects. We speculated that the relative level of these coactivators and corepressors might determine the balance of agonist and antagonist properties of mixed antagonists such as tamoxifen. Using quantitative RT-PCR we therefore measured the levels of transcripts encoding these coregulators, as well as the corepressor SMRT, and the coactivator SRC-1, in a small cohort of tamoxifen resistant and sensitive breast tumors. The results suggest that tumor sensitivity to mixed antagonists may be governed by a complex set of transcription factors, which we are only now beginning to understand.


Subject(s)
Breast Neoplasms/metabolism , Drug Resistance, Neoplasm , Estrogen Receptor Modulators/pharmacology , Tamoxifen/pharmacology , Transcription Factors/metabolism , Animals , Breast Neoplasms/drug therapy , Cell Division/drug effects , DNA-Binding Proteins/metabolism , Estrogen Receptor Modulators/therapeutic use , Female , Histone Acetyltransferases , Humans , Mifepristone/pharmacology , Nuclear Receptor Co-Repressor 2 , Nuclear Receptor Coactivator 1 , Receptors, Estrogen/agonists , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism , Receptors, Progesterone/antagonists & inhibitors , Receptors, Progesterone/metabolism , Repressor Proteins/metabolism , Tamoxifen/therapeutic use
9.
Mol Endocrinol ; 13(6): 829-36, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10379882

ABSTRACT

In the breast, data from numerous laboratories suggest that cross-talk exists between PR and growth factor and cytokine signaling pathways at multiple levels (Fig. 4). At the cell surface (level 1), progestins up-regulate growth factor and cytokine receptors. We have expanded this observation by examining the effects of progestins in the cytoplasm (level 2) where progestins regulate several intracellular effectors by increasing the levels and altering the subcellular compartmentalization of Stat5, increasing the association of Stat5 with phosphotyrosine-containing proteins and tyrosine phosphorylation of JAK2, Cbl, and Shc, and potentiating EGF-stimulated p42/p44 MAPKs, p38 MAP kinase, and JNK activities. Together, these events lead to sensitization of downstream signaling pathways to the actions of locally acting secondary factors. Finally, inside the nucleus (level 3), agonist-occupied PR synergize with nuclear transcription factors that are growth-factor regulated, to control the activity of key genes involved in breast cell fate (Figs. 1 and 4). We speculate that after progesterone treatment, orchestrated combinations of steroid hormones and growth factors or cytokines can fine tune the timing and degree of expression of a subset of genes that determine whether progestin-primed cells undergo proliferation, differentiation, or programmed cell death. The paradoxical effects of progesterone have presented a longstanding conundrum to the scientist and clinician. Why are physiological levels of progesterone proliferative in the breast but antiproliferative and protective in the uterus? If progesterone is proliferative in the breast, why is high-dose progestin therapy successful in treating breast cancer? Our intent here has been to open a dialogue addressing these questions. Our data and that of others are beginning to show that one cannot approach the question of progestin actions in isolation. Other important regulatory proteins, whose expression may vary in tissue-specific ways, work in concert with progesterone to decide cell fate. The timing and dose of progesterone may also influence the biological response. Since progestins are widely used in oral contraception, in hormone replacement therapy, and in cancer treatments, it is becoming critically important that the subtleties of their mechanisms of action be clearly understood.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Milk Proteins , Progesterone/metabolism , Signal Transduction , Animals , Breast Neoplasms/drug therapy , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Cycle/drug effects , Cell Division , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Cyclins/metabolism , Cytokines/metabolism , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Female , Gene Expression Regulation, Neoplastic , Growth Substances/metabolism , Humans , Progesterone/pharmacology , STAT5 Transcription Factor , Trans-Activators/metabolism , Transcription Factors/metabolism
10.
Circulation ; 99(20): 2688-93, 1999 May 25.
Article in English | MEDLINE | ID: mdl-10338464

ABSTRACT

BACKGROUND: Estrogens stimulate growth of breast or uterine cells but have the opposite effect on vascular smooth muscle cells, in which they protect against coronary artery disease with or without concomitant administration of progesterone. A possible cause of differences in hormone action is variable tissue-specific expression of hormone receptor. Therefore, we analyzed the structure of estrogen receptors (ERs) and progesterone receptors (PRs) in human vascular smooth muscle. METHODS AND RESULTS: RNA was isolated from human vascular smooth muscle, and the functional domains of ER-alpha and PR were characterized by reverse transcriptase and polymerase chain reaction. Interestingly, in addition to wild-type ER-alpha and PR, 5 variant ER-alpha and 2 variant PR transcripts were found. These variants contained precise deletions of exons encoding regions of the hormone-binding domain. The PR transcripts lacked exon 4 (PRDelta4) and exon 6 (PRDelta6). The ER-alpha transcripts were missing exon 4 (ERDelta4), exon 5 (ERDelta5), exon 6 (ERDelta6), exon 7 (ERDelta7), and exons 6 and 7, (ERDelta6,7). ER-beta variants were also detected. The PR variants were functionally characterized, and PRDelta6 was found to be a dominant-negative transcription inhibitor of wild-type receptors. Variant PR was present in premenopausal women but absent in postmenopausal women. CONCLUSIONS: Variant PR and ER transcripts are extensively expressed in human vascular smooth muscle. The complex tissue-specific effects of sex hormones may be mediated by the expression of heterogeneous forms of their cognate receptors. The presence of variant ERs and PRs may be of importance in altering the physiological effects of estrogens or progestins in vascular smooth muscle.


Subject(s)
Genetic Variation , Muscle, Smooth, Vascular/metabolism , RNA, Messenger/metabolism , Receptors, Estrogen/genetics , Receptors, Progesterone/genetics , Adult , Aged , Culture Techniques , Estrogen Receptor beta , Exons/genetics , Female , Gene Deletion , Genetic Variation/physiology , Humans , Male , Middle Aged , Postmenopause/metabolism , Premenopause/metabolism , Reverse Transcriptase Polymerase Chain Reaction
11.
J Biol Chem ; 273(47): 31308-16, 1998 Nov 20.
Article in English | MEDLINE | ID: mdl-9813039

ABSTRACT

During late stages of breast cancer progression, tumors frequently acquire steroid hormone resistance with concurrent amplification of growth factor receptors; this alteration predicts a poor prognosis. We show here that following treatment with the progestin, R5020, breast cancer cells undergo a "biochemical shift" in the regulation of epidermal growth factor (EGF)-stimulated signaling pathways: R5020 potentiates the effects of EGF by up-regulating EGFR, c-ErbB2 and c-ErbB3 receptors, and by enhancing EGF-stimulated tyrosine phosphorylation of signaling molecules known to associate with activated type I receptors. Independently of EGF, R5020 increases Stat5 protein levels, association of Stat5 with phosphotyrosine-containing proteins, and tyrosine phosphorylation of JAK2 and Shc. Furthermore, progestins "prime" breast cancer cells for growth signals by potentiating EGF-stimulated p42/p44 mitogen-activated protein kinase (MAPK), p38 MAP kinase, and JNK activities. Although the levels of cyclin D1, cyclin E, and p21(WAF1), are up-regulated by R5020 alone, they are synergistically up-regulated by EGF in the presence of R5020. Up-regulation of cell cycle proteins by EGF is blocked by inhibition of p42/p44 MAPK only in the presence of R5020, supporting a shift in the regulation of these cell cycle mediators from MAPK-independent to MAPK-dependent pathways. In summary, progesterone selectively increases the sensitivity of key kinase cascades to growth factors, thereby priming cells for stimulation by latent growth signals. These data support a model in which breast cancer cell growth switches from steroid hormone to growth factor dependence.


Subject(s)
Breast Neoplasms/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Epidermal Growth Factor/metabolism , Mitogen-Activated Protein Kinases , Progesterone/metabolism , Receptor Cross-Talk , Cell Cycle/physiology , Cell Line , Female , Humans , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Models, Biological , Phosphotyrosine/metabolism , Promegestone/pharmacology , Receptors, Growth Factor/biosynthesis , Signal Transduction , Up-Regulation , p38 Mitogen-Activated Protein Kinases
12.
J Biol Chem ; 273(47): 31317-26, 1998 Nov 20.
Article in English | MEDLINE | ID: mdl-9813040

ABSTRACT

STATS (signal transducers and activators of transcription) are latent transcription factors activated in the cytoplasm by diverse cell surface signaling molecules. Like progesterone receptors (PR), Stat5a and 5b are required for normal mammary gland growth and differentiation. These two proteins are up-regulated during pregnancy, a period dominated by high levels of progesterone. We now show that progestin treatment of breast cancer cells regulates Stat5a and 5b, Stat3, and Stat1 protein levels in a PR-dependent manner. In addition, progestin treatment induces translocation of Stat5 into the nucleus, possibly mediated by the association of PR and Stat5. Last, progesterone pretreatment enhances the phosphorylation of Stat5 on tyrosine 694 induced by epidermal growth factor. Functional data show that progestin pretreatment of breast cancer cells enhances the ability of prolactin to stimulate the transcriptional activity of Stat5 on a beta-casein promoter. Progesterone and epidermal growth factor synergize to control transcription from p21(WAF1) and c-fos promoters. These data demonstrate the convergence of progesterone and growth factor/cytokine signaling pathways at multiple levels, and suggest a mechanism for coordination of PR and Stat5-mediated proliferative and differentiative events in the mammary gland.


Subject(s)
Breast Neoplasms/metabolism , Epidermal Growth Factor/pharmacology , Milk Proteins , Progesterone/pharmacology , Prolactin/pharmacology , Receptors, Progesterone/metabolism , Transcriptional Activation , Biological Transport , Cell Nucleus , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , DNA-Binding Proteins/metabolism , Drug Synergism , Female , Humans , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-fos/genetics , STAT1 Transcription Factor , STAT3 Transcription Factor , STAT5 Transcription Factor , Signal Transduction , Trans-Activators/metabolism , Tumor Suppressor Proteins
13.
J Biol Chem ; 273(17): 10696-701, 1998 Apr 24.
Article in English | MEDLINE | ID: mdl-9553133

ABSTRACT

Progesterone has biphasic effects on proliferation of breast cancer cells; it stimulates growth in the first cell cycle, then arrests cells at G1/S of the second cycle accompanied by up-regulation of the cyclin-dependent kinase inhibitor, p21. We now show that progesterone regulates transcription of the p21 promoter by an unusual mechanism. This promoter lacks a canonical progesterone response element. Instead, progesterone receptors (PRs) interact with the promoter through the transcription factor Sp1 at the third and fourth of six Sp1 binding sites located downstream of nucleotide 154. Mutation of Sp1 site 3 eliminates basal transcription, and mutation of sites 3 and 4 eliminates transcriptional up-regulation by progesterone. Progesterone-mediated transcription is further prevented by overexpression of E1A, suggesting that CBP/p300 is required. Indeed, in HeLa cells, Sp1 and CBP/p300 associate with stably integrated flag-tagged PRs in a multiprotein complex. Since many signals converge on p21, cross-talk between PRs and other factors co-localized on the p21 promoter, may explain how progesterone can be either proliferative or differentiative in different target cells.


Subject(s)
Cyclins/genetics , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Progesterone/pharmacology , Trans-Activators , Transcription, Genetic/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Humans , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Protein Binding , Receptors, Progesterone/metabolism , Signal Transduction , Sp1 Transcription Factor/metabolism , Transcription Factors/metabolism , Tumor Cells, Cultured
14.
Breast Cancer Res Treat ; 48(3): 231-41, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9598870

ABSTRACT

Progesterone, through its nuclear receptors (PR), regulates the development and growth of breast cancers. PR also serve as markers of hormone dependence and prognosis in patients with this disease, and functional PR are required to mediate the antiproliferative effects of progestin therapies. We find that normal and malignant breast cells and tissues can express anomalous forms of PR transcripts. We have isolated four variant PR mRNAs that contain precise deletions of exons encoding sections of the DNA- and hormone-binding domains. The transcripts lack exon 2 (PRdelta2), exon 4 (PRdelta4), exon 6 (PRdelta6), or exons 5 and 6 (PRdelta5,6). On immunoblots, PRdelta4, delta6. and delta5, 6 cloned into the background of the PR A-isoform comigrate with similar proteins present in breast tumor extracts; delta6 and delta5, 6 are dominant-negative transcriptional inhibitors of wild-type A- and B-receptors. We propose that expression of variant PR can compromise the accuracy of receptor measurements as markers of hormone-dependent cancers, and can modify the responses of tumors to progestin therapies.


Subject(s)
Breast Neoplasms/metabolism , Receptors, Progesterone/metabolism , Breast/drug effects , Breast/metabolism , Breast Neoplasms/drug therapy , Drug Resistance , Exons , Female , Humans , Progestins/therapeutic use , RNA, Messenger/analysis , Receptors, Progesterone/drug effects , Receptors, Progesterone/genetics , Tumor Cells, Cultured
15.
J Clin Endocrinol Metab ; 82(9): 2956-61, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9284726

ABSTRACT

Progesterone is a key developmental, proliferative, and differentiative hormone in the breast and endometrium, and it can accelerate carcinogenesis in the mammary gland epithelium. In the breast and uterus, progesterone acts through two coexpressed isoforms of progesterone receptors, the B- and A-receptors. To study the function of each isoform in isolation, we previously constructed two breast cancer cell lines that stably and independently express either B-receptors (YB cells) or A-receptors (YA cells). In the present study, YA or YB cells were left untreated, or were treated with the synthetic progestin R5020, and the messages present in each cell line under the two conditions were analyzed by differential display. Two message species are described that are regulated only by B-receptors. One of these is regulated in a ligand-independent manner. A third set of messages, encoding flavin-containing monooxygenase 5 (FMO5), was induced by R5020 only in YB cells. A-receptors appear to be inhibitory. FMOs are involved in the metabolic activation of drugs and xenobiotic compounds, including the antiestrogen tamoxifen, to carcinogenic intermediates. It is possible, therefore, that by upregulating the levels of FMO5, progesterone enhances the carcinogenicity of tamoxifen in target tissues that overexpress progesterone B-receptors.


Subject(s)
Carcinogens , Oxygenases/physiology , Progesterone/metabolism , Receptors, Progesterone/physiology , Tamoxifen/adverse effects , Breast Neoplasms/pathology , Breast Neoplasms/physiopathology , Isomerism , Promegestone/pharmacology , Tumor Cells, Cultured
16.
Mol Endocrinol ; 11(6): 693-705, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9171233

ABSTRACT

Steroid receptor antagonists, such as the antiestrogen tamoxifen or the antiprogestin RU486, can have inappropriate agonist-like effects in tissues and tumors. To explain this paradox we postulated that coactivators are inadvertently brought to the promoters of DNA-bound, antagonist-occupied receptors. The human (h) progesterone receptor (PR) hinge-hormone binding domain (H-HBD) was used as bait in a two-hybrid screen of a HeLa cDNA library, in which the yeast cells were treated with RU486. We have isolated and characterized two interesting steroid receptor-interacting proteins that regulate transcription in opposite directions. The first is L7/SPA, a previously described 27-kDa protein containing a basic region leucine zipper domain, having no known nuclear function. When coexpressed with tamoxifen-occupied estrogen receptors (hER) or RU486-occupied hPR or glucocorticoid receptors (hGR), L7/SPA increases the partial agonist activity of the antagonists by 3- to 10-fold, but it has no effect on agonist-mediated transcription. The interaction of L7/SPA with hPR maps to the hinge region, and indeed, the hPR hinge region squelches L7/SPA-dependent induction of antagonist-mediated transcription. Interestingly, pure antagonists that lack partial agonist effects, such as the antiestrogen ICI164,384 or the antiprogestin ZK98299, cannot be up-regulated by L7/SPA. We also isolated, cloned, and sequenced the human homolog (hN-CoR) of the 270-kDa mouse (m) thyroid/retinoic acid receptor corepressor. Binding of hN-CoR maps to the hPR-HBD. mN-CoR, and a related human corepressor, SMRT, suppress RU486 or tamoxifen-mediated partial agonist activity by more than 90%. This suppression is completely squelched by overexpression of the hPR H-HBD. Additionally, both corepressors reverse the antagonist-dependent transcriptional up-regulation produced by L7/SPA. Our data suggest that the direction of transcription by antagonist-occupied steroid receptors can be controlled by the ratio of coactivators to corepressors recruited to the transcription complex by promoter-bound receptors. In normal tissues and in hormone-resistant breast cancers in which the agonist activity of mixed antagonists predominates, steroid receptors may be preferentially bound by coactivators. This suggests a strategy by which such partial agonist activity can be eliminated and by which candidate receptor ligands can be screened for this activity.


Subject(s)
DNA-Binding Proteins/physiology , Mifepristone/pharmacology , Nuclear Proteins/physiology , Receptors, Progesterone/agonists , Receptors, Progesterone/antagonists & inhibitors , Repressor Proteins/physiology , Ribosomal Proteins/physiology , Animals , Base Sequence , Binding Sites , COS Cells , DNA, Complementary , DNA-Binding Proteins/genetics , Gene Expression Regulation/drug effects , HeLa Cells , Humans , Leucine Zippers , Mice , Mifepristone/antagonists & inhibitors , Molecular Sequence Data , Nuclear Proteins/genetics , Nuclear Receptor Co-Repressor 1 , Nuclear Receptor Co-Repressor 2 , Receptors, Progesterone/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Repressor Proteins/genetics , Ribosomal Proteins/isolation & purification , Yeasts
17.
Mol Endocrinol ; 10(10): 1167-77, 1996 Oct.
Article in English | MEDLINE | ID: mdl-9121485

ABSTRACT

The nuclear receptors belong to a superfamily of proteins, many of which are ligand-regulated, that bind to specific DNA sequences and control specific gene transcription. Recent data show that, in addition to contacting the basal transcription machinery directly, nuclear receptors inhibit or enhance transcription by recruiting an array of coactivator or corepressor proteins to the transcription complex. In this review we define the properties of these putative coregulatory factors; we describe the basal and coregulatory factors that are currently known to interact with nuclear receptors; we suggest various mechanisms by which coactivators and corepressors act; we discuss issues that are raised by the presence of multiple, perhaps competing, coregulatory factors; and we speculate how these additional regulatory layers may explain the heterogeneity of hormone responses that are observed in normal and malignant tissues.


Subject(s)
Receptors, Cytoplasmic and Nuclear , Signal Transduction , Animals , Humans
18.
Exp Parasitol ; 76(3): 221-31, 1993 May.
Article in English | MEDLINE | ID: mdl-8500582

ABSTRACT

D. immitis third-stage larvae (L3) were cultured with fluoromethyl ketone cysteine protease inhibitors. By Day 5 in culture, none of the larvae cultured with 0.1, 0.2, 0.6, or 1.0 mM benzyloxycarbonyl-Phe-Ala-CH2F (Z-Phe-Ala-CH2F) has molted, while 63.2% of larvae in media without inhibitor had molted. At the two lower concentrations of inhibitor more larvae had initiated, but not completed, the molt. In addition to Z-Phe-Ala-CH2F, four other fluoromethyl ketone derivatives, Z-Phe-Arg-CH2F, amorpholine urea-(Mu)-Leu-Phe-CH2F, Mu-Tyr-Phe-CH2F, and Mu-Phe-Phe-CH2F, were tested to determine their effects on L3 in culture. All fluoromethyl ketones tested except Z-Phe-Arg-CH2F inhibited molting. Larvae cultured in inhibitors were determined to be alive as judged qualitatively by motility and quantitatively by reduction of 3-(4,5-diethylthiazol-2-yl)-2,5-diphenyltetrazolium. Electron microscopy demonstrated that L3 which were unable to molt after being cultured in a fluoromethyl ketone derivative had synthesized the new fourth-stage (L4) cuticle but had not shed the L3 cuticle. The same fluoromethyl ketone derivative that did not inhibit molting, Z-Phe-Arg-CH2F, was a slightly less effective inhibitor of larval extract-initiated hydrolysis of the synthetic peptide substrate, Z-Val-Leu-Arg-7-amino-4-methyl coumarin. L3 were also cultured through the molt in media containing the synthetic peptide substrate Z-Val-Leu-Arg-4- methoxy-B-naphthylamide to examine cysteine protease activity in situ. Fluorescence as seen on Days 0-4 during the molting process was first observed on the anterior tip of the larvae, and subsequently in the pharynx, with progression down the L4 as it shed the L3 cuticle.


Subject(s)
Cysteine Proteinase Inhibitors/pharmacology , Dirofilaria immitis/drug effects , Ketones/pharmacology , Analysis of Variance , Animals , Dipeptides/pharmacology , Dirofilaria immitis/enzymology , Dirofilaria immitis/physiology , Dirofilaria immitis/ultrastructure , Larva/drug effects , Larva/enzymology , Larva/physiology , Larva/ultrastructure , Microscopy, Electron
19.
Exp Parasitol ; 75(2): 213-22, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1516669

ABSTRACT

A model of cutaneous extracellular matrix was used to determine if live Dirofilaria immitis larvae secrete proteases which are active at physiological pH and capable of degrading macromolecules found in cutaneous tissue. After 72 hr, 100 third-stage larvae (L3) degraded 24% of the total matrix, while fourth-stage larvae (L4) degraded 10%. A sharp increase in the amount of matrix degraded by L3 corresponded with the onset of the molting process. L3 and L4 degraded comparable amounts of the glycoprotein and elastin components of the matrix, but molting L3 degraded nearly twice the amount of the collagen component (62% vs 35%). Characterization of proteases present in larval-soluble extracts and excretory-secretory products using synthetic substrates and protease inhibitors demonstrated cysteine-protease and metalloprotease activity. Cysteine protease activity was found in whole worm extracts of both L3 and L4. Metalloprotease was secreted at higher levels by molting L3, but was also secreted by L4. Partial separation of the metalloprotease by size-exclusion chromatography indicated that the molecular weight of the native enzyme was in the 49-54 kDa range. The cysteine protease activity was demonstrated in fractions corresponding to 34-39 kDa. The biological function of the D. immitis larval proteases remains to be conclusively determined; however, these data suggest that they are involved in degradation of components of cutaneous tissue and in the molting process.


Subject(s)
Cysteine Endopeptidases/metabolism , Dirofilaria immitis/enzymology , Metalloendopeptidases/metabolism , Amino Acid Sequence , Animals , Chromatography , Collagen/metabolism , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/isolation & purification , Dirofilaria immitis/growth & development , Elastin/metabolism , Extracellular Matrix/metabolism , Glycoproteins/metabolism , Hydrolysis , Larva/enzymology , Metalloendopeptidases/chemistry , Metalloendopeptidases/isolation & purification , Molecular Sequence Data , Oligopeptides/metabolism , Protease Inhibitors/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...