Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 52(13): 7740-7760, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-38932701

ABSTRACT

Androgen receptor- (AR-) indifference is a mechanism of resistance to hormonal therapy in prostate cancer (PC). Here we demonstrate that ONECUT2 (OC2) activates resistance through multiple drivers associated with adenocarcinoma, stem-like and neuroendocrine (NE) variants. Direct OC2 gene targets include the glucocorticoid receptor (GR; NR3C1) and the NE splicing factor SRRM4, which are key drivers of lineage plasticity. Thus, OC2, despite its previously described NEPC driver function, can indirectly activate a portion of the AR cistrome through epigenetic activation of GR. Mechanisms by which OC2 regulates gene expression include promoter binding, enhancement of genome-wide chromatin accessibility, and super-enhancer reprogramming. Pharmacologic inhibition of OC2 suppresses lineage plasticity reprogramming induced by the AR signaling inhibitor enzalutamide. These results demonstrate that OC2 activation promotes a range of drug resistance mechanisms associated with treatment-emergent lineage variation in PC and support enhanced efforts to therapeutically target OC2 as a means of suppressing treatment-resistant disease.


Subject(s)
Adenocarcinoma , Benzamides , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Nitriles , Prostatic Neoplasms , Receptors, Androgen , Receptors, Glucocorticoid , Male , Humans , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma/metabolism , Adenocarcinoma/drug therapy , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , Benzamides/pharmacology , Cell Line, Tumor , Nitriles/pharmacology , Phenylthiohydantoin/pharmacology , Phenylthiohydantoin/analogs & derivatives , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Epigenesis, Genetic , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/pathology , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/drug therapy , Animals , Cell Lineage/genetics , Mice
2.
Cell Oncol (Dordr) ; 2024 May 31.
Article in English | MEDLINE | ID: mdl-38819630

ABSTRACT

PURPOSE: Tumor heterogeneity complicates patient treatment and can be due to transitioning of cancer cells across phenotypic cell states. This process is associated with the acquisition of independence from an oncogenic driver, such as the estrogen receptor (ER) in breast cancer (BC), resulting in tumor progression, therapeutic failure and metastatic spread. The transcription factor ONECUT2 (OC2) has been shown to be a master regulator protein of metastatic castration-resistant prostate cancer (mCRPC) tumors that promotes lineage plasticity to a drug-resistant neuroendocrine (NEPC) phenotype. Here, we investigate the role of OC2 in the dynamic conversion between different molecular subtypes in BC. METHODS: We analyze OC2 expression and clinical significance in BC using public databases and immunohistochemical staining. In vitro, we perform RNA-Seq, RT-qPCR and western-blot after OC2 enforced expression. We also assess cellular effects of OC2 silencing and inhibition with a drug-like small molecule in vitro and in vivo. RESULTS: OC2 is highly expressed in a substantial subset of hormone receptor negative human BC tumors and tamoxifen-resistant models, and is associated with poor clinical outcome, lymph node metastasis and heightened clinical stage. OC2 inhibits ER expression and activity, suppresses a gene expression program associated with luminal differentiation and activates a basal-like state at the gene expression level. We also show that OC2 is required for cell growth and survival in metastatic BC models and that it can be targeted with a small molecule inhibitor providing a novel therapeutic strategy for patients with OC2 active tumors. CONCLUSIONS: The transcription factor OC2 is a driver of BC heterogeneity and a potential drug target in distinct cell states within the breast tumors.

3.
bioRxiv ; 2023 Oct 12.
Article in English | MEDLINE | ID: mdl-37905039

ABSTRACT

Androgen receptor- (AR-) indifference is a mechanism of resistance to hormonal therapy in prostate cancer (PC). Here we demonstrate that the HOX/CUT transcription factor ONECUT2 (OC2) activates resistance through multiple drivers associated with adenocarcinoma, stem-like and neuroendocrine (NE) variants. Direct OC2 targets include the glucocorticoid receptor and the NE splicing factor SRRM4, among others. OC2 regulates gene expression by promoter binding, enhancement of chromatin accessibility, and formation of novel super-enhancers. OC2 also activates glucuronidation genes that irreversibly disable androgen, thereby evoking phenotypic heterogeneity indirectly by hormone depletion. Pharmacologic inhibition of OC2 suppresses lineage plasticity reprogramming induced by the AR signaling inhibitor enzalutamide. These results demonstrate that OC2 activation promotes a range of drug resistance mechanisms associated with treatment-emergent lineage variation in PC. Our findings support enhanced efforts to therapeutically target this protein as a means of suppressing treatment-resistant disease.

4.
Int J Mol Sci ; 24(18)2023 Sep 05.
Article in English | MEDLINE | ID: mdl-37761978

ABSTRACT

Neuroendocrine prostate cancer (NEPC) is a highly aggressive subtype of prostate cancer (PC) that commonly emerges through a transdifferentiation process from prostate adenocarcinoma and evades conventional therapies. Extensive molecular research has revealed factors that drive lineage plasticity, uncovering novel therapeutic targets to be explored. A diverse array of targeting agents is currently under evaluation in pre-clinical and clinical studies with promising results in suppressing or reversing the neuroendocrine phenotype and inhibiting tumor growth and metastasis. This new knowledge has the potential to contribute to the development of novel therapeutic approaches that may enhance the clinical management and prognosis of this lethal disease. In the present review, we discuss molecular players involved in the neuroendocrine phenotype, and we explore therapeutic strategies that are currently under investigation for NEPC.


Subject(s)
Carcinoma, Neuroendocrine , Prostatic Neoplasms , Male , Humans , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Phenotype , Carcinoma, Neuroendocrine/pathology , Cell Line, Tumor
5.
Int J Mol Sci ; 25(1)2023 Dec 20.
Article in English | MEDLINE | ID: mdl-38203275

ABSTRACT

Small cell lung cancer (SCLC) stands out as the most aggressive form of lung cancer, characterized by an extremely high proliferation rate and a very poor prognosis, with a 5-year survival rate that falls below 7%. Approximately two-thirds of patients receive their diagnosis when the disease has already reached a metastatic or extensive stage, leaving chemotherapy as the remaining first-line treatment option. Other than the recent advances in immunotherapy, which have shown moderate results, SCLC patients cannot yet benefit from any approved targeted therapy, meaning that this cancer remains treated as a uniform entity, disregarding intra- or inter-tumoral heterogeneity. Continuous efforts and technological improvements have enabled the identification of new potential targets that could be used to implement novel therapeutic strategies. In this review, we provide an overview of the most recent approaches for SCLC treatment, providing an extensive compilation of the targeted therapies that are currently under clinical evaluation and inhibitor molecules with promising results in vitro and in vivo.


Subject(s)
Lung Neoplasms , Small Cell Lung Carcinoma , Humans , Small Cell Lung Carcinoma/therapy , Lung Neoplasms/drug therapy , Immunotherapy , Aggression , Technology
6.
Am J Clin Exp Urol ; 9(4): 337-349, 2021.
Article in English | MEDLINE | ID: mdl-34541032

ABSTRACT

The nuclear matrix protein Scaffold Attachment Factor B1 (SAFB1, SAFB) can act in prostate cancer (PCa) as an androgen receptor (AR) co-repressor that functions through epigenetic silencing of AR targets, such as prostate specific antigen (PSA, KLK3). Genomic profiling of SAFB1-silenced PCa cells indicated that SAFB1 may play a role in modulating intracrine androgen levels through the regulation of UDP-glucuronosyltransferase (UGT) genes, which inactivate steroid hormones. Gene silencing of SAFB1 resulted in increased levels of free dihydrotesterosterone (DHT), and increased resistance to the AR inhibitor enzalutamide. SAFB1 silencing suppressed expression of the UDP-glucuronosyltransferase family 2 member B15 gene (UGT2B15) and the closely related UGT2B17 gene, which encode proteins that irreversibly inactivate testosterone (T) and DHT. Analysis of human data indicated that genomic loss at the SAFB locus, or down-regulation of expression of the SAFB gene, is associated with aggressive PCa. These findings identify SAFB1 as an important regulator of androgen catabolism in PCa and suggest that loss or inactivation of this protein may promote AR activity by retention of active androgen in tumor cells.

9.
Mol Cancer Res ; 18(5): 671-684, 2020 05.
Article in English | MEDLINE | ID: mdl-32019810

ABSTRACT

We recently reported that restoring the CYP27A1-27hydroxycholesterol axis had antitumor properties. Thus, we sought to determine the mechanism by which 27HC exerts its anti-prostate cancer effects. As cholesterol is a major component of membrane microdomains known as lipid rafts, which localize receptors and facilitate cellular signaling, we hypothesized 27HC would impair lipid rafts, using the IL6-JAK-STAT3 axis as a model given its prominent role in prostate cancer. As revealed by single molecule imaging of DU145 prostate cancer cells, 27HC treatment significantly reduced detected cholesterol density on the plasma membranes. Further, 27HC treatment of constitutively active STAT3 DU145 prostate cancer cells reduced STAT3 activation and slowed tumor growth in vitro and in vivo. 27HC also blocked IL6-mediated STAT3 phosphorylation in nonconstitutively active STAT3 cells. Mechanistically, 27HC reduced STAT3 homodimerization, nuclear translocation, and decreased STAT3 DNA occupancy at target gene promoters. Combined treatment with 27HC and STAT3 targeting molecules had additive and synergistic effects on proliferation and migration, respectively. Hallmark IL6-JAK-STAT gene signatures positively correlated with CYP27A1 gene expression in a large set of human metastatic castrate-resistant prostate cancers and in an aggressive prostate cancer subtype. This suggests STAT3 activation may be a resistance mechanism for aggressive prostate cancers that retain CYP27A1 expression. In summary, our study establishes a key mechanism by which 27HC inhibits prostate cancer by disrupting lipid rafts and blocking STAT3 activation. IMPLICATIONS: Collectively, these data show that modulation of intracellular cholesterol by 27HC can inhibit IL6-JAK-STAT signaling and may synergize with STAT3-targeted compounds.


Subject(s)
Cholesterol/metabolism , Hydroxycholesterols/pharmacology , Interleukin-6/antagonists & inhibitors , Janus Kinase 1/antagonists & inhibitors , Membrane Microdomains/pathology , Prostatic Neoplasms/pathology , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Apoptosis , Biomarkers, Tumor/metabolism , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Male , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Mice , Mice, SCID , Prognosis , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
Clin Exp Metastasis ; 36(2): 119-137, 2019 04.
Article in English | MEDLINE | ID: mdl-30673912

ABSTRACT

The Metastasis Research Society (MRS) 17th Biennial conference on metastasis was held on the 1st to the 5th of August 2018 at Princeton University, NJ, USA. The meeting was held around themes addressing notable aspects of the understanding and treatment of metastasis and metastatic disease covering basic, translational, and clinical research. Importantly, the meeting was largely supported by our patient advocate partners including Susan G. Komen for the Cure, Theresa's Research Foundation and METAvivor. There were a total of 85 presentations from invited and selected speakers spread across the main congress and presentations from the preceding Young Investigator Satellite Meeting. Presentations are summarized in this report by session topic.


Subject(s)
Neoplasm Metastasis , Animals , Humans
12.
Nat Med ; 24(12): 1887-1898, 2018 12.
Article in English | MEDLINE | ID: mdl-30478421

ABSTRACT

Treatment of prostate cancer (PC) by androgen suppression promotes the emergence of aggressive variants that are androgen receptor (AR) independent. Here we identify the transcription factor ONECUT2 (OC2) as a master regulator of AR networks in metastatic castration-resistant prostate cancer (mCRPC). OC2 acts as a survival factor in mCRPC models, suppresses the AR transcriptional program by direct regulation of AR target genes and the AR licensing factor FOXA1, and activates genes associated with neural differentiation and progression to lethal disease. OC2 appears active in a substantial subset of human prostate adenocarcinoma and neuroendocrine tumors. Inhibition of OC2 by a newly identified small molecule suppresses metastasis in mice. These findings suggest that OC2 displaces AR-dependent growth and survival mechanisms in many cases where AR remains expressed, but where its activity is bypassed. OC2 is also a potential drug target in the metastatic phase of aggressive PC.


Subject(s)
Adenocarcinoma/drug therapy , Hepatocyte Nuclear Factor 3-alpha/genetics , Homeodomain Proteins/genetics , Prostatic Neoplasms, Castration-Resistant/drug therapy , Receptors, Androgen/genetics , Transcription Factors/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Androgens/genetics , Androgens/metabolism , Animals , Cell Line, Tumor , Disease Progression , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Homeodomain Proteins/antagonists & inhibitors , Humans , Male , Mice , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/pathology , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Signal Transduction , Transcription Factors/antagonists & inhibitors , Xenograft Model Antitumor Assays
13.
Cancer Res ; 78(21): 6086-6097, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30154147

ABSTRACT

Abnormalities in nuclear shape are a well-known feature of cancer, but their contribution to malignant progression remains poorly understood. Here, we show that depletion of the cytoskeletal regulator, Diaphanous-related formin 3 (DIAPH3), or the nuclear membrane-associated proteins, lamin A/C, in prostate and breast cancer cells, induces nuclear shape instability, with a corresponding gain in malignant properties, including secretion of extracellular vesicles that contain genomic material. This transformation is characterized by a reduction and/or mislocalization of the inner nuclear membrane protein, emerin. Consistent with this, depletion of emerin evokes nuclear shape instability and promotes metastasis. By visualizing emerin localization, evidence for nuclear shape instability was observed in cultured tumor cells, in experimental models of prostate cancer, in human prostate cancer tissues, and in circulating tumor cells from patients with metastatic disease. Quantitation of emerin mislocalization discriminated cancer from benign tissue and correlated with disease progression in a prostate cancer cohort. Taken together, these results identify emerin as a mediator of nuclear shape stability in cancer and show that destabilization of emerin can promote metastasis.Significance: This study identifies a novel mechanism integrating the control of nuclear structure with the metastatic phenotype, and our inclusion of two types of human specimens (cancer tissues and circulating tumor cells) demonstrates direct relevance to human cancer.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/21/6086/F1.large.jpg Cancer Res; 78(21); 6086-97. ©2018 AACR.


Subject(s)
Cell Nucleus/metabolism , Gene Expression Regulation, Neoplastic , Membrane Proteins/metabolism , Neoplasm Metastasis , Nuclear Proteins/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Animals , Apoptosis , Biomarkers, Tumor , Cell Line, Tumor , Cell Movement , Disease Progression , Humans , Male , Mice , Mice, SCID , Neoplasm Invasiveness , Neoplastic Cells, Circulating , Nuclear Envelope
14.
PLoS One ; 8(5): e62771, 2013.
Article in English | MEDLINE | ID: mdl-23650528

ABSTRACT

To determine the effect of retinoic acid (RA) in neuroblastoma we treated RA sensitive neuroblastoma cell lines with 9-cis RA or ATRA for 9 days, or for 5 days followed by absence of RA for another 4 days. Both isomers induced apoptosis and reduced cell density as a result of cell differentiation and/or apoptosis. Flow cytometry revealed that 9-cis RA induced apoptosis more effectively than ATRA. The expression profile of apoptosis and survival pathways was cell line specific and depended on the isomer used.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/genetics , Transcriptome/drug effects , Tretinoin/pharmacology , Alitretinoin , Cell Line, Tumor , Cell Shape/drug effects , Cell Survival , Drug Resistance, Neoplasm , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neuroblastoma
15.
Am J Pathol ; 181(5): 1573-84, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23022210

ABSTRACT

Oncosomes are tumor-derived microvesicles that transmit signaling complexes between cell and tissue compartments. Herein, we show that amoeboid tumor cells export large (1- to 10-µm diameter) vesicles, derived from bulky cellular protrusions, that contain metalloproteinases, RNA, caveolin-1, and the GTPase ADP-ribosylation factor 6, and are biologically active toward tumor cells, endothelial cells, and fibroblasts. We describe methods by which large oncosomes can be selectively sorted by flow cytometry and analyzed independently of vesicles <1 µm. Structures resembling large oncosomes were identified in the circulation of different mouse models of prostate cancer, and their abundance correlated with tumor progression. Similar large vesicles were also identified in human tumor tissues, but they were not detected in the benign compartment. They were more abundant in metastases. Our results suggest that tumor microvesicles substantially larger than exosome-sized particles can be visualized and quantified in tissues and in the circulation, and isolated and characterized using clinically adaptable methods. These findings also suggest a mechanism by which migrating tumor cells condition the tumor microenvironment and distant sites, thereby potentiating advanced disease.


Subject(s)
Cell-Derived Microparticles/pathology , Prostatic Neoplasms/blood , Prostatic Neoplasms/pathology , ADP-Ribosylation Factor 6 , Animals , Caveolin 1/metabolism , Cell Line, Tumor , Cell-Derived Microparticles/ultrastructure , Flow Cytometry , Humans , Male , Mice , Models, Biological , Neoplasm Invasiveness , Neoplasm Metastasis , Prostatic Neoplasms/ultrastructure
16.
J. physiol. biochem ; 68(3): 461-473, sept. 2012.
Article in English | IBECS | ID: ibc-122334

ABSTRACT

Increased intracelullar hormone concentration levels have been shown to be the cause of several endocrine-related cancers including breast, prostate, endometrial, ovarian, cervix, testicular, thyroid, and osteosarcoma. Deregulated expression of steroidogenic enzymes in these tumors seems to be the source of a positive balance in active steroids that bind to the corresponding nuclear receptor, thus ultimately stimulating cell proliferation. Among these enzymes, 17 Beta-hydroxysteroid (..) (AU)


Subject(s)
Humans , Endocrine Gland Neoplasms/pathology , 17-Hydroxysteroid Dehydrogenases , Cell Proliferation
17.
J Physiol Biochem ; 68(3): 461-73, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22374586

ABSTRACT

Increased intracelullar hormone concentration levels have been shown to be the cause of several endocrine-related cancers including breast, prostate, endometrial, ovarian, cervix, testicular, thyroid, and osteosarcoma. Deregulated expression of steroidogenic enzymes in these tumors seems to be the source of a positive balance in active steroids that bind to the corresponding nuclear receptor, thus ultimately stimulating cell proliferation. Among these enzymes, 17ß-hydroxysteroid dehydrogenases catalyze the interconversion between 17-ketosteroids and 17-hydroxysteroids on the last steps of sex hormones biosynthesis and metabolism. To date, 14 isoforms have been identified in vertebrates although only 13 are present in humans. Development and clinical evaluation of specific inhibitors to block their activity is currently under progress especially against the best characterized members 1 to 5. Selectivity and potency of these drugs constitute the main challenge in this new approach to cancer and steroid-dependent diseases treatment at the "pre-receptor level". Here we review the current state of knowledge regarding the deregulation of the expression of some of these enzymes in endocrine-related tumors.


Subject(s)
17-Hydroxysteroid Dehydrogenases/genetics , Neoplasms/enzymology , Receptors, Steroid/metabolism , 17-Hydroxysteroid Dehydrogenases/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Cell Proliferation , Female , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Neoplasms/drug therapy , Neoplasms/metabolism
18.
Mol Cell Endocrinol ; 339(1-2): 45-53, 2011 Jun 06.
Article in English | MEDLINE | ID: mdl-21549806

ABSTRACT

Type 11 hydroxysteroid (17-beta) dehydrogenase (HSD17B11) catalyzes the conversion of 5α-androstan-3α,17ß-diol into androsterone suggesting that it may play an important role in androgen metabolism. We previously described that overexpression of C/EBPα or C/EBPß induced HSD17B11 expression in HepG2 cells but this process was not mediated by the CCAAT boxes located within its proximal promoter region. Here, we study HSD17B11 transcriptional regulation in prostate cancer (PC) cells. Transfection experiments showed that the region -107/+18 is sufficient for promoter activity in PC cells. Mutagenesis analysis indicated that Sp1 and C/EBP binding sites found in this region are essential for promoter activity. Additional experiments demonstrated that ectopic expression of Sp1 and C/EBPα upregulated HSD17B11 expression only in PC cell lines. Through DAPA and ChIP assays, specific recruitment of Sp1 and C/EBPα to the HSD17B11 promoter was detected. These results show that HSD17B11 transcription in PC cells is regulated by Sp1 and C/EBPα.


Subject(s)
17-Hydroxysteroid Dehydrogenases/genetics , Aldehyde Oxidoreductases/genetics , Gene Expression Regulation, Neoplastic , 17-Hydroxysteroid Dehydrogenases/metabolism , 5' Untranslated Regions/genetics , Aldehyde Oxidoreductases/metabolism , Base Sequence , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Line, Tumor , Genes, Reporter , Humans , Luciferases, Renilla/biosynthesis , Luciferases, Renilla/genetics , Male , Molecular Sequence Data , Promoter Regions, Genetic , Prostatic Neoplasms , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Transcriptional Activation , Up-Regulation/genetics
19.
J Steroid Biochem Mol Biol ; 122(4): 164-71, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20638476

ABSTRACT

17ß-hydroxysteroid dehydrogenases (HSD17Bs) are enzymes that stereospecifically reduce or oxidize a keto- or hydroxy group at C17 of the steroid scaffold, respectively. Fourteen mammalian HSD17Bs have been identified so far. We previously described that the HSD17B8 gene is regulated by C/EBPß in the hepatocarcinoma cell line HepG2. Here, we analyze the 5'-flanking region of 14 promoters (HSD17B1-14) looking for CCAAT boxes and binding sites for CCAAT enhancer binding factors (C/EBPs). All promoters were found to have binding sites for these transcription factors except HSD17B1. Ectopic expression of C/EBPα or C/EBPß in HepG2 cells showed that HSD17B11 expression was induced by both transcription factors while HSD17B10 expression was only induced by C/EBPß. The first 500bp of the 5'-flanking region of both genes contain two putative binding sites for C/EBPs. Gene reporter assays showed that C/EBPß transactivated HSD17B10 but not HSD17B11. Additional experiments showed that several isoforms of C/EBPß are involved in HSD17B10 regulation. Mutation of the CCAAT box located at -30/-19 induced HSD17B10 promoter activity when only LIP was expressed, while impaired LAP-induced HSD17B10 transactivation in HepG2 cells when LAP isoforms are expressed. Taken together, our findings reveal that HSD17B10 is regulated by several isoforms of C/EBPß in HepG2 cells.


Subject(s)
3-Hydroxyacyl CoA Dehydrogenases/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Gene Expression Regulation , Promoter Regions, Genetic , Base Sequence , CCAAT-Enhancer-Binding Protein-beta/genetics , Hep G2 Cells , Humans , Molecular Sequence Data , Mutation , Protein Binding , Transcriptional Activation
20.
J Endocrinol ; 200(1): 85-92, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18852215

ABSTRACT

Hydroxysteroid (17-beta) dehydrogenase (HSD17B) are the enzymes responsible for the reversible interconversion of 17-hydroxy and 17-keto steroids. The human and mouse type 8 17beta-HSD (HSD17B8) selectively catalyze the conversion of estradiol (E2) to estrone (E1). We previously described thatHSD17B8 is transcriptionally regulated by C/EBPbeta, and that C/EBPbeta is bound to CCAAT boxes located at -5 and -46 of the transcription start site in basal conditions in HepG2 cells. Furthermore, ectopic expression of C/EBPbeta transactivated the HSD17B8 promoter activity. Here, we show that HSD17B8 expression is up-regulated in response to E2 in the estrogen receptor alpha (ERalpha) positive MCF-7 cells. Results showed that this induction is mediated by ERalpha because i) E2 did not induce HSD17B8 expression in ERalphanegative HepG2 cells, ii) ectopic expression of ERalpha restored E2-induced HSD17B8 expression, and iii) this induction was blocked by the anti-ER ICI 182,780. Additional experiments showed that no estrogen response element was necessary for this regulation. However, the CCAAT boxes located at the HSD17B8 proximal promoter were required for E2-induced transcription. Furthermore, co-immunoprecipitation studies revealed tethering of ERalphatoC/EBPbeta in response to E2 in cells expressing ERalpha. Additionally, chromatin immunoprecipitation assays demonstrated that, in response to E2, ERalpha is recruited to the CCAAT boxes in which C/EBPbeta is already bound. Taken together, our results reveal that ERalpha is involved in the transcriptional regulation of HSD17B8 gene in response to E2 through its interaction with C/EBPbeta.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Estradiol/metabolism , Estrogen Receptor alpha/metabolism , Gene Expression , Oxidoreductases/genetics , Proteins/genetics , Receptor Cross-Talk , Signal Transduction , CCAAT-Enhancer-Binding Protein-beta/genetics , Cell Line , Estrogen Receptor alpha/genetics , Humans , Oxidoreductases/metabolism , Protein Binding , Proteins/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...