Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
Mol Cell ; 70(4): 679-694.e7, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29775582

ABSTRACT

Enhancers are thought to activate transcription by physically contacting promoters via looping. However, direct assays demonstrating these contacts are required to mechanistically verify such cellular determinants of enhancer function. Here, we present versatile cell-free assays to further determine the role of enhancer-promoter contacts (EPCs). We demonstrate that EPC is linked to mutually stimulatory transcription at the enhancer and promoter in vitro. SRC-3 was identified as a critical looping determinant for the estradiol-(E2)-regulated GREB1 locus. Surprisingly, the GREB1 enhancer and promoter contact two internal gene body SRC-3 binding sites, GBS1 and GBS2, which stimulate their transcription. Utilizing time-course 3C assays, we uncovered SRC-3-dependent dynamic chromatin interactions involving the enhancer, promoter, GBS1, and GBS2. Collectively, these data suggest that the enhancer and promoter remain "poised" for transcription via their contacts with GBS1 and GBS2. Upon E2 induction, GBS1 and GBS2 disengage from the enhancer, allowing direct EPC for active transcription.


Subject(s)
Breast Neoplasms/genetics , Chromatin/metabolism , Estrogens/pharmacology , Gene Expression Regulation, Neoplastic , Nuclear Receptor Coactivator 3/metabolism , Transcription, Genetic , Binding Sites , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Chromatin/genetics , Enhancer Elements, Genetic , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Humans , Nuclear Receptor Coactivator 3/genetics , Promoter Regions, Genetic , Protein Binding , Tumor Cells, Cultured
2.
Proc Natl Acad Sci U S A ; 120(4): e2216436120, 2023 01 24.
Article in English | MEDLINE | ID: mdl-36656865

ABSTRACT

Enhancers not only activate target promoters to stimulate messenger RNA (mRNA) synthesis, but they themselves also undergo transcription to produce enhancer RNAs (eRNAs), the significance of which is not well understood. Transcription at the participating enhancer-promoter pair appears coordinated, but it is unclear why and how. Here, we employ cell-free transcription assays using constructs derived from the human GREB1 locus to demonstrate that transcription at an enhancer and its target promoter is interdependent. This interdependence is observable under conditions where direct enhancer-promoter contact (EPC) takes place. We demonstrate that transcription activation at a participating enhancer-promoter pair is dependent on i) the mutual availability of the enhancer and promoter, ii) the state of transcription at both the enhancer and promoter, iii) local abundance of both eRNA and mRNA, and iv) direct EPC. Our results suggest transcriptional interdependence between the enhancer and the promoter as the basis of their transcriptional concurrence and coordination throughout the genome. We propose a model where transcriptional concurrence, coordination and interdependence are possible if the participating enhancer and promoter are entangled in the form of EPC, reside in a proteinaceous bubble, and utilize shared transcriptional resources and regulatory inputs.


Subject(s)
Enhancer Elements, Genetic , RNA , Humans , Enhancer Elements, Genetic/genetics , Promoter Regions, Genetic/genetics , RNA/genetics , RNA, Messenger/genetics , Transcriptional Activation , Transcription, Genetic , Gene Expression Regulation
3.
Proc Natl Acad Sci U S A ; 120(23): e2221707120, 2023 06 06.
Article in English | MEDLINE | ID: mdl-37253006

ABSTRACT

Steroid receptor coactivator 3 (SRC-3) is most strongly expressed in regulatory T cells (Tregs) and B cells, suggesting that it plays an important role in the regulation of Treg function. Using an aggressive E0771 mouse breast cell line syngeneic immune-intact murine model, we observed that breast tumors were "permanently eradicated" in a genetically engineered tamoxifen-inducible Treg-cell-specific SRC-3 knockout (KO) female mouse that does not possess a systemic autoimmune pathological phenotype. A similar eradication of tumor was noted in a syngeneic model of prostate cancer. A subsequent injection of additional E0771 cancer cells into these mice showed continued resistance to tumor development without the need for tamoxifen induction to produce additional SRC-3 KO Tregs. SRC-3 KO Tregs were highly proliferative and preferentially infiltrated into breast tumors by activating the chemokine (C-C motif) ligand (Ccl) 19/Ccl21/chemokine (C-C motif) receptor (Ccr)7 signaling axis, generating antitumor immunity by enhancing the interferon-γ/C-X-C motif chemokine ligand (Cxcl) 9 signaling axis to facilitate the entrance and function of effector T cells and natural killer cells. SRC-3 KO Tregs also show a dominant effect by blocking the immune suppressive function of WT Tregs. Importantly, a single adoptive transfer of SRC-3 KO Tregs into wild-type E0771 tumor-bearing mice can completely abolish preestablished breast tumors by generating potent antitumor immunity with a durable effect that prevents tumor reoccurrence. Therefore, treatment with SRC-3-deleted Tregs represents an approach to completely block tumor growth and recurrence without the autoimmune side effects that typically accompany immune checkpoint modulators.


Subject(s)
Breast Neoplasms , Mammary Neoplasms, Animal , Nuclear Receptor Coactivator 3 , Animals , Female , Male , Mice , Ligands , Mice, Knockout , Nuclear Receptor Coactivator 3/genetics , T-Lymphocytes, Regulatory , Tamoxifen/pharmacology
4.
J Mol Cell Cardiol ; 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38969334

ABSTRACT

We recently discovered that steroid receptor coactivators (SRCs) SRCs-1, 2 and 3, are abundantly expressed in cardiac fibroblasts (CFs) and their activation with the SRC small molecule stimulator MCB-613 improves cardiac function and dramatically lowers pro-fibrotic signaling in CFs post-myocardial infarction. These findings suggest that CF-derived SRC activation could be beneficial in the mitigation of chronic heart failure after ischemic insult. However, the cardioprotective mechanisms by which CFs contribute to cardiac pathological remodeling are unclear. Here we present studies designed to identify the molecular and cellular circuitry that governs the anti-fibrotic effects of an MCB-613 derivative, MCB-613-10-1, in CFs. We performed cytokine profiling and whole transcriptome and proteome analyses of CF-derived signals in response to MCB-613-10-1. We identified the NRF2 pathway as a direct MCB-613-10-1 therapeutic target for promoting resistance to oxidative stress in CFs. We show that MCB-613-10-1 promotes cell survival of anti-fibrotic CFs exposed to oxidative stress by suppressing apoptosis. We demonstrate that an increase in HMOX1 expression contributes to CF resistance to oxidative stress-mediated apoptosis via a mechanism involving SRC co-activation of NRF2, hence reducing inflammation and fibrosis. We provide evidence that MCB-613-10-1 acts as a protectant against oxidative stress-induced mitochondrial damage. Our data reveal that SRC stimulation of the NRF2 transcriptional network promotes resistance to oxidative stress and highlights a mechanistic approach toward addressing pathologic cardiac remodeling.

5.
FASEB J ; 37(12): e23313, 2023 12.
Article in English | MEDLINE | ID: mdl-37962238

ABSTRACT

Although we have shown that steroid receptor coactivator-2 (SRC-2), a member of the p160/SRC family of transcriptional coregulators, is essential for decidualization of both human and murine endometrial stromal cells, SRC-2's role in the earlier stages of the implantation process have not been adequately addressed. Using a conditional SRC-2 knockout mouse (SRC-2d/d ) in timed natural pregnancy studies, we show that endometrial SRC-2 is required for embryo attachment and adherence to the luminal epithelium. Implantation failure is associated with the persistent expression of Mucin 1 and E-cadherin on the apical surface and basolateral adherens junctions of the SRC-2d/d luminal epithelium, respectively. These findings indicate that the SRC-2d/d luminal epithelium fails to exhibit a plasma membrane transformation (PMT) state known to be required for the development of uterine receptivity. Transcriptomics demonstrated that the expression of genes involved in steroid hormone control of uterine receptivity were significantly disrupted in the SRC-2d/d endometrium as well as genes that control epithelial tight junctional biology and the emergence of the epithelial mesenchymal transition state, with the latter sharing similar biological properties with PMT. Collectively, these findings uncover a new role for endometrial SRC-2 in the induction of the luminal epithelial PMT state, which is a prerequisite for the development of uterine receptivity and early pregnancy establishment.


Subject(s)
Embryo Implantation , Uterus , Animals , Female , Humans , Mice , Pregnancy , Embryo Implantation/genetics , Endometrium/metabolism , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Mice, Knockout , Nuclear Receptor Coactivator 2/genetics , Uterus/metabolism
6.
Proc Natl Acad Sci U S A ; 117(49): 31353-31364, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33229578

ABSTRACT

Progressive remodeling of the heart, resulting in cardiomyocyte (CM) loss and increased inflammation, fibrosis, and a progressive decrease in cardiac function, are hallmarks of myocardial infarction (MI)-induced heart failure. We show that MCB-613, a potent small molecule stimulator of steroid receptor coactivators (SRCs) attenuates pathological remodeling post-MI. MCB-613 decreases infarct size, apoptosis, hypertrophy, and fibrosis while maintaining significant cardiac function. MCB-613, when given within hours post MI, induces lasting protection from adverse remodeling concomitant with: 1) inhibition of macrophage inflammatory signaling and interleukin 1 (IL-1) signaling, which attenuates the acute inflammatory response, 2) attenuation of fibroblast differentiation, and 3) promotion of Tsc22d3-expressing macrophages-all of which may limit inflammatory damage. SRC stimulation with MCB-613 (and derivatives) is a potential therapeutic approach for inhibiting cardiac dysfunction after MI.


Subject(s)
Cyclohexanones/pharmacology , Myocardial Infarction/physiopathology , Pyridines/pharmacology , Receptors, Steroid/metabolism , Ventricular Remodeling/drug effects , Animals , Cell Differentiation/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis , Heart Function Tests , Inflammation/pathology , Macrophages/drug effects , Macrophages/pathology , Mice , Myocardial Infarction/genetics , Myocardial Infarction/pathology , RAW 264.7 Cells , RNA/genetics , RNA/metabolism , Transcription, Genetic/drug effects
7.
Breast Cancer Res ; 24(1): 73, 2022 10 31.
Article in English | MEDLINE | ID: mdl-36316775

ABSTRACT

BACKGROUND: The tumor immune microenvironment (TIME) generated by cancer-infiltrating immune cells has a crucial role in promoting or suppressing breast cancer progression. However, whether the steroid receptor coactivator-3 (SRC-3) modulates TIME to progress breast cancer is unclear. Therefore, the present study evaluates whether SRC-3 generates a tumor-promoting TIME in breast tumors using a syngeneic immune-intact mouse model of breast cancer. METHODS: We employed E0771 and 4T1 breast cancer in immune-intact syngeneic female C57BL/6 and BALB/c mice, respectively. SI-2, a specific small-molecule inhibitor of SRC-3, was administered daily (2.5 mg/kg) to E0771 and 4T1 breast tumor-bearing immune-intact mice. In addition, SRC-3 knockdown (KD)-E0771 and SRC-3 KD-4T1 cells and their parental breast cancer cells were injected into their syngeneic immune-intact female mice versus immune-deficiency mice to validate that the host immune system is required for breast tumor suppression by SRC-3 KD in immune-intact mice. Furthermore, tumor-infiltrating immune cells (such as CD4+, CD8+, CD56+, and Foxp3+ cells) in E0771 and 4T1 breast cancers treated with SI-2 and in SRC-3 KD E0771 and 4T1 breast cancers were determined by immunohistochemistry. Additionally, cytokine levels in SI-2-treated and SRC-3 KD E0771 breast tumors and their control cancers were defined with a Mouse Cytokine Array. RESULTS: SRC-3 inhibition by SI-2 significantly suppressed the progression of breast cancer cells (E0771 and 4T1) into breast cancers in immune-intact syngeneic female mice. SRC-3 KD-E0771 and -4T1 breast cancer cells did not produce well-developed tumors in immune-intact syngeneic female mice compared to their parental cells, but SRC-3 KD breast cancers were well developed in immune-defective host mice. SRC-3 inhibition by SI-2 and SRC-3 KD effectively increased the numbers of cytotoxic immune cells, such as CD4+ and CD8+ T cells and CD56+ NK cells, and Interferon γ (Ifng) in breast cancers compared to vehicle. However, SI-2 treatment reduced the number of tumor-infiltrating CD4+/Foxp3+ regulatory T (Treg) cells compared to vehicle treatment. In addition, SRC-3 inhibition by SI-2 and SRC-3 KD increased C-X-C motif chemokine ligand 9 (Cxcl9) expression in breast cancer to recruit C-X-C motif chemokine receptor 3 (Cxcr3)-expressing cytotoxic immune cells into breast tumors. CONCLUSIONS: SRC-3 is a critical immunomodulator in breast cancer, generating a protumor immune microenvironment. SRC-3 inhibition by SI-2 or SRC-3 KD activates the Cxcl9/Cxcr3 axis in breast tumors and enhances the antitumor immune microenvironment to suppress breast cancer progression.


Subject(s)
Neoplasms , Nuclear Receptor Coactivator 3 , Tumor Microenvironment , Animals , Female , Mice , Cell Line, Tumor , Cytokines/metabolism , Forkhead Transcription Factors , Mice, Inbred BALB C , Mice, Inbred C57BL , Nuclear Receptor Coactivator 3/metabolism
8.
Reproduction ; 164(2): 41-54, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35679138

ABSTRACT

Although a non-malignant gynecological disorder, endometriosis displays some pathogenic features of malignancy, such as cell proliferation, migration, invasion and adaptation to hypoxia. Current treatments of endometriosis include pharmacotherapy and/or surgery, which are of limited efficacy and often associated with adverse side effects. Therefore, to develop more effective therapies to treat this disease, a broader understanding of the underlying molecular mechanisms that underpin endometriosis needs to be attained. Using immortalized human endometriotic epithelial and stromal cell lines, we demonstrate that the early growth response 1 (EGR1) transcription factor is essential for cell proliferation, migration and invasion, which represent some of the pathogenic properties of endometriotic cells. Genome-wide transcriptomics identified an EGR1-dependent transcriptome in human endometriotic epithelial cells that potentially encodes a diverse spectrum of proteins that are known to be involved in tissue pathologies. To underscore the utility of this transcriptomic data set, we demonstrate that carbonic anhydrase 9 (CA9), a homeostatic regulator of intracellular pH, is not only a molecular target of EGR1 but is also important for maintaining many of the cellular properties of human endometriotic epithelial cells that are also ascribed to EGR1. Considering therapeutic intervention strategies are actively being developed for EGR1 and CAIX in the treatment of other pathologies, we believe EGR1 and its transcriptome (which includes CA9) will offer not only a new conceptual framework to advance our understanding of endometriosis but will also furnish new molecular vulnerabilities to be leveraged as potential therapeutic options in the future treatment of endometriosis.


Subject(s)
Early Growth Response Protein 1 , Endometriosis , Cell Movement , Early Growth Response Protein 1/genetics , Endometriosis/metabolism , Endometrium/metabolism , Epithelial Cells/metabolism , Female , Humans , Stromal Cells/metabolism , Transcription Factors/metabolism
9.
Mol Cell ; 51(2): 185-99, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-23850489

ABSTRACT

Chromatin immunoprecipitation studies have mapped protein occupancies at many genomic loci. However, a detailed picture of the complexity of coregulators (CoRs) bound to a defined enhancer along with a transcription factor is missing. To address this, we used biotin-DNA pull-down assays coupled with mass spectrometry-immunoblotting to identify at least 17 CoRs from nuclear extracts bound to 17ß-estradiol (E2)-liganded estrogen receptor-α on estrogen response elements (EREs). Unexpectedly, these complexes initially are biochemically stable and contain certain atypical corepressors. Addition of ATP dynamically converts these complexes to an "activated" state by phosphorylation events, primarily mediated by DNA-dependent protein kinase. Importantly, a "natural" ERE-containing enhancer and nucleosomal EREs recruit similar complexes. We further discovered the mechanism whereby H3K4me3 stimulates ERα-mediated transcription as compared with unmodified nucleosomes. H3K4me3 templates promote specific CoR dynamics in the presence of ATP and AcCoA, as manifested by CBP/p300 and SRC-3 dismissal and SAGA and TFIID stabilization/recruitment.


Subject(s)
Breast Neoplasms/metabolism , DNA-Binding Proteins/metabolism , DNA/metabolism , Estrogen Receptor alpha/metabolism , Gene Expression Regulation, Neoplastic , Nucleosomes/metabolism , Proteomics , Response Elements/genetics , Breast Neoplasms/genetics , Chromatin Immunoprecipitation , DNA/genetics , DNA-Activated Protein Kinase/genetics , DNA-Activated Protein Kinase/metabolism , DNA-Binding Proteins/genetics , E1A-Associated p300 Protein/genetics , E1A-Associated p300 Protein/metabolism , Estrogen Receptor alpha/genetics , Estrogens/pharmacology , Female , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , HeLa Cells , Humans , MCF-7 Cells , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Receptor Coactivator 3/genetics , Nuclear Receptor Coactivator 3/metabolism , Nucleosomes/genetics , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phosphorylation , Promoter Regions, Genetic , Sialoglycoproteins/genetics , Sialoglycoproteins/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Trans-Activators , Transcription, Genetic , Transcriptional Activation
10.
Genes Dev ; 27(3): 274-87, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23388826

ABSTRACT

Steroid receptor coactivator 3 (SRC-3) is an oncogenic nuclear receptor coactivator that plays a significant role in drug resistance. Using a lentiviral cDNA library rescue screening approach, we identified a SRC-3 downstream gene-TRAF4 (tumor necrosis factor [TNF] receptor associated-factor 4)-that functions in cell resistance to cytotoxic stress. TRAF4 expression is positively correlated with SRC-3 expression in human breast cancers. Similar to that observed for SRC-3 overexpression, breast cancer cells overexpressing TRAF4 are more resistant to stress-induced death. Here, we further dissected the underlying molecular mechanism for SRC-3 and TRAF4-mediated resistance to cytotoxic agents. We observed that SRC-3 expression is inversely correlated with the expression of p53-regulated proapoptotic genes in breast cancers and further found that SRC-3 and TRAF4 overexpression diminished cytotoxic stress-induced up-regulation of the tumor suppressor p53 protein. To determine the mechanism, we showed that the TRAF domain of TRAF4 bound to the N-terminal TRAF-like region of the deubiquitinase HAUSP (herpesvirus-associated ubiquitin-specific protease; also named USP7) and blocked the access of p53 to the same region of HAUSP. This TRAF4-mediated inhibition of HAUSP then led to the loss of p53 deubiquitination and its stabilization in response to cellular stress. Consistent with this cellular function, we also found that TRAF4 overexpression in breast cancer patients was associated significantly with poor prognosis. Because of SRC-3's ability to abrogate p53 function, our results suggest that SRC-3 overexpression may be especially important in tumors in which p53 is not mutated.


Subject(s)
Nuclear Receptor Coactivator 3/metabolism , Stress, Physiological/physiology , TNF Receptor-Associated Factor 4/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Endopeptidases/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , HEK293 Cells , Humans , Mice , Nuclear Receptor Coactivator 3/genetics , Protein Binding , Protein Stability , Ubiquitin-Specific Peptidase 7 , Ubiquitin-Specific Proteases , Ubiquitination
11.
Cytokine ; 123: 154745, 2019 11.
Article in English | MEDLINE | ID: mdl-31226438

ABSTRACT

Although salivary gland cancers comprise only ∼3-6% of head and neck cancers, treatment options for patients with advanced-stage disease are limited. Because of their rarity, salivary gland malignancies are understudied compared to other exocrine tissue cancers. The comparative lack of progress in this cancer field is particularly evident when it comes to our incomplete understanding of the key molecular signals that are causal for the development and/or progression of salivary gland cancers. Using a novel conditional transgenic mouse (K5:RANKL), we demonstrate that Receptor Activator of NFkB Ligand (RANKL) targeted to cytokeratin 5-positive basal epithelial cells of the salivary gland causes aggressive tumorigenesis within a short period of RANKL exposure. Genome-wide transcriptomic analysis reveals that RANKL markedly increases the expression levels of numerous gene families involved in cellular proliferation, migration, and intra- and extra-tumoral communication. Importantly, cross-species comparison of the K5:RANKL transcriptomic dataset with The Cancer Genome Atlas cancer signatures reveals the strongest molecular similarity with cancer subtypes of the human head and neck squamous cell carcinoma. These studies not only provide a much needed transcriptomic resource to mine for novel molecular targets for therapy and/or diagnosis but validates the K5:RANKL transgenic as a preclinical model to further investigate the in vivo oncogenic role of RANKL signaling in salivary gland tumorigenesis.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasm Proteins/metabolism , RANK Ligand/metabolism , Salivary Gland Neoplasms/metabolism , Salivary Glands/metabolism , Transcription, Genetic , Transcriptome , Animals , Epithelium/metabolism , Epithelium/pathology , Mice , Mice, Transgenic , Neoplasm Proteins/genetics , RANK Ligand/genetics , Salivary Gland Neoplasms/genetics , Salivary Gland Neoplasms/pathology , Salivary Glands/pathology
12.
Proc Natl Acad Sci U S A ; 113(18): 4970-5, 2016 May 03.
Article in English | MEDLINE | ID: mdl-27084884

ABSTRACT

Protein-protein interactions (PPIs) play a central role in most biological processes, and therefore represent an important class of targets for therapeutic development. However, disrupting PPIs using small-molecule inhibitors (SMIs) is challenging and often deemed as "undruggable." We developed a cell-based functional assay for high-throughput screening to identify SMIs for steroid receptor coactivator-3 (SRC-3 or AIB1), a large and mostly unstructured nuclear protein. Without any SRC-3 structural information, we identified SI-2 as a highly promising SMI for SRC-3. SI-2 meets all of the criteria of Lipinski's rule [Lipinski et al. (2001) Adv Drug Deliv Rev 46(1-3):3-26] for a drug-like molecule and has a half-life of 1 h in a pharmacokinetics study and a reasonable oral availability in mice. As a SRC-3 SMI, SI-2 can selectively reduce the transcriptional activities and the protein concentrations of SRC-3 in cells through direct physical interactions with SRC-3, and selectively induce breast cancer cell death with IC50 values in the low nanomolar range (3-20 nM), but not affect normal cell viability. Furthermore, SI-2 can significantly inhibit primary tumor growth and reduce SRC-3 protein levels in a breast cancer mouse model. In a toxicology study, SI-2 caused minimal acute cardiotoxicity based on a hERG channel blocking assay and an unappreciable chronic toxicity to major organs based on histological analyses. We believe that this work could significantly improve breast cancer treatment through the development of "first-in-class" drugs that target oncogenic coactivators.


Subject(s)
Benzimidazoles/pharmacology , Hydrazones/pharmacology , Nuclear Receptor Coactivator 3/antagonists & inhibitors , Animals , Benzimidazoles/pharmacokinetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Hydrazones/pharmacokinetics , Mice , Nuclear Receptor Coactivator 3/metabolism , Protein Processing, Post-Translational/drug effects , Transcription, Genetic/drug effects
13.
Biol Reprod ; 98(6): 856-869, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29546395

ABSTRACT

Speckle-type poz protein (SPOP) is an E3-ubiquitin ligase adaptor for turnover of a diverse number of proteins involved in key cellular processes such as chromatin remodeling, transcriptional regulation, and cell signaling. Genomic analysis revealed that SPOP somatic mutations are found in a subset of endometrial cancers, suggesting that these mutations act as oncogenic drivers of this gynecologic malignancy. These studies also raise the question as to the role of wild-type SPOP in normal uterine function. To address this question, we generated a mouse model (Spopd/d) in which SPOP is ablated in uterine cells that express the PGR. Fertility studies demonstrated that SPOP is required for embryo implantation and for endometrial decidualization. Molecular analysis revealed that expression levels of the PGR at the protein and transcript level are significantly reduced in the Spopd/d uterus. While this result was unexpected, this finding explains in part the dysfunctional phenotype of the Spopd/d uterus. Moderate increased levels of the ESR1, GATA2, and SRC2 were detected in the Spopd/d uterus, suggesting that SPOP is required to maintain the proteome for normal uterine function. With age, the Spopd/d endometrium exhibits large glandular cysts with foci of epithelial proliferation, further supporting a role for SPOP in maintaining a healthy uterus. Collectively, studies on the Spopd/d mouse support an important role for SPOP in normal uterine function and suggest that this mouse model may prove useful to study the role of SPOP-loss-of-function mutations in the etiopathogenesis of endometrial cancer.


Subject(s)
Embryo Implantation/physiology , Infertility/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Uterus/metabolism , Animals , Endometrium/metabolism , Estrogen Receptor alpha/metabolism , Female , GATA2 Transcription Factor/metabolism , Infertility/genetics , Mice , Mice, Knockout , Nuclear Proteins/genetics , Pregnancy , Repressor Proteins/genetics , Ubiquitin-Protein Ligase Complexes
14.
Biol Reprod ; 98(1): 15-27, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29186366

ABSTRACT

Infertility and early embryo miscarriage is linked to inadequate endometrial decidualization. Although transcriptional reprogramming is known to drive decidualization in response to progesterone, the key signaling effectors that directly mediate this hormone response are not fully known. This knowledge gap is clinically significant because identifying the early signals that directly mediate progesterone-driven decidualization will address some of the current limitations in diagnosing and therapeutically treating patients at most risk for early pregnancy loss. We recently revealed that the promyelocytic leukemia zinc finger (PLZF) is a direct target of the progesterone receptor and is essential for decidualization of human endometrial stromal cells (hESCs). The purpose of this current work was to identify the genome-wide transcriptional program that is controlled by PLZF during hESC decidualization using an established in vitro hESC culture model, siRNA-mediated knockdown methods, and RNA-sequencing technology followed by bioinformatic analysis and validation. We discovered that PLZF is critical in the regulation of genes that are involved in cellular processes that are essential for the archetypal morphological and functional changes that occur when hESCs transform into epithelioid decidual cells such as proliferation and cell motility. We predict that the transcriptome datasets identified in this study will not only contribute to a broader understanding of PLZF-dependent endometrial decidualization at the molecular level but may advance the development of more effective molecular diagnostics and therapeutics for the clinical management of female infertility and subfertility that is based on a dysfunctional endometrium.


Subject(s)
Decidua/physiology , Endometrium/cytology , Promyelocytic Leukemia Zinc Finger Protein/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Computational Biology , Female , Gene Expression Regulation/physiology , Humans , Promyelocytic Leukemia Zinc Finger Protein/genetics , RNA Interference , Stromal Cells/cytology , Stromal Cells/metabolism , Transcriptome
15.
Annu Rev Med ; 65: 279-92, 2014.
Article in English | MEDLINE | ID: mdl-24111892

ABSTRACT

Transcriptional coregulators (coactivators and corepressors) have emerged as the principal modulators of the functions of nuclear receptors and other transcription factors. During the decade since the discovery of steroid receptor coactivator-1 (SRC-1), the first authentic coregulator, more than 400 coregulators have been identified and characterized, and deciphering their function has contributed significantly to our understanding of their role in human physiology. Deregulated expression of coregulators has been implicated in diverse disease states and related pathologies. The advancement of molecular technologies has enabled us to better characterize the molecular associations of the SRC family of coactivators with other protein complexes in the context of gene regulation. These continuing discoveries not only expand our knowledge of the roles of coactivators in various human diseases but allow us to discover novel coactivator-targeting strategies for therapeutic intervention in these diseases.


Subject(s)
Energy Metabolism , Neoplasms/metabolism , Nuclear Receptor Coactivators/physiology , Reproduction , Gene Expression Regulation , Humans , Neoplasm Metastasis , Neoplasms/pathology , Nuclear Receptor Coactivators/metabolism , Protein Conformation , Protein Processing, Post-Translational
16.
Mol Cell ; 29(4): 465-76, 2008 Feb 29.
Article in English | MEDLINE | ID: mdl-18313384

ABSTRACT

SRC-3/AIB1 is a steroid receptor coactivator with potent growth-promoting activity, and its overexpression is sufficient to induce tumorigenesis. Previous studies indicate that the cellular level of SRC-3 is tightly regulated by both ubiquitin-dependent and ubiquitin-independent proteasomal degradation pathways. Atypical protein kinase C (aPKC) is frequently overexpressed in cancers. In the present study, we show that aPKC phosphorylates and specifically stabilizes SRC-3 in a selective ER-dependent manner. We further demonstrate that an acidic residue-rich region in SRC-3 is an important determinant for aPKC-mediated phosphorylation and stabilization. The mechanism of the aPKC-mediated stabilization appears due to a decreased interaction between SRC-3 and the C8 subunit of the 20S core proteasome, thus preventing SRC-3 degradation. Our results demonstrate a potent signaling mechanism for regulating SRC-3 levels in cells by coordinate enzymatic inhibition of both ubiquitin-dependent and ubiquitin-independent proteolytic pathways.


Subject(s)
Histone Acetyltransferases/metabolism , Isoenzymes/metabolism , Protein Kinase C/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Breast Neoplasms/metabolism , Cell Line , Endoplasmic Reticulum/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Female , Gene Expression Regulation , Histone Acetyltransferases/chemistry , Histone Acetyltransferases/genetics , Humans , Isoenzymes/genetics , Mice , Molecular Sequence Data , Nuclear Receptor Coactivator 3 , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Protein Kinase C/genetics , Protein Subunits/genetics , Protein Subunits/metabolism , Sequence Alignment , Trans-Activators/chemistry , Trans-Activators/genetics , Transcription Factors/genetics
17.
Proc Natl Acad Sci U S A ; 110(17): 6997-7002, 2013 Apr 23.
Article in English | MEDLINE | ID: mdl-23559371

ABSTRACT

The p160 steroid receptor coactivators (SRCs) SRC-1, SRC-2 [nuclear receptor coactivator (NCOA)2], and SRC-3 [amplified in breast cancer 1 (AIB1)/NCOA3] are key pleiotropic "master regulators" of transcription factor activity necessary for cancer cell proliferation, survival, metabolism, and metastasis. SRC overexpression and overactivation occur in numerous human cancers and are associated with poor clinical outcomes and resistance to therapy. In prostate cancer (PC), the p160 SRCs play critical roles in androgen receptor transcriptional activity, cell proliferation, and resistance to androgen deprivation therapy. We recently demonstrated that the E3 ubiquitin ligase adaptor speckle-type poxvirus and zinc finger (POZ) domain protein (SPOP) interacts directly with SRC-3 and promotes its cullin 3-dependent ubiquitination and proteolysis in breast cancer, thus functioning as a potential tumor suppressor. Interestingly, somatic heterozygous missense mutations in the SPOP substrate-binding cleft recently were identified in up to 15% of human PCs (making SPOP the gene most commonly affected by nonsynonymous point mutations in PC), but their contribution to PC pathophysiology remains unknown. We now report that PC-associated SPOP mutants cannot interact with SRC-3 protein or promote its ubiquitination and degradation. Our data suggest that wild-type SPOP plays a critical tumor suppressor role in PC cells, promoting the turnover of SRC-3 protein and suppressing androgen receptor transcriptional activity. This tumor suppressor effect is abrogated by the PC-associated SPOP mutations. These studies provide a possible explanation for the role of SPOP mutations in PC, and highlight the potential of SRC-3 as a therapeutic target in PC.


Subject(s)
Gene Expression Regulation/physiology , Nuclear Proteins/genetics , Nuclear Receptor Coactivator 3/metabolism , Prostatic Neoplasms/genetics , Repressor Proteins/genetics , Analysis of Variance , Cell Line, Tumor , Electrophoresis, Polyacrylamide Gel , Genetic Vectors/genetics , HEK293 Cells , Humans , Immunoblotting , Immunoprecipitation , Lentivirus , Male , Mutation, Missense/genetics , Nuclear Proteins/metabolism , Prostatic Neoplasms/physiopathology , Real-Time Polymerase Chain Reaction , Receptors, Androgen/metabolism , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tetrazolium Salts , Thiazoles
18.
Front Immunol ; 15: 1389041, 2024.
Article in English | MEDLINE | ID: mdl-38698860

ABSTRACT

Steroid receptor coactivators (SRCs) are master regulators of transcription that play key roles in human physiology and pathology. SRCs are particularly important for the regulation of the immune system with major roles in lymphocyte fate determination and function, macrophage activity, regulation of nuclear factor κB (NF-κB) transcriptional activity and other immune system biology. The three members of the p160 SRC family comprise a network of immune-regulatory proteins that can function independently or act in synergy with each other, and compensate for - or moderate - the activity of other SRCs. Recent evidence indicates that the SRCs are key participants in governing numerous aspects of CD4+ T cell biology. Here we review findings that establish the SRCs as essential regulators of regulatory T cells (Tregs) and T helper 17 (Th17) cells, with a focus on their crucial roles in Treg immunity in cancer and Treg-Th17 cell phenotypic plasticity.


Subject(s)
T-Lymphocytes, Regulatory , Th17 Cells , Humans , Neoplasms/immunology , Neoplasms/metabolism , Nuclear Receptor Coactivators/metabolism , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Th17 Cells/metabolism
19.
PLoS One ; 19(4): e0289902, 2024.
Article in English | MEDLINE | ID: mdl-38683834

ABSTRACT

Mantle cell lymphoma (MCL) has a poor prognosis and high relapse rates despite current therapies, necessitating novel treatment regimens. Inhibition of SRC-3 show effectiveness in vivo and in vitro in other B cell lymphomas. Additionally, previous studies have shown that SRC-3 is highly expressed in the lymph nodes of B cell non-Hodgkin's lymphoma patients, suggesting SRC-3 may play a role in the progression of B cell lymphoma. This study aimed to investigate novel SRC-3 inhibitors, SI-10 and SI-12, in mantle cell lymphoma. The cytotoxic effects of SI-10 and SI-12 were evaluated in vitro and demonstrated dose-dependent cytotoxicity in a panel of MCL cell lines. The in vivo efficacy of SI-10 was confirmed in two ibrutinib-resistant models: an immunocompetent disseminated A20 mouse model of B-cell lymphoma and a human PDX model of MCL. Notably, SI-10 treatment also resulted in a significant extension of survival in vivo with low toxicity in both ibrutinib-resistant murine models. We have investigated SI-10 as a novel anti-lymphoma compound via the inhibition of SRC-3 activity. These findings indicate that targeting SRC-3 should be investigated in combination with current clinical therapeutics as a novel strategy to expand the therapeutic index and to improve lymphoma outcomes.


Subject(s)
Adenine/analogs & derivatives , Lymphoma, Mantle-Cell , Lymphoma, Mantle-Cell/drug therapy , Lymphoma, Mantle-Cell/pathology , Animals , Humans , Mice , Cell Line, Tumor , Adenine/pharmacology , Adenine/therapeutic use , Piperidines/pharmacology , Piperidines/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Drug Resistance, Neoplasm/drug effects , Xenograft Model Antitumor Assays , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Female
20.
J Med Chem ; 67(7): 5333-5350, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38551814

ABSTRACT

Steroid receptor coactivator 3 (SRC-3) is a critical mediator of many intracellular signaling pathways that are crucial for cancer proliferation and metastasis. In this study, we performed structure-activity relationship exploration and drug-like optimization of the hit compound SI-2, guided by in vitro/in vivo metabolism studies and cytotoxicity assays. Our efforts led to the discovery of two lead compounds, SI-10 and SI-12. Both compounds exhibit potent cytotoxicity against a panel of human cancer cell lines and demonstrate acceptable pharmacokinetic properties. A biotinylated estrogen response element pull-down assay demonstrated that SI-12 could disrupt the recruitment of SRC-3 and p300 in the estrogen receptor complex. Importantly, SI-10 and SI-12 significantly inhibited tumor growth and metastasis in vivo without appreciable acute toxicity. These results demonstrate the potential of SI-10 and SI-12 as drug candidates for cancer therapy, given their potent SRC-3 inhibition and promising pharmacokinetic and toxicity profiles.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Nuclear Receptor Coactivator 3/metabolism , Cell Line , Structure-Activity Relationship , Signal Transduction , Cell Proliferation , Cell Line, Tumor , Antineoplastic Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL