Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters











Publication year range
1.
Tumour Biol ; 44(1): 187-203, 2022.
Article in English | MEDLINE | ID: mdl-36093651

ABSTRACT

BACKGROUND: The alternative NF-κB pathway is activated by the NF-κB-inducing kinase (NIK) mediated phosphorylation of the inhibitor of κ-B kinase α (IKKα). IKKα then phosphorylates p100/NFKB2 to result in its processing to the active p52 subunit. Evidence suggests that basal breast cancers originate within a subpopulation of luminal progenitor cells which is expanded by signaling to IKKα. OBJECTIVE: To determine the role of IKKα in the development of basal tumors. METHODS: Kinase dead IkkαAA/AA mice were crossed with the C3(1)-TAg mouse model of basal mammary cancer. Tumor growth and tumor numbers in WT and IkkαAA/AA mice were assessed and immunopathology, p52 expression and stem/progenitor 3D colony forming assays were performed. Nik-/- mammary glands were isolated and mammary colonies were characterized. RESULTS: While tumor growth was slower than in WT mice, IkkαAA/AA tumor numbers and pathology were indistinguishable from WT tumors. Both WT and IkkαAA/AA tumors expressed p52 except those IkkαAA/AA tumors where NIK, IKKαAA/AA and ErbB2 were undetectable. Colonies formed by WT and IkkαAA/AA mammary cells were nearly all luminal/acinar however, colony numbers and sizes derived from IkkαAA/AA cells were reduced. In contrast to IkkαAA/AA mice, virgin Nik-/- mammary glands were poorly developed and colonies were primarily derived from undifferentiated bipotent progenitor cells. CONCLUSIONS: C3(1)-TAg induced mammary tumors express p100/p52 even without functional IKKα. Therefore the development of basal-like mammary cancer does not strictly rely on IKKα activation. Signal-induced stabilization of NIK may be sufficient to mediate processing of p100NFKB2 which can then support basal-like mammary tumor formation. Lastly, in contrast to the pregnancy specific role of IKKα in lobuloalveogenesis, NIK is obligatory for normal mammary gland development.


Subject(s)
I-kappa B Kinase , Mammary Neoplasms, Animal , Animals , Female , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Mammary Neoplasms, Animal/genetics , Mice , NF-kappa B/metabolism , Phosphorylation , Pregnancy , Signal Transduction
2.
Breast Cancer Res ; 24(1): 40, 2022 06 09.
Article in English | MEDLINE | ID: mdl-35681213

ABSTRACT

BACKGROUND: The Bcl-3 protein is an atypical member of the inhibitor of -κB family that has dual roles as a transcriptional repressor and a coactivator for dimers of NF-κB p50 and p52. Bcl-3 is expressed in mammary adenocarcinomas and can promote tumorigenesis and survival signaling and has a key role in tumor metastasis. In this study, we have investigated the role of Bcl-3 in the normal mammary gland and impact on tumor pathology. METHODS: We utilized bcl-3-/- mice to study mammary gland structure in virgins and during gestation, lactation and early involution. Expression of involution-associated genes and proteins and putative Bcl-3 target genes was examined by qRT-PCR and immunoblot analysis. Cell autonomous branching morphogenesis and collagen I invasion properties of bcl-3-/- organoids were tested in 3D hydrogel cultures. The role of Bcl-3 in tumorigenesis and tumor pathology was also assessed using a stochastic carcinogen-induced mammary tumor model. RESULTS: Bcl-3-/- mammary glands demonstrated reduced branching complexity in virgin and pregnant mice. This defect was recapitulated in vitro where significant defects in bud formation were observed in bcl-3-/- mammary organoid cultures. Bcl-3-/- organoids showed a striking defect in protrusive collective fibrillary collagen I invasion associated with reduced expression of Fzd1 and Twist2. Virgin and pregnant bcl-3-/- glands showed increased apoptosis and rapid increases in lysosomal cell death and apoptosis after forced weaning compared to WT mice. Bcl-2 and Id3 are strongly induced in WT but not bcl-3-/- glands in early involution. Tumors in WT mice were predominately adenocarcinomas with NF-κB activation, while bcl-3-/- lesions were largely squamous lacking NF-κB and with low Bcl-2 expression. CONCLUSIONS: Collectively, our results demonstrate that Bcl-3 has a key function in mammary gland branching morphogenesis, in part by regulation of genes involved in extracellular matrix invasion. Markedly reduced levels of pro-survival proteins expression in bcl-3 null compared to WT glands 24 h post-weaning indicate that Bcl-3 has a role in moderating the rate of early phase involution. Lastly, a reduced incidence of bcl-3-/- mammary adenocarcinomas versus squamous lesions indicates that Bcl-3 supports the progression of epithelial but not metaplastic cancers.


Subject(s)
Adenocarcinoma , B-Cell Lymphoma 3 Protein , Breast Neoplasms , Carcinoma, Squamous Cell , Mammary Glands, Animal , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Apoptosis/genetics , B-Cell Lymphoma 3 Protein/metabolism , Breast Neoplasms/pathology , Carcinogenesis/metabolism , Carcinoma, Squamous Cell/pathology , Collagen/metabolism , Epithelial Cells/metabolism , Female , Lactation , Mammary Glands, Animal/metabolism , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Pregnancy , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism
3.
Cells ; 8(10)2019 10 19.
Article in English | MEDLINE | ID: mdl-31635050

ABSTRACT

SRC-3/AIB1 (Amplified in Breast Cancer-1) is a nuclear receptor coactivator for the estrogen receptor in breast cancer cells. It is also an intrinsically disordered protein when not engaged with transcriptional binding partners and degraded upon transcriptional coactivation. Given the amplified expression of SRC-3 in breast cancers, the objective of this study was to determine how increasing SRC-3 protein levels are regulated in MCF-7 breast cancer cells. We found that endogenous SRC-3 was expelled from the nucleus in vesicle-like spheres under normal growth conditions suggesting that this form of nuclear exclusion of SRC-3 is a homeostatic mechanism for regulating nuclear SRC-3 protein. Only SRC-3 not associated with CREB-binding protein (CBP) was extruded from the nucleus. We found that overexpression in MCF-7 cells results in aneuploid senescence and cell death with frequent formation of nuclear aggregates which were consistently juxtaposed to perinuclear microtubules. Transfected SRC-3 was SUMOylated and caused redistribution of nuclear promyelocytic leukemia (PML) bodies and perturbation of the nuclear membrane lamin B1, hallmarks of nucleophagy. Increased SRC-3 protein-induced autophagy and resulted in SUMO-1 localization to the nuclear membrane and formation of protrusions variously containing SRC-3 and chromatin. Aspects of SRC-3 overexpression and toxicity were recapitulated following treatment with clinically relevant agents that stabilize SRC-3 in breast cancer cells. We conclude that amplified SRC-3 levels have major impacts on nuclear protein quality control pathways and may mark cancer cells for sensitivity to protein stabilizing therapeutics.


Subject(s)
CREB-Binding Protein/metabolism , Nuclear Receptor Coactivator 3/metabolism , Apoptosis/genetics , Apoptosis/physiology , Autophagy/genetics , Autophagy/physiology , CREB-Binding Protein/genetics , Cell Cycle Checkpoints/genetics , Cell Cycle Checkpoints/physiology , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cellular Senescence/drug effects , Cellular Senescence/genetics , Cellular Senescence/physiology , Cyclin E/genetics , Electrophoresis, Polyacrylamide Gel , Female , Humans , MCF-7 Cells , Microscopy, Fluorescence , Microtubules/metabolism , Mitosis/genetics , Mitosis/physiology , Mutagenesis, Site-Directed , Nuclear Envelope/metabolism , Nuclear Proteins/metabolism , Nuclear Receptor Coactivator 3/genetics , Phosphorylation , Promyelocytic Leukemia Protein/genetics , Promyelocytic Leukemia Protein/metabolism , Proteasome Endopeptidase Complex/metabolism , SUMO-1 Protein/genetics , SUMO-1 Protein/metabolism , Tamoxifen/pharmacology
4.
J Mammary Gland Biol Neoplasia ; 23(3): 109-123, 2018 09.
Article in English | MEDLINE | ID: mdl-29876871

ABSTRACT

Cellular inhibitor of apoptosis proteins-1 and -2 (cIAP1/2) are integral to regulation of apoptosis and signaling by the tumor necrosis factor (TNF) and related family of receptors. The expression of cIAP2 in tissues is typically low and considered functionally redundant with cIAP1, however cIAP2 can be activated by a variety of cellular stresses. Members of the TNFR family and their ligands have essential roles in mammary gland biology. We have found that cIAP2-/- virgin mammary glands have reduced ductal branching and delayed lobuloalveogenesis in early pregnancy. Post-lactational involution involves two phases where the first phase is reversible and is mediated, in part, by TNFR family ligands. In cIAP2-/- mice mammary glands appeared engorged at mid-lactation accompanied by enhanced autophagic flux and decreased cIAP1 protein expression. Severely stretched myoepithelium was associated with BIM-EL expression and other indicators of anoikis. Within 24 h after forced or natural weaning, cIAP2-/- glands had nearly completed involution. The TNF-related weak inducer of apoptosis (Tweak) which results in degradation of cIAP1 through its receptor, Fn14, began to increase in late lactation and was significantly increased in cIAP2-/- relative to WT mice by 12 h post weaning accompanied by decreased cIAP1 protein expression. Carcinogen/progesterone-induced mammary tumorigenesis was significantly delayed in cIAP2-/- mice and tumors contained high numbers of apoptotic cells. We conclude that cIAP2 has a critical role in the mammary gland wherein it prevents rapid involution induced by milk stasis-induced stress associated with Tweak activation and contributes to the survival of mammary tumor cells.


Subject(s)
Baculoviral IAP Repeat-Containing 3 Protein/metabolism , Carcinogenesis/metabolism , Lactation/metabolism , Mammary Glands, Animal/metabolism , Signal Transduction/physiology , Animals , Apoptosis , Female , Male , Mice , Mice, Inbred C57BL , Pregnancy , Weaning
5.
Cancer Prev Res (Phila) ; 11(2): 69-80, 2018 02.
Article in English | MEDLINE | ID: mdl-29101208

ABSTRACT

Recent studies have shown that progesterone receptor (PR)-expressing cells respond to progesterone in part through the induction of the receptor activator of NF-κB ligand (RANKL), which acts in a paracrine manner to induce expansion of a RANK-expressing luminal progenitor cell population. The RANK+ population in human breast tissue from carriers of BRCA1 mutations (BRCA1mut/+) as well as the luminal progenitor population in Brca1-deficient mouse mammary glands is abnormally amplified. Remarkably, mouse Brca1+/- and human BRCA1mut/+ progenitor cells are able to form colonies in vitro in the absence of progesterone, demonstrating a hormone-independent proliferative capacity. Our research has demonstrated that proliferation in BRCA1-deficient cells results in a DNA damage response (DDR) that activates a persistent NF-κB signal, which supplants progesterone/RANKL signaling for an extended time period. Thus, the transcriptional targets normally activated by RANKL that promote a proliferative response in luminal progenitors can contribute to the susceptibility of mammary epithelial cells to BRCA1-mutated breast cancers as a consequence of DDR-induced NF-κB. Together, these latest findings mark substantial progress in uncovering the mechanisms driving high rates of breast tumorigenesis in BRCA1 mutation carriers and ultimately reveal possibilities for nonsurgical prevention strategies. Cancer Prev Res; 11(2); 69-80. ©2017 AACR.


Subject(s)
BRCA1 Protein/genetics , Breast Neoplasms/pathology , Breast/pathology , Cell Transformation, Neoplastic/pathology , Mutation , NF-kappa B/metabolism , Breast/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Female , Humans , Signal Transduction
6.
Cell Stem Cell ; 19(1): 52-65, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27292187

ABSTRACT

Human BRCA1 mutation carriers and BRCA1-deficient mouse mammary glands contain an abnormal population of mammary luminal progenitors that can form 3D colonies in a hormone-independent manner. The intrinsic cellular regulatory defect in these presumptive breast cancer precursors is not known. We have discovered that nuclear factor kappaB (NF-κB) (p52/RelB) is persistently activated in a subset of BRCA1-deficient mammary luminal progenitors. Hormone-independent luminal progenitor colony formation required NF-κB, ataxia telangiectasia-mutated (ATM), and the inhibitor of kappaB kinase, IKKα. Progesterone (P4)-stimulated proliferation resulted in a marked enhancement of DNA damage foci in Brca1(-/-) mouse mammary. In vivo, NF-κB inhibition prevented recovery of Brca1(-/-) hormone-independent colony-forming cells. The majority of human BRCA1(mut/+) mammary glands showed marked lobular expression of nuclear NF-κB. We conclude that the aberrant proliferative capacity of Brca1(-/-) luminal progenitor cells is linked to the replication-associated DNA damage response, where proliferation of mammary progenitors is perpetuated by damage-induced, autologous NF-κB signaling.


Subject(s)
BRCA1 Protein/deficiency , Breast/pathology , DNA Damage , Mammary Glands, Animal/pathology , NF-kappa B/metabolism , Stem Cells/pathology , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , BRCA1 Protein/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Knockdown Techniques , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mice , NF-kappa B p52 Subunit/metabolism , Phosphorylation/drug effects , Phosphoserine/metabolism , Progesterone/pharmacology , Protein Binding/drug effects , Sesquiterpenes/pharmacology , Signal Transduction/drug effects , Stem Cells/drug effects , Stem Cells/metabolism , Tumor Stem Cell Assay
7.
Endocrinology ; 155(7): 2480-91, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24823389

ABSTRACT

Vasomotor thermo-dysregulation (hot flashes) are an often debilitating symptom of menopause. Effective treatment is achieved primarily through activation of the estrogen receptor (ER)α with estrogens but is also associated with increased risk for breast and uterine cancer. In this study, we have tested novel compounds lacking the B ring of 17-hydroxy-ß-estradiol (E2) (A-CD compounds) with differing ratios of ERα:ERß binding affinities for the ability to reduce diurnal/nocturnal tail-skin temperatures (TSTs) in the ovariectomized female rat menopausal hot flash model. Normal mammary tissue expresses the predominantly antiproliferative ERß. Therefore, we hypothesized that a preferential ERß agonist with fractional ERα activity would safely reduce TSTs. The A-CD compound, L17, is a preferential ERß agonist that has a ratio of ERß:ERα binding affinity relative to E2 of 9.3 (where ERß:ERα for E2, 1.0). In the ovariectomized rat, daily administration of low doses (1 mg/kg) of the A-CD compound TD81 (ERα:ERß relative affinity, 15.2) was ineffective in temperature regulation, whereas L17 showed a trend toward TST reduction. Both E2 and the A-CD compound, TD3 (ERß:ERα relative affinity, 5.0), also reduced TSTs but had marked proliferative effects on mammary and uterine tissues. At 2 mg/kg, L17 strongly reduced TSTs even more effectively than E2 but, importantly, had only minimal effect on uterine weight and mammary tissues. Both E2- and L17-treated rats showed similar weight reduction over the treatment period. E2 is rapidly metabolized to highly reactive quinones, and we show that L17 has 2-fold greater metabolic stability than E2. Finally, L17 and E2 similarly mediated induction of c-fos expression in neurons within the rat thermoregulatory hypothalamic median preoptic nucleus. Thus, the A-CD compound, L17, may represent a safe and effective approach to the treatment of menopausal hot flashes.


Subject(s)
Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Menopause , Models, Animal , Vasomotor System/physiology , Adiposity/drug effects , Animals , Binding, Competitive , Body Temperature/drug effects , Body Temperature/physiology , Estradiol/chemistry , Estradiol/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/agonists , Estrogen Receptor beta/agonists , Estrogens/chemistry , Estrogens/metabolism , Estrogens/pharmacology , Female , Hot Flashes/metabolism , Hot Flashes/physiopathology , Humans , Immunohistochemistry , Ligands , Mammary Glands, Human/drug effects , Mammary Glands, Human/growth & development , Mammary Glands, Human/metabolism , Molecular Structure , Ovariectomy , Preoptic Area/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Wistar , Uterus/drug effects , Uterus/growth & development , Uterus/metabolism , Vasomotor System/drug effects , Weight Gain/drug effects
8.
Mol Cancer Ther ; 13(7): 1882-93, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24785256

ABSTRACT

Acquired resistance to selective estrogen receptor (ER) modulators (SERM) and downregulators (SERD) is a significant clinical problem in the treatment of estrogen (E2) receptor-positive (ER(+)) breast cancers. There are two ER subtypes, ERα and ERß, which promote and inhibit breast cancer cell proliferation, respectively. Although ER(+) breast cancers typically express a high ratio of ERα to ERß, the acquisition of SERM resistance in vitro and in vivo is associated with increased relative expression of the ERß. On some gene enhancers, ERß has been shown to function in opposition to the ERα in the presence of E2. Here, we demonstrate that two different ERß agonists, WAY-20070 and a novel "A-CD" estrogen called L17, produce a marked reduction in G(2)-M phase correlated with effects on cyclin D1 and cyclin E expression in a SERM/SERD-resistant breast cancer cell line. ERß agonists recruited both the ERα and ERß to the Bcl-2 E2-response element strongly reducing Bcl-2 mRNA and protein in an ERß-dependent manner. L17 recruited RIP140 to the Bcl-2 promoter in cells overexpressing ERß. Exposure to the ERß ligands also resulted in increased processing of LC3-I to LC3-II, indicative of enhanced autophagic flux. The coaddition of ERß agonist and the autophagy inhibitor chloroquine resulted in a significant accumulation of sub-G1 DNA which was completely prevented by the addition of the caspase inhibitor Z-VAD-FMK. We propose that combined therapies with an ERß agonist and an inhibitor of autophagy may provide the basis for a novel approach to the treatment of SERM/SERD-resistant breast cancers.


Subject(s)
Breast Neoplasms/drug therapy , Estrogen Receptor beta/agonists , Estrogens/pharmacology , Oxazoles/pharmacology , Phenols/pharmacology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Autophagy/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Female , Humans , Ligands , Selective Estrogen Receptor Modulators/pharmacology , Signal Transduction/drug effects
9.
PLoS One ; 8(2): e57426, 2013.
Article in English | MEDLINE | ID: mdl-23437386

ABSTRACT

The cold inducible RNA binding protein (CIRBP) responds to a wide array of cellular stresses, including short wavelength ultraviolet light (UVC), at the transcriptional and post-translational level. CIRBP can bind the 3'untranslated region of specific transcripts to stabilize them and facilitate their transport to ribosomes for translation. Here we used RNA interference and oligonucleotide microarrays to identify potential downstream targets of CIRBP induced in response to UVC. Twenty eight transcripts were statistically increased in response to UVC and these exhibited a typical UVC response. Only 5 of the 28 UVC-induced transcripts exhibited a CIRBP-dependent pattern of expression. Surprisingly, 3 of the 5 transcripts (IL1B, IL8 and TNFAIP6) encoded proteins important in inflammation with IL-1ß apparently contributing to IL8 and TNFAIP6 expression in an autocrine fashion. UVC-induced IL1B expression could be inhibited by pharmacological inhibition of NFκB suggesting that CIRBP was affecting NF-κB signaling as opposed to IL1B mRNA stability directly. Bacterial lipopolysaccharide (LPS) was used as an activator of NF-κB to further study the potential link between CIRBP and NFκB. Transfection of siRNAs against CIRBP reduced the extent of the LPS-induced phosphorylation of IκBα, NF-κB DNA binding activity and IL-1ß expression. The present work firmly establishes a novel link between CIRBP and NF-κB signaling in response to agents with diverse modes of action. These results have potential implications for disease states associated with inflammation.


Subject(s)
Interleukin-1beta/genetics , NF-kappa B/genetics , RNA, Messenger/biosynthesis , RNA-Binding Proteins/genetics , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Cold Temperature , Fibroblasts , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Humans , I-kappa B Proteins/genetics , I-kappa B Proteins/immunology , Interleukin-1beta/immunology , Interleukin-8/genetics , Interleukin-8/immunology , Lipopolysaccharides/pharmacology , NF-KappaB Inhibitor alpha , NF-kappa B/immunology , Oligonucleotide Array Sequence Analysis , Phosphorylation/drug effects , Phosphorylation/radiation effects , Primary Cell Culture , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Small Interfering/genetics , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/immunology , Signal Transduction/drug effects , Signal Transduction/radiation effects , Ultraviolet Rays
10.
Cell Cycle ; 11(21): 4009-19, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23032264

ABSTRACT

Cellular inhibitor of apoptosis proteins (cIAP1 and cIAP2) function to prevent apoptosis and are often overexpressed in various cancers. However, mutations in cIAP1/2 can activate the alternative NFκB pathway through IκBα-kinase-α (IKKα) and are associated with hematopoetic malignancies. In the current study, we found that knockdown of cIAP2 in human mammary epithelial cells resulted in activation of MDM2 through increased SUMOylation and profound reduction of the pool of MDM2 not phosphorylated at Ser166. cIAP2 siRNA markedly decreased p53 levels, which were rescued by addition of the MDM2 inhibitor, Nutlin3a. An IAP antagonist, which induces cIAP degradation, transiently increased MDM2 mRNA. Simultaneous transfection of siRNA for cIAP2 and IKKα reduced MDM2 protein, while expression of a kinase-dead IKKß strongly increased non-Ser166 P-MDM2. Inhibition of either IKKα or -ß partially rescued p53 levels, while concomitant IKKα/ß inhibition fully rescued p53 after cIAP2 knockdown. Surprisingly, IKKα knockdown alone increased SUMO-MDM2, suggesting that in the absence of activation, IKKα can prevent MDM2 SUMOylation. cIAP2 knockdown disrupted the interaction between the MDM2 SUMO ligase, PIAS1 and IKKα. Partial knockdown of cIAP2 cooperated with (V12) H-ras-transfected mammary epithelial cells to enhance colony formation. In summary, our data identify a novel role for cIAP2 in maintaining wild-type p53 levels by preventing both an NFκB-mediated increase and IKKα/-ß-dependent transcriptional and post-translational modifications of MDM2. Thus, mutations or reductions in cIAP2 could contribute to cancer promotion, in part, through downregulation of p53.


Subject(s)
I-kappa B Kinase/metabolism , Inhibitor of Apoptosis Proteins/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Baculoviral IAP Repeat-Containing 3 Protein , Cell Line , Down-Regulation/drug effects , Enzyme Inhibitors/pharmacology , Humans , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/genetics , Imidazoles/pharmacology , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/genetics , Mutation , Phosphorylation , Piperazines/pharmacology , Protein Inhibitors of Activated STAT/metabolism , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/genetics , RNA, Messenger/metabolism , Sumoylation , Ubiquitin-Protein Ligases
12.
J Med Chem ; 54(2): 433-48, 2011 Jan 27.
Article in English | MEDLINE | ID: mdl-21190382

ABSTRACT

Long-term use of estrogen supplements by women leads to an increased risk of breast and uterine cancers. Possible mechanisms include metabolism of estradiol and compounds related to tumor-initiating quinones, and ligand-induced activation of the estrogen receptors ERα and ERß which can cause cancer cell proliferation, depending on the ratio of receptors present. One therapeutic goal would be to create a spectrum of compounds of variable potency for ERα and ERß, which are resistant to quinone formation, and to determine an optimum point in this spectrum. We describe the synthesis, modeling, binding affinities, hormone potency, and a measure of quinone formation for a new family of A-CD estrogens, where the A-C bond is formed by ring coupling. Some substituents on the A-ring increase hormone potency, and one compound is much less quinone-forming than estradiol. These compounds span a wide range of receptor subtype selectivities and may be useful in hormone replacement therapy.


Subject(s)
Estradiol Congeners/chemical synthesis , Estradiol/chemistry , Estrogen Receptor alpha/agonists , Estrogen Receptor beta/agonists , Animals , Binding, Competitive , Cell Line , Estradiol Congeners/chemistry , Estradiol Congeners/pharmacology , Fluorine/chemistry , Hepatocytes/cytology , Hepatocytes/drug effects , Humans , Ligands , Male , Models, Molecular , Molecular Structure , Quinones/metabolism , Rats , Rats, Sprague-Dawley , Response Elements , Stereoisomerism , Structure-Activity Relationship , Thermodynamics , Transcriptional Activation/drug effects
13.
J Cell Biochem ; 107(3): 448-59, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19350539

ABSTRACT

Estrogen receptor (ER)-positive breast cancer cells have low levels of constitutive NF-kappaB activity while ER negative (-) cells and hormone-independent cells have relatively high constitutive levels of NF-kappaB activity. In this study, we have examined the aspects of mutual repression between the ERalpha and NF-kappaB proteins in ER+ and ER- hormone-independent cells. Ectopic expression of the ERalpha reduced cell numbers in ER+ and ER- breast cancer cell lines while NF-kappaB-binding activity and the expression of several NF-kappaB-regulated proteins were reduced in ER- cells. ER overexpression in ER+/E2-independent LCC1 cells only weakly inhibited the predominant p50 NF-kappaB. GST-ERalpha fusion protein pull downs and in vivo co-immunoprecipitations of NF-kappaB:ERalpha complexes showed that the ERalpha interacts with p50 and p65 in vitro and in vivo. Inhibition of NF-kappaB increased the expression of diverse E2-regulated proteins. p50 differentially associated directly with the ER:ERE complex in LCC1 and MCF-7 cells by supershift analysis while p65 antibody reduced ERalpha:ERE complexes in the absence of a supershift. ChIP analysis demonstrated that NF-kappaB proteins are present on an endogenous ERE. Together these results demonstrate that the ER and NF-kappaB undergo mutual repression, which may explain, in part, why expression of the ERalpha in ER- cells does not confer growth signaling. Secondly, the acquisition of E2-independence in ER+ cells is associated with predominantly p50:p50 NF-kappaB, which may reflect alterations in the ER in these cells. Since the p50 homodimer is less sensitive to the presence of the ER, this may allow for the activation of both pathways in the same cell.


Subject(s)
Breast Neoplasms/metabolism , Estrogen Receptor alpha/metabolism , NF-kappa B/metabolism , Binding Sites , Breast Neoplasms/genetics , Cell Line, Tumor , Female , Humans , NF-kappa B p50 Subunit/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , Transfection
14.
Apoptosis ; 12(4): 657-69, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17252199

ABSTRACT

The Bcl-2 gene is positively regulated by estrogen (E2) primarily through E2-response elements in the coding region and a putative p53 negative regulatory element (NRE) containing a short upstream open reading frame (uORF). The ability of mutant p53 to repress or induce Bcl-2 expression is controversial. In this study E2-receptor positive (ER(+))/wild-type p53 MCF-7cells were transfected with p53Delta291, which lacks a nuclear localization signal or a DNA binding domain mutant, p53(173L). Both p53 mutants but especially p53Delta291 increased Bcl-2 protein expression from a CMV-NRE-Bcl-2 cDNA construct in an NRE-position/orientation independent manner as well as from a 1.7 kb Bcl-2 promoter reporter gene. Bcl-2 protein expression prevented the p53Delta291-mediated increase in Bcl-2 promoter activity although immunoprecipitation demonstrated that only a small proportion of the wild-type p53 but not p53Delta91 protein interacts with Bcl-2. Unless levels of ectopically expressed mutant p53 were extremely high, stable expression of mutant p53 in MCF-7 cells moderately increased Bcl-2 protein levels. Expression of mutant p53 did not alter E2 regulation of Bcl-2, however, mutation of the uORF prevented regulation by both mutant p53 and E2. Adenovirus-mediated overexpression of WT p53 strongly reduced Bcl-2 expression in ER(-)/mut p53 MDA-MB-231 cells. Taken together these data support the position that mutant p53 behaves in a dominant "positive" manner relieving repression by WT p53 or another Bcl-2 transcriptional inhibitor in a manner independent of nuclear translocation.


Subject(s)
Breast Neoplasms/metabolism , Gene Expression Regulation , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Estrogen/metabolism , Tumor Suppressor Protein p53/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Mutation , Promoter Regions, Genetic , Proto-Oncogene Proteins c-bcl-2/genetics , Regulatory Sequences, Nucleic Acid , Transcription, Genetic , Tumor Suppressor Protein p53/genetics
15.
Apoptosis ; 11(4): 589-605, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16528475

ABSTRACT

The chemotherapeutic drug paclitaxel induces microtubular stabilization and mitotic arrest associated with increased survivin expression. Survivin is a member of the inhibitor of apoptosis (iap) family which is highly expressed in during G2/M phase where it regulates spindle formation during mitosis. It is also constitutively overexpressed in most cancer cells where it may play a role in chemotherapeutic resistance. MCF-7 breast cancer cells stably overexpressing the sense strand of survivin (MCF-7(survivin-S) cells) were more resistant to paclitaxel than cells depleted of survivin (MCF-7(survivin-AS) despite G2/M arrest in both cell lines. However, survivin overexpression did not protect cells relative to control MCF-7(pcDNA3) cells. Paclitaxel-induced cytotoxicity can be enhanced by retinoic acid and here we show that RA strongly reduces survivin expression in MCF-7 cells and prevents paclitaxel-mediated induction of survivin expression. Mitochondrial release of cytochrome c after paclitaxel alone or in combination with RA was weak, however robust Smac release was observed. While RA/paclitaxel-treated MCF-7 (pcDNA3) cultures contained condensed apoptotic nuclei, MCF-7(survivin-S) nuclei were morphologically distinct with hypercondensed disorganized chromatin and released mitochondrial AIF-1. RA also reduced paclitaxel-associated levels of cyclin B1 expression consistent with mitotic exit. MCF-7(survivin-S) cells displayed a 30% increase in >2N/<4N ploidy while there was no change in this compartment in vector control cells following RA/paclitaxel. We propose that RA sensitizes MCF-7 cells to paclitaxel at least in part through survivin downregulation and the promotion of aberrant mitotic progression resulting in apoptosis. In addition we provide biochemical and morphological data which suggest that RA-treated MCF-7(survivin-S) cells can also undergo catastrophic mitosis when exposed to paclitaxel.


Subject(s)
Antineoplastic Agents, Phytogenic/toxicity , Apoptosis , Microtubule-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , Paclitaxel/toxicity , Tretinoin/pharmacology , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Apoptosis Inducing Factor/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/ultrastructure , Caspases/metabolism , Cell Cycle , Cell Line, Tumor , Cell Nucleus/ultrastructure , Cyclin B/metabolism , Cyclin B1 , Drug Synergism , Female , Humans , Inhibitor of Apoptosis Proteins , Paclitaxel/antagonists & inhibitors , Ploidies , Survivin
16.
J Cell Biochem ; 90(4): 692-708, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-14587026

ABSTRACT

All-trans retinoic acid (ATRA) can down regulate the anti-apoptotic protein Bcl-2 and the cell cycle proteins cyclin D1 and cdk2 in estrogen receptor-positive breast cancer cells. We show here that retinoids can also reduce expression of the inhibitor of apoptosis protein, survivin. Here we have compared the regulation of these proteins in MCF-7 and ZR-75 breast cancer cells by natural and synthetic retinoids selective for the RA receptors (RARs) alpha, beta, and gamma then correlated these with growth inhibition, induction of apoptosis and chemosensitization to Taxol. In both cell lines ATRA and 9-cis RA induced the most profound decreases in cyclin D1 and cdk2 expression and also mediated the largest growth inhibition. The RARalpha agonist, Ro 40-6055 also strongly downregulated these proteins although did not produce an equivalent decrease in S-phase cells. Only ATRA induced RARbeta expression. ATRA, 9-cis RA and 4-HPR initiated the highest level of apoptosis as determined by mitochondrial Bax translocation, while only ATRA and 9-cis RA strongly reduced Bcl-2 and survivin protein expression. Enumeration of dead cells over 96 h correlated well with downregulation of both survivin and Bcl-2. Simultaneous retinoid-mediated reduction of both these proteins also predicted optimal Taxol sensitization. 4-HPR was much weaker than the natural retinoids with respect to Taxol sensitization, consistent with the proposed requirement for reduced Bcl-2 in this synergy. Neither the extent of cell cycle protein regulation nor AP-1 inhibition fully predicted the antiproliferative effect of the synthetic retinoids suggesting that growth inhibition requires regulation of a spectrum of RAR-regulated gene products in addition even to pivotal cell cycle proteins.


Subject(s)
Apoptosis/drug effects , Retinoids/pharmacology , CDC2-CDC28 Kinases/metabolism , Cell Division/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Clone Cells/drug effects , Cyclin D1/metabolism , Cyclin-Dependent Kinase 2 , Gene Expression Regulation/drug effects , Humans , Immunohistochemistry , Inhibitor of Apoptosis Proteins , Ligands , Microtubule-Associated Proteins/metabolism , Neoplasm Proteins , Paclitaxel/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Retinoic Acid/metabolism , Retinoids/chemical synthesis , Retinoids/chemistry , S Phase/drug effects , Survivin , Transcription Factor AP-1/metabolism
17.
Mol Cell Biol ; 23(19): 6887-900, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12972607

ABSTRACT

About one-third of breast cancers express a functional estrogen (beta-estradiol [E2]) receptor (ER) and are initially dependent on E2 for growth and survival but eventually progress to hormone independence. We show here that ER(+), E2-independent MCF-7/LCC1 cells derived from E2-dependent MCF-7 cells contain elevated basal NF-kappaB activity and elevated expression of the transcriptional coactivator Bcl-3 compared with the parental MCF-7 line. LCC1 NF-kappaB activity consists primarily of p50 dimers, although low levels of a p65/p50 complex are also present. The ER(-) breast cancer cell lines harbor abundant levels of both NF-kappaB complexes. In contrast, nuclear extracts from MCF-7 cells contain a significantly lower level of p50 and p65 than do LCC1 cells. Estrogen withdrawal increases both NF-kappaB DNA binding activity and expression of Bcl-3 in MCF-7 and LCC1 cells in vitro and in vivo. Tumors derived from MCF-7 cells ectopically expressing Bcl-3 remain E2 dependent but display a markedly higher tumor establishment and growth rate compared to controls. Expression of a stable form of IkappaBalpha in LCC1 cells severely reduced nuclear expression of p65 and the p65/p50 DNA binding heterodimer. Whereas LCC1 tumors in nude mice were stable or grew, LCC1(IkappaBalpha) tumors regressed after E2 withdrawal. Thus, both p50/Bcl-3- and p65/p50-associated NF-kappaB activities are activated early in progression and serve differential roles in growth and hormone independence, respectively. We propose that E2 withdrawal may initiate selection for hormone independence in breast cancer cells by activation of NF-kappaB and Bcl-3, which could then supplant E2 by providing both survival and growth signals.


Subject(s)
Breast Neoplasms/genetics , Estrogens/metabolism , Gene Expression Regulation, Neoplastic , NF-kappa B/metabolism , Proto-Oncogene Proteins/metabolism , Animals , B-Cell Lymphoma 3 Protein , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Transformed , Cell Nucleus/chemistry , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Dimerization , Female , Humans , Mice , Mice, Nude , Models, Biological , NF-kappa B/antagonists & inhibitors , NF-kappa B/chemistry , Neoplasm Proteins/genetics , Neoplasm Transplantation , Oncogenes , Ovariectomy , Receptors, Estrogen/metabolism , Transcription Factors , Transcriptional Activation
18.
J Biol Chem ; 278(16): 14219-29, 2003 Apr 18.
Article in English | MEDLINE | ID: mdl-12480939

ABSTRACT

MCF-7 and ZR-75 breast cancer cells infected with an adenovirus constitutively expressing high levels of cyclin D1 demonstrated widespread mitochondrial translocation of Bax and cytochrome c release that was approximately doubled after the addition of all-trans retinoic acid (RA) or Bcl-2 antisense oligonucleotide. By comparison, the percentage of cells in Lac Z virus-infected cultures containing translocated Bax and cytoplasmic cytochrome c was markedly less even after RA treatment. Despite this, RA-treated Lac Z and untreated cyclin D1 virus-infected cultures contained similarly low proportions of cells with active caspase or cells that were permeable to propidium iodide. Bax activation was p53-dependent and accompanied by arrest in G(2) phase. Although constitutive Bcl-2 expression prevented Bax activation, it did not alter cyclin D1-induced cell cycle arrest, illustrating the independence of these events. Both RA and antisense Bcl-2 oligonucleotide decreased Bcl-2 protein levels and markedly increased caspase activity and apoptosis in cyclin D1-infected cells. Thus amplified cyclin D1 expression initiates an apoptotic signal inhibited by different levels of cellular Bcl-2 at two points. Whereas high cellular levels of Bcl-2 prevent mitochondrial Bax translocation, lower levels can prevent apoptosis by inhibition of caspase activation.


Subject(s)
Caspases/metabolism , Cyclin D1/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Suppressor Protein p53/metabolism , Apoptosis , Biological Transport , Cell Cycle , Cell Death , Cytochrome c Group/metabolism , Enzyme Activation , Epitopes , Humans , Immunoblotting , Immunohistochemistry , Lac Operon , Membrane Potentials , Microscopy, Fluorescence , Mitochondria/metabolism , Oligonucleotides, Antisense/pharmacology , Plasmids/metabolism , Propidium/pharmacology , Protein Binding , Protein Structure, Tertiary , Protein Transport , Proto-Oncogene Proteins/metabolism , Signal Transduction , Subcellular Fractions/metabolism , Time Factors , Tretinoin/metabolism , Tumor Cells, Cultured , bcl-2-Associated X Protein
SELECTION OF CITATIONS
SEARCH DETAIL