Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Pigment Cell Melanoma Res ; 36(5): 365-377, 2023 09.
Article in English | MEDLINE | ID: mdl-37341054

ABSTRACT

An increasing number of cancer subtypes are treated with front-line immunotherapy. However, approaches to overcome primary and acquired resistance remain limited. Preclinical mouse models are often used to investigate resistance mechanisms, novel drug combinations, and delivery methods; yet most of these models lack the genetic diversity and mutational patterns observed in human tumors. Here we describe a series of 13 C57BL/6J melanoma cell lines to address this gap in the field. The Ohio State University-Moffitt Melanoma Exposed to Radiation (OSUMMER) cell lines are derived from mice expressing endogenous, melanocyte-specific, and clinically relevant Nras driver mutations (Q61R, Q61K, or Q61L). Exposure of these animals to a single, non-burning dose of ultraviolet B accelerates the onset of spontaneous melanomas with mutational patterns akin to human disease. Furthermore, in vivo irradiation selects against potent tumor antigens, which could prevent the outgrowth of syngeneic cell transfers. Each OSUMMER cell line possesses distinct in vitro growth properties, trametinib sensitivity, mutational signatures, and predicted antigenicity. Analysis of OSUMMER allografts shows a correlation between strong, predicted antigenicity and poor tumor outgrowth. These data suggest that the OSUMMER lines will be a valuable tool for modeling the heterogeneous responses of human melanomas to targeted and immune-based therapies.


Subject(s)
Cell Line, Tumor , Melanoma , Animals , Mice , Cell Line, Tumor/radiation effects , GTP Phosphohydrolases/genetics , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Membrane Proteins/genetics , Mice, Inbred C57BL , Mutation/genetics , Proto-Oncogene Proteins B-raf/genetics
2.
Pigment Cell Melanoma Res ; 36(1): 6-18, 2023 01.
Article in English | MEDLINE | ID: mdl-36148789

ABSTRACT

Melanin is a free-radical scavenger, antioxidant, and broadband absorber of ultraviolet (UV) radiation which protects the skin from environmental carcinogenesis. However, melanin synthesis and UV-induced reactive melanin species are also implicated in melanocyte genotoxicity. Here, we attempted to reconcile these disparate functions of melanin using a UVB-sensitive, NRAS-mutant mouse model, TpN. We crossed TpN mice heterozygous for an inactivating mutation in Tyrosinase to produce albino and black littermates on a C57BL/6J background. These animals were then exposed to a single UVB dose on postnatal day three when keratinocytes in the skin have yet to be melanized. Approximately one-third (35%) of black mice were protected from UVB-accelerated tumor formation. However, melanoma growth rates, tumor mutational burdens, and gene expression profiles were similar in melanomas from black and albino mice. Skin from albino mice contained more cyclobutane pyrimidine dimer (CPD) positive cells than black mice 1-h post-irradiation. However, this trend gradually reversed over time with CPDs becoming more prominent in black than albino melanocytes at 48 h. These results show that in the absence of epidermal pigmentation, melanocytic melanin limits the tumorigenic effects of acute UV exposure but fails to protect melanocytes from UVB-induced mutagenesis.


Subject(s)
Melanoma , Skin Neoplasms , Mice , Animals , Melanins/metabolism , Skin Neoplasms/pathology , Mice, Inbred C57BL , Melanocytes/metabolism , Melanoma/pathology , Ultraviolet Rays , Mutagenesis
3.
Nat Commun ; 13(1): 3153, 2022 06 07.
Article in English | MEDLINE | ID: mdl-35672316

ABSTRACT

A distinct profile of NRAS mutants is observed in each tumor type. It is unclear whether these profiles are determined by mutagenic events or functional differences between NRAS oncoproteins. Here, we establish functional hallmarks of NRAS mutants enriched in human melanoma. We generate eight conditional, knock-in mouse models and show that rare melanoma mutants (NRAS G12D, G13D, G13R, Q61H, and Q61P) are poor drivers of spontaneous melanoma formation, whereas common melanoma mutants (NRAS Q61R, Q61K, or Q61L) induce rapid tumor onset with high penetrance. Molecular dynamics simulations, combined with cell-based protein-protein interaction studies, reveal that melanomagenic NRAS mutants form intramolecular contacts that enhance BRAF binding affinity, BRAF-CRAF heterodimer formation, and MAPK > ERK signaling. Along with the allelic series of conditional mouse models we describe, these results establish a mechanistic basis for the enrichment of specific NRAS mutants in human melanoma.


Subject(s)
Melanoma , Monomeric GTP-Binding Proteins/standards , Skin Neoplasms , Animals , Disease Models, Animal , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Melanoma/genetics , Melanoma/pathology , Membrane Proteins/genetics , Mice , Mutation , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Signal Transduction/genetics , Skin Neoplasms/genetics
4.
Life Sci Alliance ; 4(9)2021 09.
Article in English | MEDLINE | ID: mdl-34210801

ABSTRACT

BRAF-mutant melanomas are more likely than NRAS-mutant melanomas to arise in anatomical locations protected from chronic sun damage. We hypothesized that this discrepancy in tumor location is a consequence of the differential sensitivity of BRAF and NRAS-mutant melanocytes to ultraviolet light (UV)-mediated carcinogenesis. We tested this hypothesis by comparing the mutagenic consequences of a single neonatal, ultraviolet-AI (UVA; 340-400 nm) or ultraviolet-B (UVB; 280-390 nm) exposure in mouse models heterozygous for mutant Braf or homozygous for mutant Nras Tumor onset was accelerated by UVB, but not UVA, and the resulting melanomas contained recurrent mutations affecting the RING domain of MAP3K1 and Actin-binding domain of Filamin A. Melanomas from UVB-irradiated, Braf-mutant mice averaged twice as many single-nucleotide variants and five times as many dipyrimidine variants than tumors from similarly irradiated Nras-mutant mice. A mutational signature discovered in UVB-accelerated tumors mirrored COSMIC signatures associated with human skin cancer and was more prominent in Braf- than Nras-mutant murine melanomas. These data show that a single UVB exposure yields a greater burden of mutations in murine tumors driven by oncogenic Braf.


Subject(s)
Melanoma/etiology , Monomeric GTP-Binding Proteins/genetics , Mutagenesis/radiation effects , Mutation/radiation effects , Proto-Oncogene Proteins B-raf/genetics , Ultraviolet Rays/adverse effects , Animals , Biomarkers, Tumor , Disease Models, Animal , Disease Susceptibility , Genetic Predisposition to Disease , Melanoma/metabolism , Melanoma/pathology , Mice
5.
Carcinogenesis ; 41(12): 1660-1670, 2020 12 31.
Article in English | MEDLINE | ID: mdl-32894276

ABSTRACT

Over 70% of breast cancers express the estrogen receptor (ER) and depend on ER activity for survival and proliferation. While hormone therapies that target receptor activity are initially effective, patients invariably develop resistance which is often associated with activation of the phosphoinositide 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway. While the mechanism by which estrogen regulates proliferation is not fully understood, one gene target of ER, growth regulation by estrogen in breast cancer 1 (GREB1), is required for hormone-dependent proliferation. However, the molecular function by which GREB1 regulates proliferation is unknown. Herein, we validate that knockdown of GREB1 results in growth arrest and that exogenous GREB1 expression initiates senescence, suggesting that an optimal level of GREB1 expression is necessary for proliferation of breast cancer cells. Under both of these conditions, GREB1 is able to regulate signaling through the PI3K/Akt/mTOR pathway. GREB1 acts intrinsically through PI3K to regulate phosphatidylinositol (3,4,5)-triphosphate levels and Akt activity. Critically, growth suppression of estrogen-dependent breast cancer cells by GREB1 knockdown is rescued by expression of constitutively activated Akt. Together, these data identify a novel molecular function by which GREB1 regulates breast cancer proliferation through Akt activation and provides a mechanistic link between estrogen signaling and the PI3K pathway.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Estrogen/metabolism , Apoptosis , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Proliferation , Female , Humans , Neoplasm Proteins/genetics , Phosphatidylinositol 3-Kinase/genetics , Proto-Oncogene Proteins c-akt/genetics , Tumor Cells, Cultured
6.
Breast Cancer Res ; 22(1): 41, 2020 05 05.
Article in English | MEDLINE | ID: mdl-32370801

ABSTRACT

BACKGROUND: In utero endocrine disruption is linked to increased risk of breast cancer later in life. Despite numerous studies establishing this linkage, the long-term molecular changes that predispose mammary cells to carcinogenic transformation are unknown. Herein, we investigated how endocrine disrupting compounds (EDCs) drive changes within the stroma that can contribute to breast cancer susceptibility. METHODS: We utilized bisphenol A (BPA) as a model of estrogenic endocrine disruption to analyze the long-term consequences in the stroma. Deregulated genes were identified by RNA-seq transcriptional profiling of adult primary fibroblasts, isolated from female mice exposed to in utero BPA. Collagen staining, collagen imaging techniques, and permeability assays were used to characterize changes to the extracellular matrix. Finally, gland stiffness tests were performed on exposed and control mammary glands. RESULTS: We identified significant transcriptional deregulation of adult fibroblasts exposed to in utero BPA. Deregulated genes were associated with cancer pathways and specifically extracellular matrix composition. Multiple collagen genes were more highly expressed in the BPA-exposed fibroblasts resulting in increased collagen deposition in the adult mammary gland. This transcriptional reprogramming of BPA-exposed fibroblasts generates a less permeable extracellular matrix and a stiffer mammary gland. These phenotypes were only observed in adult 12-week-old, but not 4-week-old, mice. Additionally, diethylstilbestrol, known to increase breast cancer risk in humans, also increases gland stiffness similar to BPA, while bisphenol S does not. CONCLUSIONS: As breast stiffness, extracellular matrix density, and collagen deposition have been directly linked to breast cancer risk, these data mechanistically connect EDC exposures to molecular alterations associated with increased disease susceptibility. These alterations develop over time and thus contribute to cancer risk in adulthood.


Subject(s)
Endocrine Disruptors/toxicity , Extracellular Matrix/pathology , Mammary Glands, Animal/pathology , Prenatal Exposure Delayed Effects/pathology , Stromal Cells/pathology , Animals , Benzhydryl Compounds/toxicity , Estrogens, Non-Steroidal/pharmacology , Extracellular Matrix/drug effects , Extracellular Matrix/immunology , Female , Fibroblasts/immunology , Fibroblasts/pathology , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/immunology , Mammary Glands, Animal/metabolism , Mice , Phenols/toxicity , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/metabolism , Stromal Cells/drug effects , Stromal Cells/immunology , Transcriptome
7.
Endocr Relat Cancer ; 25(7): 735-746, 2018 07.
Article in English | MEDLINE | ID: mdl-29695586

ABSTRACT

Activation of the transcription factor estrogen receptor α (ERα) and the subsequent regulation of estrogen-responsive genes play a crucial role in the development and progression of the majority of breast cancers. One gene target of ERα, growth regulation by estrogen in breast cancer 1 (GREB1), is associated with proliferation and regulation of ERα activity in estrogen-responsive breast cancer cells. The GREB1 gene encodes three distinct isoforms: GREB1a, GREB1b and GREB1c, whose molecular functions are largely unknown. Here, we investigate the role of these isoforms in regulation of ERα activity and proliferation. Interaction between GREB1 and ERα was mapped to the amino terminus shared by all GREB1 variants. Analysis of isoform-specific regulation of ERα activity suggests none of the GREB1 isoforms possess potent co-regulator activity. Exogenous expression of GREB1a resulted in elevated expression of some ER-target genes, independent of ERα activity. Despite this slight specificity of GREB1a for gene regulation, exogenous expression of either GREB1a or GREB1b resulted in decreased proliferation in both ER-positive and ER-negative breast carcinoma cell lines, demonstrating an ER-independent function of GREB1. Interestingly, we show an increase in the expression of GREB1b and GREB1c mRNA in malignant breast tissue compared to normal patient samples, suggesting a selective preference for these isoforms during malignant transformation. Together, these data suggest GREB1a has an isoform-specific function as a transcriptional regulator while all isoforms share an ER-independent activity that regulates proliferation.


Subject(s)
Breast Neoplasms/genetics , Estrogen Receptor alpha/metabolism , Neoplasm Proteins/therapeutic use , Protein Isoforms/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Female , Humans , Neoplasm Proteins/pharmacology
8.
Endocrinology ; 158(10): 3435-3447, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28938483

ABSTRACT

In utero exposure to the endocrine disrupting compound bisphenol A (BPA) is known to disrupt mammary gland development and increase tumor susceptibility in rodents. It is unclear whether different periods of in utero development might be more susceptible to BPA exposure. We exposed pregnant CD-1 mice to BPA at different times during gestation that correspond to specific milestones of in utero mammary gland development. The mammary glands of early-life and adult female mice, exposed in utero to BPA, were morphologically and molecularly (estrogen receptor-α and Ki67) evaluated for developmental abnormalities. We found that BPA treatment occurring before mammary bud invasion into the mesenchyme [embryonic day (E)12.5] incompletely resulted in the measured phenotypes of mammary gland defects. Exposing mice up to the point at which the epithelium extends into the precursor fat pad (E16.5) resulted in a nearly complete BPA phenotype and exposure during epithelial extension (E15.5 to E18.5) resulted in a partial phenotype. Furthermore, the relative differences in phenotypes between exposure windows highlight the substantial correlations between early-life molecular changes (estrogen receptor-α and Ki67) in the stroma and the epithelial elongation defects in mammary development. These data further implicate BPA action in the stroma as a critical mediator of epithelial phenotypes.


Subject(s)
Benzhydryl Compounds/pharmacology , Estrogen Receptor alpha/drug effects , Estrogens, Non-Steroidal/pharmacology , Ki-67 Antigen/drug effects , Mammary Glands, Animal/drug effects , Phenols/pharmacology , Prenatal Exposure Delayed Effects , Amniotic Fluid/chemistry , Animals , Chromatography, High Pressure Liquid , Estrogen Receptor alpha/metabolism , Female , Immunohistochemistry , Ki-67 Antigen/metabolism , Mammary Glands, Animal/embryology , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mice , Phenotype , Pregnancy , Time Factors
9.
Clin Immunol ; 176: 12-22, 2017 03.
Article in English | MEDLINE | ID: mdl-28039018

ABSTRACT

Recent studies implicate innate immunity to systemic lupus erythematosus (SLE) pathogenesis. Toll-like receptor (TLR)8 is estrogen-regulated and binds viral ssRNA to stimulate innate immune responses, but recent work indicates that microRNA (miR)-21 within extracellular vesicles (EVs) can also trigger this receptor. Our objective was to examine TLR8 expression/activation to better understand sex-biased responses involving TLR8 in SLE. Our data identify an estrogen response element that promotes STAT1 expression and demonstrate STAT1-dependent transcriptional activation of TLR8 with estrogen stimulation. In lieu of viral ssRNA activation, we explored EV-encapsulated miR-21 as an endogenous ligand and observed induction of both TLR8 and cytokine expression in vitro. Moreover, extracellular miR detection was found predominantly within EVs. Thus, just as a cytokine or chemokine, EV-encapsulated miR-21 can act as an inflammatory signaling molecule, or miRokine, by virtue of being an endogenous ligand of TLR8. Collectively, our data elucidates a novel innate inflammatory pathway in SLE.


Subject(s)
Estrogens/metabolism , Lupus Erythematosus, Systemic/metabolism , MicroRNAs/metabolism , STAT1 Transcription Factor/metabolism , Signal Transduction/physiology , Toll-Like Receptor 8/metabolism , Cell Line, Tumor , Chemokines/metabolism , Humans , Immunity, Innate/immunology , Inflammation/immunology , Inflammation/metabolism , Ligands , Lupus Erythematosus, Systemic/immunology , MCF-7 Cells
10.
PLoS Genet ; 12(8): e1006224, 2016 08.
Article in English | MEDLINE | ID: mdl-27487356

ABSTRACT

Antisense transcription is a prevalent feature at mammalian promoters. Previous studies have primarily focused on antisense transcription initiating upstream of genes. Here, we characterize promoter-proximal antisense transcription downstream of gene transcription starts sites in human breast cancer cells, investigating the genomic context of downstream antisense transcription. We find extensive correlations between antisense transcription and features associated with the chromatin environment at gene promoters. Antisense transcription downstream of promoters is widespread, with antisense transcription initiation observed within 2 kb of 28% of gene transcription start sites. Antisense transcription initiates between nucleosomes regularly positioned downstream of these promoters. The nucleosomes between gene and downstream antisense transcription start sites carry histone modifications associated with active promoters, such as H3K4me3 and H3K27ac. This region is bound by chromatin remodeling and histone modifying complexes including SWI/SNF subunits and HDACs, suggesting that antisense transcription or resulting RNA transcripts contribute to the creation and maintenance of a promoter-associated chromatin environment. Downstream antisense transcription overlays additional regulatory features, such as transcription factor binding, DNA accessibility, and the downstream edge of promoter-associated CpG islands. These features suggest an important role for antisense transcription in the regulation of gene expression and the maintenance of a promoter-associated chromatin environment.


Subject(s)
Antisense Elements (Genetics)/genetics , Nuclear Proteins/genetics , Promoter Regions, Genetic , Transcription, Genetic , Antisense Elements (Genetics)/biosynthesis , Chromatin/genetics , CpG Islands/genetics , Gene Expression Regulation, Fungal , Genomics , Histone Code/genetics , Histones/genetics , Humans , Nuclear Proteins/biosynthesis , Nucleosomes/genetics , Protein Binding/genetics , Sequence Alignment
11.
Mol Endocrinol ; 29(3): 384-95, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25594248

ABSTRACT

The pervasive nature of estrogenic industrial and dietary compounds is a growing health concern linked to cancer, obesity, and neurological disorders. Prior analyses of endocrine disruptor action have focused primarily on the short-term consequences of exposure. However, these studies are unlikely to reflect the consequences of constant exposures common to industrialized countries. Here we examined the global effects of long-term endocrine disruption on gene transcription and estrogen signaling. Estrogen-dependent breast cancer cell lines were chronically treated with physiologically relevant levels of bisphenol A or genistein for more than 70 passages. Microarray analysis demonstrated global reprogramming of the transcriptome when compared with a similarly cultured control cell line. Estrogen-responsive targets showed diminished expression in both the presence and absence of estrogen. Estrogen receptor recruitment, H3K4 monomethylation, and deoxyribonuclease accessibility were reduced at nearby response elements. Based on these observations, we investigated the potential of long-term endocrine disruptor exposure to initiate persistent transcriptional reprogramming. Culture of chronically exposed cell lines in the absence of the endocrine disruptors did not reverse many of the signaling defects that accumulated during treatment. Taken together, these data demonstrate that chronic exposure to endocrine disrupting compounds can permanently alter physiological hormone signaling.


Subject(s)
Endocrine Disruptors/toxicity , Estrogens/pharmacology , Cell Line, Tumor , Early Growth Response Protein 3/genetics , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Genetic Loci , Humans , Ligands , Receptors, Estrogen/metabolism , Signal Transduction/drug effects
12.
Clin Immunol ; 151(1): 66-77, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24525049

ABSTRACT

Females of child-bearing age are more resistant to infectious disease and have an increased risk of systemic lupus erythematosus (SLE). We hypothesized that estrogen-induced gene expression could establish an immunoactivated state which would render enhanced defense against infection, but may be deleterious in autoimmune development. Using peripheral blood mononuclear cells (PBMCs), we demonstrate enhanced responses with immunogen stimulation in the presence of 17ß-estradiol (E2) and gene array analyses reveal toll-like receptor 8 (TLR8) as an E2-responsive candidate gene. TLR8 expression levels are up-regulated in SLE and PBMCs stimulated with TLR8 agonist display a female sex-biased, E2-sensitive response. Moreover, we identify a putative ERα-binding region near the TLR8 locus and blocking ERα expression significantly decreases E2-mediated TLR8 induction. Our findings characterize TLR8 as a novel estrogen target gene that can lower the inflammatory threshold and implicate an IFNα-independent inflammatory mechanism that could contribute to higher SLE incidence in women.


Subject(s)
Endosomes/drug effects , Estradiol/pharmacology , Estrogen Receptor alpha/immunology , Leukocytes, Mononuclear/drug effects , Lupus Erythematosus, Systemic/immunology , Toll-Like Receptor 8/immunology , Animals , Binding Sites , Cell Line, Tumor , Cells, Cultured , Endosomes/immunology , Endosomes/metabolism , Estrogen Receptor alpha/genetics , Female , Gene Expression Regulation , Humans , Imidazoles/pharmacology , Immunologic Factors/pharmacology , Interferon-alpha/immunology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/pathology , Male , Mice , Mice, Inbred C57BL , Protein Binding , Sex Factors , Signal Transduction , Toll-Like Receptor 8/agonists , Toll-Like Receptor 8/genetics
13.
Mol Cell Endocrinol ; 380(1-2): 25-31, 2013 Nov 05.
Article in English | MEDLINE | ID: mdl-23545159

ABSTRACT

The glucocorticoid receptor (GR) functions to regulate a wide group of physiological processes through hormone inducible interaction with genomic loci and subsequent manipulation of the transcriptional output of target genes. Despite expression in a wide variety of tissues, the GR has diverse roles that are regulated tightly in a cell type specific manner. With the advent of whole genome approaches, the details of that diversity and the mechanisms regulating them are beginning to be elucidated. This review aims describe the recent advances detailing the role chromatin structure plays in dictating GR specificity.


Subject(s)
Chromatin/genetics , Glucocorticoids/physiology , Animals , Binding Sites , Chromatin/metabolism , Chromatin Assembly and Disassembly , Genome, Human , Humans , Nucleic Acid Conformation , Promoter Regions, Genetic , Protein Binding , Receptors, Glucocorticoid/physiology
14.
J Clin Invest ; 123(1): 493-508, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23257359

ABSTRACT

Cyclin D1b is a splice variant of the cell cycle regulator cyclin D1 and is known to harbor divergent and highly oncogenic functions in human cancer. While cyclin D1b is induced during disease progression in many cancer types, the mechanisms underlying cyclin D1b function remain poorly understood. Herein, cell and human tumor xenograft models of prostate cancer were utilized to resolve the downstream pathways that are required for the protumorigenic functions of cyclin D1b. Specifically, cyclin D1b was found to modulate the expression of a large transcriptional network that cooperates with androgen receptor (AR) signaling to enhance tumor cell growth and invasive potential. Notably, cyclin D1b promoted AR-dependent activation of genes associated with metastatic phenotypes. Further exploration determined that transcriptional induction of SNAI2 (Slug) was essential for cyclin D1b-mediated proliferative and invasive properties, implicating Slug as a critical driver of disease progression. Importantly, cyclin D1b expression highly correlated with that of Slug in clinical samples of advanced disease. In vivo analyses provided strong evidence that Slug enhances both tumor growth and metastatic phenotypes. Collectively, these findings reveal the underpinning mechanisms behind the protumorigenic functions of cyclin D1b and demonstrate that the convergence of the cyclin D1b/AR and Slug pathways results in the activation of processes critical for the promotion of lethal tumor phenotypes.


Subject(s)
Cyclin D1/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Signal Transduction , Transcription Factors/metabolism , Alternative Splicing/genetics , Animals , Cyclin D1/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Receptors, Androgen/genetics , Snail Family Transcription Factors , Transcription Factors/genetics , Transcriptional Activation/genetics , Transplantation, Heterologous
15.
Mol Cell Biol ; 32(10): 1805-17, 2012 May.
Article in English | MEDLINE | ID: mdl-22451486

ABSTRACT

Steroid hormone receptors initiate a genetic program tightly regulated by the chromatin environment of the responsive regions. Using the glucocorticoid receptor (GR) as a model factor for transcriptional initiation, we classified chromatin structure through formaldehyde-assisted isolation of regulatory elements (FAIRE). We looked at dynamic changes in FAIRE signals during GR activation specifically at regions of receptor interaction. We found a distribution of GR-responsive regions with diverse responses to activation and chromatin modulation. The majority of GR binding regions demonstrate increases in FAIRE signal in response to ligand. However, the majority GR-responsive regions shared a similar FAIRE signal in the basal chromatin state, suggesting a common chromatin structure for GR recruitment. Supporting this notion, global FAIRE sequencing (seq) data indicated an enrichment of signal surrounding the GR binding site prior to activation. Brg-1 knockdown showed response element-specific effects of ATPase-dependent chromatin remodeling. FAIRE induction was universally decreased by Brg-1 depletion, but to varying degrees in a target specific manner. Taken together, these data suggest classes of nuclear receptor response regions that react to activation through different chromatin regulatory events and identify a chromatin structure that classifies the majority of response elements tested.


Subject(s)
Chromatin/metabolism , Receptors, Glucocorticoid/metabolism , Binding Sites , Cell Line, Tumor , Chromatin/chemistry , Chromatin Assembly and Disassembly , Formaldehyde , Humans , Ligands , Protein Binding , Response Elements/physiology
16.
J Biol Chem ; 286(10): 8117-8127, 2011 Mar 11.
Article in English | MEDLINE | ID: mdl-21212260

ABSTRACT

D-type cyclins regulate cellular outcomes in part through cyclin-dependent, kinase-independent mechanisms that modify transcription factor action, and recent in vivo studies showed that cyclin D1 associates with a large number of transcriptional regulators in cells of the retina and breast. Given the frequency of cyclin D1 alterations in cancer, it is imperative to delineate the molecular mechanisms by which cyclin D1 controls key transcription factor networks in human disease. Prostate cancer was used as a paradigm because this tumor type is reliant at all stages of the disease on androgen receptor (AR) signaling, and cyclin D1 has been shown to negatively modulate AR-dependent expression of prostate-specific antigen (KLK3/PSA). Strategies were employed to control cyclin D1 expression under conditions of hormone depletion, and the effect of cyclin D1 on subsequent androgen-dependent gene expression was determined using unbiased gene expression profiling. Modulating cyclin D1 conferred widespread effects on androgen signaling and revealed cyclin D1 to be a selective effector of hormone action. A subset of androgen-induced target genes, known to be directly regulated by AR, was strongly suppressed by cyclin D1. Analyses of AR occupancy at target gene regulatory loci of clinical relevance demonstrated that cyclin D1 limits AR residence after hormone stimulation. Together, these findings reveal a new function for cyclin D1 in controlling hormone-dependent transcriptional outcomes and demonstrate a pervasive role for cyclin D1 in regulating transcription factor dynamics.


Subject(s)
Androgens/metabolism , Cyclin D1/metabolism , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Signal Transduction , Cell Line, Tumor , Cyclin D1/genetics , Genetic Loci/genetics , Humans , Kallikreins/biosynthesis , Kallikreins/genetics , Male , Prostate-Specific Antigen/biosynthesis , Prostate-Specific Antigen/genetics , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics
17.
Clin Cancer Res ; 15(17): 5338-49, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19706803

ABSTRACT

PURPOSE: Alternative CCND1 splicing results in cyclin D1b, which has specialized, protumorigenic functions in prostate not shared by the cyclin D1a (full length) isoform. Here, the frequency, tumor relevance, and mechanisms controlling cyclin D1b were challenged. EXPERIMENTAL DESIGN: First, relative expression of both cyclin D1 isoforms was determined in prostate adenocarcinomas. Second, relevance of the androgen axis was determined. Third, minigenes were created to interrogate the role of the G/A870 polymorphism (within the splice site), and findings were validated in primary tissue. Fourth, the effect of G/A870 on cancer risk was assessed in two large case-control studies. RESULTS: Cyclin D1b is induced in tumors, and a significant subset expressed this isoform in the absence of detectable cyclin D1a. Accordingly, the isoforms showed noncorrelated expression patterns, and hormone status did not alter splicing. Whereas G/A870 was not independently predictive of cancer risk, A870 predisposed for transcript-b production in cells and in normal prostate. The influence of A870 on overall transcript-b levels was relieved in tumors, indicating that aberrations in tumorigenesis likely alter the influence of the polymorphism. CONCLUSIONS: These studies reveal that cyclin D1b is specifically elevated in prostate tumorigenesis. Cyclin D1b expression patterns are distinct from that observed with cyclin D1a. The A870 allele predisposes for transcript-b production in a context-specific manner. Although A870 does not independently predict cancer risk, tumor cells can bypass the influence of the polymorphism. These findings have major implications for the analyses of D-cyclin function in the prostate and provide the foundation for future studies directed at identifying potential modifiers of the G/A870 polymorphism.


Subject(s)
Alternative Splicing/genetics , Cyclin D1/genetics , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/genetics , Alleles , Case-Control Studies , Cyclin D1/metabolism , Genotype , Humans , Male , Polymorphism, Genetic , Prostatic Neoplasms/pathology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Tissue Array Analysis
18.
J Biol Chem ; 283(50): 34976-82, 2008 Dec 12.
Article in English | MEDLINE | ID: mdl-18922793

ABSTRACT

Dermatomyositis (DM) is an autoimmune disease, which is often accompanied by the development of disease-specific autoantibodies directed against the SNF2-superfamily helicase, Mi-2. Recent evidence suggests that ultraviolet radiation exposure may be an important risk factor for the development of not only the disease but also specific autoimmunity against Mi-2. Consequently, we investigated the effects of ultraviolet radiation on Mi-2 protein expression. We observed an increase in protein levels upon ultraviolet radiation exposure in cell culture systems. These changes in expression occur quite rapidly, are maximized just 1 h following exposure, and are unique to Mi-2 when compared with other members of the NuRD complex. Changes in protein levels are not mediated through transcriptional mechanisms. Treatment results in a more efficiently translated message through regulatory elements in the 5'-UTR region of the transcript. Investigation into protein half-life further demonstrated increased stability of Mi-2 following UV exposure. Taken together, we describe a system by which Mi-2 protein expression can be quickly increased following UV exposure and then maintained up to 16 h later. These data provide a novel regulation of an important transcriptional regulator and provide insight into the possible mechanisms of the development of DM and associated autoantibodies.


Subject(s)
Autoantigens/metabolism , DNA Helicases/metabolism , Protein Biosynthesis/radiation effects , Ultraviolet Rays , 5' Untranslated Regions , Autoantibodies/chemistry , Cell Line, Tumor , Cell Separation , Flow Cytometry , Humans , Keratinocytes/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex , Models, Biological , Plasmids/metabolism , Risk Factors , Time Factors , Transcription, Genetic
19.
Cancer Res ; 68(14): 5628-38, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18632615

ABSTRACT

Cyclin D1 is a key mediator of cell cycle progression that is aberrantly regulated in multiple cancers, especially in breast cancers. A number of studies have indicated that a polymorphism in a splice donor site in the cyclin D1 gene is associated with alternative splicing and the production of the alternative cyclin D1b transcript. Furthermore, this polymorphism is selectively associated with disease outcomes. However, relatively little is known regarding the protein product of the alternatively spliced message, cyclin D1b. Using antibodies specific for cyclin D1b, it was found that this protein is readily detectable in a number of cancer cell lines and primary breast cancers. Whereas cyclin D1b interacts with cyclin-dependent kinase 4 (CDK4), it is relatively inefficient at mediating RB phosphorylation and cell cycle progression in model systems due to the lack of exon 5 of cyclin D1-encoded sequences. However, cyclin D1b protein levels are not significantly attenuated by DNA damage or antiestrogen treatment, indicating that the protein may have significant effect on the response to such therapeutic modalities. Whereas enforced expression of cyclin D1b was not sufficient to abrogate DNA damage checkpoint responses, it did efficiently overcome cell cycle arrest mediated by antiestrogen therapeutics. This action of cyclin D1b was not associated with effects on estrogen receptor activity, but was rather dependent on functional association with CDK4. Combined, these studies indicate that the cyclin D1b protein is aberrantly regulated and could contribute to therapeutic failure in the context of ER-positive breast cancer.


Subject(s)
Cyclins/genetics , Cyclins/physiology , Drug Resistance, Neoplasm , Estrogen Antagonists/metabolism , Gene Expression Regulation, Neoplastic , Receptors, Estrogen/metabolism , Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cisplatin/pharmacology , Cyclin D , Cyclin-Dependent Kinase 4/metabolism , Gene Expression Profiling , HeLa Cells , Humans , Phosphorylation , Polymorphism, Genetic
20.
Endocr Relat Cancer ; 13(4): 979-94, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17158750

ABSTRACT

The androgen receptor (AR) mediates the effects of male steroid hormones (androgens) and contributes to a wide variety of physiological and pathophysiological conditions. As such, the regulatory mechanisms governing AR activity are of high significance. Concerted effort has been placed on delineating the mechanisms that control AR activity in prostate cancer, as AR is required for survival and proliferation in this tumor type. Moreover, AR is the central therapeutic target for metastatic prostate cancers, and recurrent tumors evade therapy by restoring AR activity. It is increasingly apparent that AR cofactors which modulate receptor activity can contribute to prostate cancer growth or progression, and this has been particularly well established for AR coactivators. The present review is focused on the role of AR corepressors in governing androgen action, with a specific emphasis on their activities in prostate cancer.


Subject(s)
Prostatic Neoplasms/metabolism , Receptors, Androgen/physiology , Repressor Proteins/physiology , Humans , Male , Prostatic Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...