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










Publication year range
1.
Genes (Basel) ; 15(5)2024 04 27.
Article in English | MEDLINE | ID: mdl-38790192

ABSTRACT

TR2 and TR4 (NR2C1 and NR2C2, respectively) are evolutionarily conserved nuclear orphan receptors capable of binding direct repeat sequences in a stage-specific manner. Like other nuclear receptors, TR2 and TR4 possess important roles in transcriptional activation or repression with developmental stage and tissue specificity. TR2 and TR4 bind DNA and possess the ability to complex with available cofactors mediating developmental stage-specific actions in primitive and definitive erythrocytes. In erythropoiesis, TR2 and TR4 are required for erythroid development, maturation, and key erythroid transcription factor regulation. TR2 and TR4 recruit and interact with transcriptional corepressors or coactivators to elicit developmental stage-specific gene regulation during hematopoiesis.


Subject(s)
Hematopoiesis , Humans , Animals , Hematopoiesis/genetics , Nuclear Receptor Subfamily 2, Group C, Member 2/metabolism , Nuclear Receptor Subfamily 2, Group C, Member 2/genetics , Erythropoiesis/genetics , Gene Expression Regulation, Developmental
2.
Sci Adv ; 5(10): eaax6366, 2019 10.
Article in English | MEDLINE | ID: mdl-31633027

ABSTRACT

Alternative lengthening of telomeres (ALT) is known to use homologous recombination (HR) to replicate telomeric DNA in a telomerase-independent manner. However, the detailed process remains largely undefined. It was reported that nuclear receptors COUP-TFII and TR4 are recruited to the enriched GGGTCA variant repeats embedded within ALT telomeres, implicating nuclear receptors in regulating ALT activity. Here, we identified a function of nuclear receptors in ALT telomere maintenance that involves a direct interaction between COUP-TFII/TR4 and FANCD2, the key protein in the Fanconi anemia (FA) DNA repair pathway. The COUP-TFII/TR4-FANCD2 complex actively induces the DNA damage response by recruiting endonuclease MUS81 and promoting the loading of the PCNA-POLD3 replication complex in ALT telomeres. Furthermore, the COUP-TFII/TR4-mediated ALT telomere pathway does not require the FA core complex or the monoubiquitylation of FANCD2, key steps in the canonical FA pathway. Thus, our findings reveal that COUP-TFII/TR4 regulates ALT telomere maintenance through a novel noncanonical FANCD2 pathway.


Subject(s)
COUP Transcription Factor II/metabolism , Fanconi Anemia Complementation Group D2 Protein/metabolism , Nuclear Receptor Subfamily 2, Group C, Member 2/metabolism , Telomere/metabolism , Amino Acid Motifs , COUP Transcription Factor II/antagonists & inhibitors , COUP Transcription Factor II/genetics , Cell Line, Tumor , DNA Polymerase III/metabolism , DNA Repair , DNA-Binding Proteins/metabolism , Endonucleases/metabolism , Fanconi Anemia/genetics , Fanconi Anemia/pathology , Fanconi Anemia Complementation Group D2 Protein/antagonists & inhibitors , Fanconi Anemia Complementation Group D2 Protein/genetics , G2 Phase , Humans , Mutagenesis, Site-Directed , Nuclear Receptor Subfamily 2, Group C, Member 2/antagonists & inhibitors , Nuclear Receptor Subfamily 2, Group C, Member 2/genetics , Proliferating Cell Nuclear Antigen/chemistry , Proliferating Cell Nuclear Antigen/metabolism , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Telomere Homeostasis
3.
Biochem Biophys Res Commun ; 508(1): 210-216, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30473219

ABSTRACT

The aim of this study was to investigate the effect of miR-4778-3p on the radiosensitivity of cervical cancer cells and to elucidate the underlying mechanism. Tissue samples were collected from eight patients with cervical cancer prior to chemoradiotherapy. MicroRNA chip analyses, RT-PCR, gene transfection, CCK8, wound healing and Transwell assays, colony-forming assay, western blot, and the Dual-Luciferase Reporter Assay System were used to evaluate the role of miR-4778-3p in cervical cancer radiosensitivity and its relationships with target molecules NR2C2 and Med19. Thirty-two differentially expressed miRNA molecules (fold-change > 2; p < 0.05) associated with cervical cancer radioresistance were identified. The expression of miR-4778-3p was significantly lower in recurrent or metastatic patients than in control subjects. In vitro studies using radioresistant HeLa and SiHa cervical cancer cell lines showed that miR-4778-3p upregulation significantly inhibited cell proliferation, invasiveness, and migration after irradiation. There was also a significant increase in apoptosis and a significant decrease in the proportion of cells at the G2/M phase. Further, miR-4778-3p upregulation led to increased expression of apoptosis-related molecules, such as Bax, Caspase-3, Caspase-8, and Caspase-9. Reporter gene assays showed that miR-4778-3p bound specifically to NR2C2 and Med19 and negatively regulated their expression. Thus, miR-4778-3p reduces the vitality, proliferation, and migration of radioresistant cervical cancer cells and may regulate the radiosensitivity of cervical cancer by targeting and regulating NR2C2 and Med19 expression.


Subject(s)
Mediator Complex/metabolism , MicroRNAs/metabolism , Nuclear Receptor Subfamily 2, Group C, Member 2/metabolism , Uterine Cervical Neoplasms/genetics , Apoptosis , Cell Cycle , Cell Proliferation , Female , Humans , Mediator Complex/genetics , MicroRNAs/genetics , Nuclear Receptor Subfamily 2, Group C, Member 2/genetics , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/radiotherapy
4.
Blood ; 130(23): 2537-2547, 2017 12 07.
Article in English | MEDLINE | ID: mdl-29018082

ABSTRACT

The orphan nuclear receptors TR4 (NR2C2) and TR2 (NR2C1) are the DNA-binding subunits of the macromolecular complex, direct repeat erythroid-definitive, which has been shown to repress ε- and γ-globin transcription during adult definitive erythropoiesis. Previous studies implied that TR2 and TR4 act largely in a redundant manner during erythroid differentiation; however, during the course of routine genetic studies, we observed multiple variably penetrant phenotypes in the Tr4 mutants, suggesting that indirect effects of the mutation might be masked by multiple modifying genes. To test this hypothesis, Tr4+/- mutant mice were bred into a congenic C57BL/6 background and their phenotypes were reexamined. Surprisingly, we found that homozygous Tr4 null mutant mice expired early during embryogenesis, around embryonic day 7.0, and well before erythropoiesis commences. We further found that Tr4+/- erythroid cells failed to fully differentiate and exhibited diminished proliferative capacity. Analysis of Tr4+/- mutant erythroid cells revealed that reduced TR4 abundance resulted in decreased expression of genes required for heme biosynthesis and erythroid differentiation (Alad and Alas2), but led to significantly increased expression of the proliferation inhibitory factor, cyclin dependent kinase inhibitor (Cdkn1c) These studies support a vital role for TR4 in promoting erythroid maturation and proliferation, and demonstrate that TR4 and TR2 execute distinct, individual functions during embryogenesis and erythroid differentiation.


Subject(s)
Cell Differentiation/genetics , Erythroid Cells/cytology , Erythroid Cells/metabolism , Nuclear Receptor Subfamily 2, Group C, Member 2/genetics , Anemia/blood , Anemia/genetics , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Proliferation/genetics , Erythropoiesis/genetics , Gene Expression Regulation, Developmental , Genes, Lethal , Heterozygote , Homozygote , Lymphopoiesis/genetics , Mice , Mice, Knockout , Mutation , Myelopoiesis/genetics , Nuclear Receptor Subfamily 2, Group C, Member 2/metabolism
5.
Sci Rep ; 7(1): 12600, 2017 10 03.
Article in English | MEDLINE | ID: mdl-28974699

ABSTRACT

NF-κB signaling is essential for osteoclast differentiation and skeletal homeostasis. We have reported recently that NUMB-like (NUMBL) protein modulates osteoclastogenesis by down regulating NF-κB activation. Herein, we decipher the mechanism underlying this phenomenon. We found that whereas NUMBL mRNA expression decreases upon stimulation of wild type (WT) bone marrow macrophages (BMMs) with RANKL, TAK1 deficiency in these cells leads to increased NUMBL and decreased TRAF6 and NEMO expression. These changes were restored upon WT-TAK1 expression, but not with catalytically inactive TAK1-K63W, suggesting that TAK1 enzymatic activity is required for these events. Forced expression of NUMBL inhibits osteoclast differentiation and function as evident by reduction in all hallmarks of osteoclastogenesis. Conversely, NUMBL-null BMMs, show increased osteoclast differentiation and mRNA expression of osteoclast marker genes. Post-translationally, K48-linked poly-ubiquitination of NUMBL is diminished in TAK1-null BMMs compared to elevated K48-poly-ubiquitination in WT cells, indicating increased stability of NUMBL in TAK1-null conditions. Further, our studies show that NUMBL directly interacts with TRAF6 and NEMO, and induces their K48-poly-ubiquitination mediated proteasomal degradation. Collectively, our data suggest that NUMBL and TAK1 are reciprocally regulated and that NUMBL acts as an endogenous regulator of NF-κB signaling and osteoclastogenesis by targeting the TAK1-TRAF6-NEMO axis.


Subject(s)
Intracellular Signaling Peptides and Proteins/genetics , Nerve Tissue Proteins/genetics , Nuclear Receptor Subfamily 2, Group C, Member 2/genetics , Osteogenesis/genetics , TNF Receptor-Associated Factor 6/genetics , Animals , Cell Differentiation/genetics , Gene Expression Regulation, Developmental/genetics , Mice , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , NF-kappa B/genetics , Osteoclasts/metabolism , Proteasome Endopeptidase Complex/genetics , RNA, Messenger/genetics , Signal Transduction
6.
Biochem Biophys Res Commun ; 490(2): 423-428, 2017 08 19.
Article in English | MEDLINE | ID: mdl-28623141

ABSTRACT

Sirtuin 7 (SIRT7) is an NAD+-dependent deacetylase/deacylase, but only a limited number of SIRT7 substrates have been identified. Recently, we found that Sirt7 knockout mice are resistant to high-fat diet-induced fatty liver, and that SIRT7 positively regulates the protein level of TR4, a nuclear receptor involved in lipid metabolism, by inhibiting the CUL4B/DDB1/DCAF1 E3 ubiquitin ligase complex. However, the mechanism by which SIRT7 inhibits the E3 ubiquitin ligase complex was not identified. Here, we demonstrate that SIRT7 binds directly to DDB1 and deacetylates DDB1 at Lys1121. K1121R-DDB1 (a deacetylation-mimicking mutant) displayed reduced binding with DCAF1. The expression of TR4 protein and TR4 target genes, including Cd36, Cidea, Cidec and Pparg1, was increased in K1121R-DDB1-overexpressing Hepa1-6 cells compared to WT-DDB1-overexpressing cells. Our results indicate that the SIRT7-mediated deacetylation of K1121 attenuates the activity of the CUL4B/DDB1/DCAF1 E3 ubiquitin ligase complex by reducing binding between DDB1 and DCAF1, leading to the increased expression of TR4.


Subject(s)
DNA-Binding Proteins/metabolism , Nuclear Receptor Subfamily 2, Group C, Member 2/metabolism , Sirtuins/metabolism , Acetylation , Animals , Gene Expression Regulation , HEK293 Cells , Humans , Mice , Mice, Knockout , Nuclear Receptor Subfamily 2, Group C, Member 2/genetics , Protein Binding , Protein Interaction Maps , Proteolysis , Sirtuins/genetics
7.
Mol Cell Biochem ; 430(1-2): 139-147, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28210900

ABSTRACT

Renilla luciferase reporter is a widely used internal control in dual luciferase reporter assay system, where its transcription is driven by a constitutively active promoter. However, the authenticity of the Renilla luciferase response in some experimental settings has recently been questioned. Testicular receptor 4 (TR4, also known as NR2C2) belongs to the subfamily 2 of nuclear receptors. TR4 binds to a direct repeat regulatory element in the promoter of a variety of target genes and plays a key role in tumorigenesis, lipoprotein regulation, and central nervous system development. In our experimental system using murine pituitary corticotroph tumor AtT20 cells to investigate TR4 actions on POMC transcription, we found that overexpression of TR4 resulted in reduced Renilla luciferase expression whereas knockdown TR4 increased Renilla luciferase expression. The TR4 inhibitory effect was mediated by the TR4 DNA-binding domain and behaved similarly to the GR and its agonist, Dexamethasone. We further demonstrated that the chimeric intron, commonly present in various Renilla plasmid backbones such as pRL-Null, pRL-SV40, and pRL-TK, was responsible for TR4's inhibitory effect. The results suggest that an intron-free Renilla luciferase reporter may provide a satisfactory internal control for TR4 at certain dose range. Our findings advocate caution on the use of Renilla luciferase as an internal control in TR4-directed studies to avoid misleading data interpretation.


Subject(s)
Dexamethasone/pharmacology , Genes, Reporter , Luciferases, Renilla/biosynthesis , Neoplasm Proteins/metabolism , Nuclear Receptor Subfamily 2, Group C, Member 2/metabolism , Pituitary Neoplasms/metabolism , Animals , Cell Line, Tumor , False Positive Reactions , Luciferases, Renilla/genetics , Mice , Neoplasm Proteins/agonists , Neoplasm Proteins/genetics , Nuclear Receptor Subfamily 2, Group C, Member 2/agonists , Nuclear Receptor Subfamily 2, Group C, Member 2/genetics , Pituitary Neoplasms/genetics , Pituitary Neoplasms/pathology , Pro-Opiomelanocortin/biosynthesis , Pro-Opiomelanocortin/genetics
8.
Oncotarget ; 7(43): 69149-69158, 2016 Oct 25.
Article in English | MEDLINE | ID: mdl-27708250

ABSTRACT

We recently demonstrated that the orphan nuclear receptor testicular receptor 4 (TR4) is a potent regulator of corticotroph tumor growth and hormone secretion. The Ras/Raf/MEK/ERK pathway is commonly overactivated in human tumors and we have demonstrated that corticotroph tumor TR4 is activated by ERK1/2-mediated phosphorylation. We evaluated effects of MEK-162, a selective, non-ATP-competitive allosteric inhibitor of MEK1/2, on murine and human in vitro and in vivo corticotroph tumor proliferation and adrenocorticotrophic hormone (ACTH) secretion. MEK-162 treatment dose-dependently inhibited corticotroph tumor proliferation, induced apoptosis, reduced pro-opiomelanocortin (POMC) mRNA levels and inhibited ACTH secretion in vitro. Similar findings were obtained in human corticotroph tumor primary cultures (n = 5). These actions of MEK-162 were augmented in the presence of TR4 overexpression, suggesting that TR4 levels may serve as a predictive biomarker of MEK-162 corticotroph tumor responsiveness. Additionally, MEK-162 treatment reduced TR4 protein expression and blocked recruitment of TR4 to bind its consensus site on the POMC promoter (-854bp to -637bp), elucidating multiple mechanisms to control TR4 corticotroph tumor actions. In a murine corticotroph tumor in vivo model of Cushing's disease, MEK-162 treatment inhibited tumor growth and reduced tumor-derived circulating plasma ACTH, and corticosterone levels. These results demonstrate the potent actions of MEK-162 to inhibit corticotroph tumor growth and hormone secretion in vitro and in vivo via TR4-dependent and independent mechanisms, and raise the possibility of MEK-162 as a novel therapy for Cushing's disease.


Subject(s)
Benzimidazoles/pharmacology , Cell Proliferation/drug effects , MAP Kinase Signaling System/drug effects , Pituitary ACTH Hypersecretion/drug therapy , ACTH-Secreting Pituitary Adenoma/drug therapy , ACTH-Secreting Pituitary Adenoma/genetics , ACTH-Secreting Pituitary Adenoma/metabolism , Adenoma/drug therapy , Adenoma/genetics , Adenoma/metabolism , Adrenocorticotropic Hormone/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression/drug effects , Humans , Mice , Nuclear Receptor Subfamily 2, Group C, Member 2/genetics , Nuclear Receptor Subfamily 2, Group C, Member 2/metabolism , Pituitary ACTH Hypersecretion/genetics , Pituitary ACTH Hypersecretion/metabolism , Pro-Opiomelanocortin/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Tumor Cells, Cultured
10.
Lung Cancer ; 89(3): 320-8, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26144287

ABSTRACT

OBJECTIVES: Aberrant expression of testicular orphan receptor 4 (TR4) has been shown to regulate biological processes near solid tumors. However, the role of TR4 in non-small cell lung cancer (NSCLC) patient prognosis and the development of NSCLC cancer cells are unclear. METHODS: Immunohistochemical analysis was used to evaluate the correlation between TR4 expression and clinicopathological characteristics in 291 cases of NSCLC specimens. A knockdown and overexpression of TR4 was performed to assess the role of TR4. Transwell and colony formation assays were completed to investigate the metastatic and proliferative abilities. Quantitative real-time PCR, Western blotting and immunofluorescence staining were carried out to analyze the epithelial-to-mesenchymal transition (EMT) phenotype. RESULTS: Immunohistochemical evaluation of clinical samples revealed that most of the lung cancer tissues were strongly positive for TR4, whereas the tissues that stained weakly positive or negative for TR4 expression were shown in the paired normal tissues. Moreover, higher levels of TR4 expression were significantly associated with higher lymph node metastases, TNM stages, tumor thrombus in vena and poor prognosis. We observed that downregulation and up-regulation of TR4 with stable cell transfection significantly influence the proliferation, invasive and metastatic abilities of NSCLC lines. In addition, aberrant TR4 expression could modulate the expression levels of several EMT related markers. CONCLUSIONS: Collectively, our results show TR4 expression in NSCLC samples is significantly associated with poor clinicopathological features, and TR4 plays an important role in the metastatic capacity of NSCLC cells by EMT regulation.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/mortality , Epithelial-Mesenchymal Transition/genetics , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Nuclear Receptor Subfamily 2, Group C, Member 2/genetics , Adult , Aged , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Movement/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Grading , Neoplasm Metastasis , Neoplasm Staging , Phenotype , Prognosis , Survival Analysis
11.
Bioinformatics ; 31(15): 2537-44, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25840044

ABSTRACT

MOTIVATION: Modeling regulatory networks using expression data observed in a differentiation process may help identify context-specific interactions. The outcome of the current algorithms highly depends on the quality and quantity of a single time-course dataset, and the performance may be compromised for datasets with a limited number of samples. RESULTS: In this work, we report a multi-layer graphical model that is capable of leveraging many publicly available time-course datasets, as well as a cell lineage-specific data with small sample size, to model regulatory networks specific to a differentiation process. First, a collection of network inference methods are used to predict the regulatory relationships in individual public datasets. Then, the inferred directional relationships are weighted and integrated together by evaluating against the cell lineage-specific dataset. To test the accuracy of this algorithm, we collected a time-course RNA-Seq dataset during human erythropoiesis to infer regulatory relationships specific to this differentiation process. The resulting erythroid-specific regulatory network reveals novel regulatory relationships activated in erythropoiesis, which were further validated by genome-wide TR4 binding studies using ChIP-seq. These erythropoiesis-specific regulatory relationships were not identifiable by single dataset-based methods or context-independent integrations. Analysis of the predicted targets reveals that they are all closely associated with hematopoietic lineage differentiation.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , Erythroid Cells/metabolism , Erythropoiesis/genetics , Gene Expression Regulation , Gene Regulatory Networks , Stem Cells/metabolism , Algorithms , Cells, Cultured , Chromatin Immunoprecipitation , Erythroid Cells/cytology , Genome, Human , High-Throughput Nucleotide Sequencing/methods , Humans , Nuclear Receptor Subfamily 2, Group C, Member 2/genetics , Nuclear Receptor Subfamily 2, Group C, Member 2/metabolism , Prions/genetics , Prions/metabolism , ROC Curve , Sample Size , Stem Cells/cytology , Transcription Factors/genetics , Transcription Factors/metabolism
12.
J Immunol ; 194(7): 3116-26, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25740946

ABSTRACT

Th17-derived Th1 lymphocytes, termed nonclassic, differ from classic Th1 cells because of the presence of retinoic acid orphan receptor (ROR)C2 and the surface expression of CD161 and CCR6. We demonstrate in this article that nonclassic Th1 cells, like Th17 cells, have a marked RORC2 and IL17A demethylation, whereas classic Th1 cells exhibit a complete methylation of these genes. The analysis of RORC2 DNA methylation in the CD4(+)CD161(+) and CD4(+)CD161(-) naive Th subsets from umbilical cord blood surprisingly revealed comparable hypermethylation levels. PCR analysis at the single-cell level revealed that RORC2 mRNA was expressed by none of the CD4(+)CD161(-) and present only in a minority of CD4(+)CD161(+) naive Th cells. These findings provide two important novel observations on the physiology of human Th17 cells: 1) they confirm at the epigenetic level the origin of nonclassic Th1 cells from Th17 cells, also identifying in the RORC2 and IL17A methylation status a novel tool for their distinction from classic Th1 cells, and 2) they demonstrate that RORC2-expressing cells are only a minority in the subset of CD4(+)CD161(+) naive Th cells, which are known to contain all Th17 cell precursors.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , DNA Methylation , Interleukin-17/genetics , Nuclear Receptor Subfamily 2, Group C, Member 2/genetics , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , CD4 Antigens/metabolism , Cytokines/biosynthesis , Gene Expression Regulation , Humans , Immunophenotyping , Interferon-gamma/genetics , Models, Biological , NK Cell Lectin-Like Receptor Subfamily B/metabolism , Phenotype , Precursor Cells, T-Lymphoid/metabolism , Promoter Regions, Genetic , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism
13.
Cancer Gene Ther ; 21(10): 411-5, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25104727

ABSTRACT

Docetaxel-based therapy is one of the first-line options for castration-resistant prostate cancer (CRPC). However, a large proportion of CRPC patients show different extents of docetaxel resistance. The current study aims to investigate the role of testicular nuclear receptor 4 (TR4) in docetaxel resistance in CRPC. TR4 expression level in prostate biopsy samples from CRPC patients treated with docetaxel was measured by immunohistochemistry (IHC). Alternation of TR4 expression in prostate cancer (PCa) cell line PC3 was applied to find out the influence of TR4 on half-maximal inhibitory concentration (IC50), cell viability and cell apoptosis. Patients who failed to achieve prostate-specific antigen (PSA) response (<50% PSA reduction from baseline) after docetaxel-based chemotherapy had a comparatively higher TR4 expression than those who achieved PSA response (⩾50% PSA reduction from baseline). Knocking down TR4 in PC3 cells led to a lower IC50 dose, poorer cell viability and more cell apoptosis when treated with docetaxel, whereas overexpression of TR4 in PC3 led to a higher IC50 dose, better cell viability and less cell apoptosis. TR4 enhances the chemo-resistance of docetaxel in CRPC. It may serve as a biomarker to determine the prognosis of docetaxel-based therapy and as a potential therapy target to combine with docetaxel to better suppress CRPC.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/genetics , Nuclear Receptor Subfamily 2, Group C, Member 2/genetics , Prostatic Neoplasms, Castration-Resistant/genetics , Taxoids/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Docetaxel , Dose-Response Relationship, Drug , Gene Expression , Genetic Vectors/genetics , Humans , Immunohistochemistry , Inhibitory Concentration 50 , Lentivirus/genetics , Male , Nuclear Receptor Subfamily 2, Group C, Member 2/metabolism , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Transduction, Genetic
14.
Carcinogenesis ; 35(6): 1399-406, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24583925

ABSTRACT

Testicular nuclear receptor 4 (TR4), a member of the nuclear receptor superfamily, plays important roles in metabolism, fertility and aging. The linkage of TR4 functions in cancer progression, however, remains unclear. Using three different mouse models, we found TR4 could prevent or delay prostate cancer (PCa)/prostatic intraepithelial neoplasia development. Knocking down TR4 in human RWPE1 and mouse mPrE normal prostate cells promoted tumorigenesis under carcinogen challenge, suggesting TR4 may play a suppressor role in PCa initiation. Mechanism dissection in both in vitro cell lines and in vivo mice studies found that knocking down TR4 led to increased DNA damage with altered DNA repair system that involved the modulation of ATM expression at the transcriptional level, and addition of ATM partially interrupted the TR4 small interfering RNA-induced tumorigenesis in cell transformation assays. Immunohistochemical staining in human PCa tissue microarrays revealed ATM expression is highly correlated with TR4 expression. Together, these results suggest TR4 may function as a tumor suppressor to prevent or delay prostate tumorigenesis via regulating ATM expression at the transcriptional level.


Subject(s)
Cell Transformation, Neoplastic/genetics , DNA Damage , DNA Repair , Nuclear Receptor Subfamily 2, Group C, Member 2/genetics , Prostatic Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Gene Expression , Humans , Immunohistochemistry , Male , Mice , Mice, Knockout , Nuclear Receptor Subfamily 2, Group C, Member 2/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Prostatic Intraepithelial Neoplasia/genetics , Prostatic Intraepithelial Neoplasia/metabolism , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Transcription, Genetic , Tumor Suppressor Proteins/metabolism
15.
Cell Mol Biol Lett ; 17(2): 309-22, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22396141

ABSTRACT

The testicular receptor 4 (TR4) is a member of the nuclear receptor superfamily that controls various biological activities. A protective role of TR4 against oxidative stress has recently been discovered. We here examined the protective role of TR4 against ionizing radiation (IR) and found that small hairpin RNA mediated TR4 knockdown cells were highly sensitive to IR-induced cell death. IR exposure increased the expression of TR4 in scramble control small hairpin RNA expressing cells but not in TR4 knockdown cells. Examination of IR-responsive molecules found that the expression of Gadd45a, the growth arrest and DNA damage response gene, was dramatically decreased in Tr4 deficient (TR4KO) mice tissues and could not respond to IR stimulation in TR4KO mouse embryonic fibroblast cells. This TR4 regulation of GADD45A was at the transcriptional level. Promoter analysis identified four potential TR4 response elements located in intron 3 and exon 4 of the GADD45A gene. Reporter and chromatin immunoprecipitation (ChIP) assays provided evidence indicating that TR4 regulated the GADD45A expression through TR4 response elements located in intron 3 of the GADD45A gene. Together, we find that TR4 is essential in protecting cells from IR stress. Upon IR challenges, TR4 expression is increased, thereafter inducing GADD45A through transcriptional regulation. As GADD45A is directly involved in the DNA repair pathway, this suggests that TR4 senses genotoxic stress and up-regulates GADD45A expression to protect cells from IR-induced genotoxicity.


Subject(s)
Cell Cycle Proteins/metabolism , Nuclear Proteins/metabolism , Nuclear Receptor Subfamily 2, Group C, Member 2/antagonists & inhibitors , Radiation, Ionizing , Animals , Apoptosis/radiation effects , Cell Cycle Checkpoints/radiation effects , Cell Cycle Proteins/genetics , Cells, Cultured , Chromatin Immunoprecipitation , DNA Repair/radiation effects , Exons , Fibroblasts/metabolism , Introns , Mice , Mice, Knockout , Nuclear Proteins/genetics , Nuclear Receptor Subfamily 2, Group C, Member 2/genetics , Nuclear Receptor Subfamily 2, Group C, Member 2/metabolism , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering/metabolism , Up-Regulation
16.
Proc Natl Acad Sci U S A ; 108(46): 18808-13, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-22042865

ABSTRACT

Sickle cell disease (SCD) is a hematologic disorder caused by a missense mutation in the adult ß-globin gene. Higher fetal hemoglobin (HbF) levels in red blood cells of SCD patients have been shown to improve morbidity and mortality. We previously found that nuclear receptors TR2 and TR4 repress expression of the human embryonic ε-globin and fetal γ-globin genes in definitive erythroid cells. Because forced expression of TR2/TR4 in murine adult erythroid cells paradoxically enhanced fetal γ-globin gene expression in transgenic mice, we wished to determine if forced TR2/TR4 expression in a SCD model mouse would result in elevated HbF synthesis and thereby alleviate the disease phenotype. In a "humanized" sickle cell model mouse, forced TR2/TR4 expression increased HbF abundance from 7.6% of total hemoglobin to 18.6%, accompanied by increased hematocrit from 23% to 34% and reticulocyte reduction from 61% to 18%, indicating a significant reduction in hemolysis. Moreover, forced TR2/TR4 expression reduced hepatosplenomegaly and liver parenchymal necrosis and inflammation in SCD mice, indicating alleviation of usual pathophysiological characteristics. This article shows that genetic manipulation of nonglobin proteins, or transcription factors regulating globin gene expression, can ameliorate the disease phenotype in a SCD model animal. This proof-of-concept study demonstrates that modulating TR2/TR4 activity in SCD patients may be a promising therapeutic approach to induce persistent HbF accumulation and for treatment of the disease.


Subject(s)
Anemia, Sickle Cell/genetics , Fetal Hemoglobin/genetics , Nuclear Receptor Subfamily 2, Group C, Member 1/genetics , Nuclear Receptor Subfamily 2, Group C, Member 2/genetics , Animals , Bone Marrow Cells/cytology , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Phenotype , Spleen/cytology , Transgenes , beta-Thalassemia/genetics , gamma-Globins/metabolism
17.
Reprod Biol Endocrinol ; 9: 138, 2011 Oct 13.
Article in English | MEDLINE | ID: mdl-21995792

ABSTRACT

BACKGROUND: Successful reproductive efforts require the establishment of a situation favorable for reproduction that requires integration of both behavior and internal physiological events. TR4 nuclear receptor is known to be involved in male fertility via controlling spermatogenesis, yet its roles in regulating other biological events related to reproduction have not been completely revealed. METHODS: Male TR4 knockout (TR4 -/-) and wild type mice were used for the sexual behavior and penile dysfunction studies. Mice were sacrificed for histological examination and corresponding genes profiles were analyzed by quantitative RT-PCR. Reporter gene assays were performed. RESULTS: We describe an unexpected finding of priapism in TR4 -/- mice. As a transcriptional factor, we demonstrated that TR4 transcriptionally modulates a key enzyme regulating penis erection and neuronal nitric oxide synthese NOS (nNOS). Thereby, elimination of TR4 results in nNOS reduction in both mRNA and protein levels, consequently may lead to erectile dysfunction. In addition, male TR4 -/- mice display defects in sexual and social behavior, with increased fear or anxiety, as well as reduced mounting, intromission, and ejaculation. Reduction of ER alpha, ER beta, and oxytocin in the hypothalamus may contribute to defects in sexual behavior and stress response. CONCLUSIONS: Together, these results provide in vivo evidence of important TR4 roles in penile physiology, as well as in male sexual behavior. In conjunction with previous finding, TR4 represents a key factor that controls male fertility via regulating behavior and internal physiological events.


Subject(s)
Fertility , Nuclear Receptor Subfamily 2, Group C, Member 2/physiology , Penis/metabolism , Priapism/metabolism , Sexual Behavior, Animal , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Gene Expression Regulation, Enzymologic , Genes, Reporter , Male , Mice , Mice, Knockout , Muscle, Smooth/growth & development , Muscle, Smooth/metabolism , Muscle, Smooth/pathology , Muscle, Smooth/physiopathology , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Nuclear Receptor Subfamily 2, Group C, Member 2/genetics , Penis/growth & development , Penis/pathology , Penis/physiopathology , Priapism/pathology , Priapism/physiopathology , Promoter Regions, Genetic , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Response Elements , Severity of Illness Index , Transcriptional Activation
18.
FEBS Lett ; 585(17): 2763-7, 2011 Sep 02.
Article in English | MEDLINE | ID: mdl-21843524

ABSTRACT

We show that TR4 facilitates lipid accumulation in 3T3-L1 adipocytes via induction of the FATP1 gene. Further study showed that TR4 transactivated FATP1 5' promoter activity via direct binding to the TR4 responsive element located at the FATP1 5' promoter region. Constitutive overexpression of TR4 in 3T3-L1 adipocytes resulted in increased lipid accumulation, accompanied by an increase in fatty acid uptake. However, small interfering RNA knockdown of FATP1 abolished TR4-enhanced fatty acid uptake. Moreover, microRNA-mediated silencing of TR4 in 3T3-L1 adipocytes drastically reduced basal FATP1 5' promoter activity and FATP1 expression with reduced lipid accumulation.


Subject(s)
Adipocytes/metabolism , Fatty Acid Transport Proteins/metabolism , Fatty Acids/metabolism , Nuclear Receptor Subfamily 2, Group C, Member 2/metabolism , 3T3-L1 Cells , Animals , Blotting, Western , CD36 Antigens/genetics , CD36 Antigens/metabolism , Chromatin Immunoprecipitation , Coenzyme A Ligases/genetics , Coenzyme A Ligases/metabolism , Fatty Acid Transport Proteins/genetics , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Mice , MicroRNAs/genetics , Nuclear Receptor Subfamily 2, Group C, Member 2/genetics , Reverse Transcriptase Polymerase Chain Reaction
19.
J Cell Physiol ; 222(2): 347-56, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19859911

ABSTRACT

The thyroid hormone receptor (TR) and human testicular orphan receptor 4 (TR4) belong to the nuclear hormone receptor superfamily. They are ligand-dependent transcription factors. TR and TR4 bind to a similar thyroid response element (TRE), known as a direct repeat with four nucleotide spacing (DR4). This study examined the possible interaction or cross-talking between those two receptors. We hypothesized that protein-protein interaction between TR4 and TR may promote TR-mediated transcriptional activity. Glutathione S-transferase pull-down and immunoprecipitation assays showed direct interaction between TR and TR4. Electrophoretic mobility-shift assay demonstrated that TR and TR4 could co-occupy the same TRE. The interaction between TR4 and TR may enhance regulation of genes targeted by TR, such as furin, fibrinogen, cdk2 and p21 expression. We found that TR4 function is similar with TR as TR4 alone could regulate expression of some TR target genes, and could increase cell migration or inhibit cell proliferation. Importantly, the TR-dependent inhibition of cell proliferation and stimulation of cell migration are more enhanced in the HepG2-TR cells stably over-expressing TR4. Overall, TR4 not only has modulation abilities similar to TR but also can cross-talk with TR and promote the TR signaling pathway.


Subject(s)
Nuclear Receptor Subfamily 2, Group C, Member 2/metabolism , Receptor Cross-Talk , Signal Transduction , Thyroid Hormone Receptors alpha/metabolism , Animals , Cell Line , Cell Movement , Cell Proliferation , Electrophoretic Mobility Shift Assay , Furin/genetics , Haplorhini , Humans , Immunoprecipitation , Ligands , Male , Nuclear Receptor Subfamily 2, Group C, Member 2/genetics , Promoter Regions, Genetic , Protein Binding , Response Elements , Signal Transduction/genetics , Thyroid Hormone Receptors alpha/genetics , Time Factors , Transcriptional Activation , Transfection , Triiodothyronine/metabolism
20.
Biochim Biophys Acta ; 1789(11-12): 734-40, 2009.
Article in English | MEDLINE | ID: mdl-19800043

ABSTRACT

Nuclear receptors can be activated by chemicals, metabolites, hormones or environmental compounds to regulate gene expression. Bioassay-guided screening of mouse tissue extracts found that natural fatty acids of a certain carbon length and level of unsaturation could activate the mouse orphan nuclear receptor, testicular orphan receptor 4 (TR4). Subsequent experiments focused on gamma-linoleic acid, a compound identified during screening of mouse tissues that exerts regulatory activity in TR4 transactivation assays. gamma-linoleic acid positively modulates TR4 activity to promote the expression of downstream genes such as apolipoprotein E (ApoE) and phosphoenolpyruvate carboxykinase, and to activate a reporter carrying direct repeat 1 from the ApoE promoter. It also induced the interaction of TR4 with transcription coregulators such as RIP140 and PCAF. Comparisons of transactivation by TR4 and the metabolism-related peroxisome proliferator-activated nuclear receptors indicate that gamma-linoleic acid regulation is specific to TR4. The data suggest that TR4 might exert its physiological function by sensing certain lipids. Identifying these compounds could be useful for examining the physiological pathways in which TR4 and its target genes are involved.


Subject(s)
Fatty Acids/pharmacology , Nuclear Receptor Subfamily 2, Group C, Member 2/genetics , Tissue Extracts/pharmacology , Transcriptional Activation/drug effects , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Base Sequence , COS Cells , Cell Line , Chlorocebus aethiops , Fatty Acids/chemistry , Gas Chromatography-Mass Spectrometry , Humans , Male , Mice , Molecular Structure , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Receptor Interacting Protein 1 , Nuclear Receptor Subfamily 2, Group C, Member 2/metabolism , Peroxisome Proliferator-Activated Receptors/genetics , Peroxisome Proliferator-Activated Receptors/metabolism , Phosphoenolpyruvate Carboxykinase (ATP)/genetics , Phosphoenolpyruvate Carboxykinase (ATP)/metabolism , Protein Binding/drug effects , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Testis/chemistry , Testis/metabolism , Tissue Extracts/chemistry , Two-Hybrid System Techniques , gamma-Linolenic Acid/pharmacology , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...