Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Nat Commun ; 14(1): 6117, 2023 09 30.
Article in English | MEDLINE | ID: mdl-37777542

ABSTRACT

The proline synthesis is importantly involved in tumor growth under hypoxia, while the underlying mechanism remains to be further investigated. Here we show that pyrroline-5-carpoxylate reductase-1 (PYCR1), displaying a constant nuclear localization, is phosphorylated by nuclear IGF1R at Tyrosine 135 under hypoxia; this phosphorylation promotes the binding of PYCR1 to ELK4 and thus PYCR1 recruitment to ELK4-targeted genes promoter. Under hypoxia, ELK4-binding ability and enzymatic activity of PYCR1 are both required for ELK4-Sirt7-mediated transcriptional repression and cell growth maintenance, in which PYCR1-catalyzed NAD+ production stimulates the deacetylation activity of Sirt7 on H3K18ac that restrains genes transcription. Functionally, PYCR1 Tyr-135 phosphorylation exerts supportive effect on tumor growth under hypoxia, and the level of PYCR1 Tyr-135 phosphorylation is associated with malignancy of colorectal cancer (CRC). These data uncover the relationship between the compartmentally metabolic activity of PYCR1 and genes transcription regulation, and highlight the oncogenic role of PYCR1 during CRC development.


Subject(s)
Neoplasms , Oxidoreductases , Humans , Cell Line, Tumor , ets-Domain Protein Elk-4/metabolism , Hypoxia/genetics , Neoplasms/genetics , Receptor, IGF Type 1/genetics , delta-1-Pyrroline-5-Carboxylate Reductase
2.
Balkan Med J ; 40(6): 409-414, 2023 10 20.
Article in English | MEDLINE | ID: mdl-37519006

ABSTRACT

Background: Cervical cancer (CC) is a prevalent gynecological carcinoma, and patients infected with human papillomavirus (HPV) have a higher morbidity rate. Aims: To explore the effects of ETS-like transcription factor 4 (ELK4) in patients with HPV+ CC. Study design: In vitro cell lines and human-sample study. Methods: The ELK4 levels in human tissue (65 HPV+ CC tissue and 25 HPV− normal cervical tissue) and cell lines (human cervical epithelial immortalized cell line H8 and CC cell lines HeLa [HPV18], CaSki [HPV16], and SiHa [HPV−]) were quantified using qRT-PCR and western blot assay. ELK4 knockdown transfection was effective and confirmed by western blotting. The MTT and EDU assays were used to evaluate cell viability and proliferation, respectively. Flow cytometry was used to detect the CC cell cycle stage. Stem cell markers, such as cluster of differentiation 133 (CD133), CD44, and aldehyde dehydrogenase 1, and the cervicospheres formed were measured. ChIP-qPCR and luciferase activity experiments were used to assess the bond between ELK4 and F-box protein 22 (FBXO22). Results: ELK4 was highly expressed in the HPV+ CC tissue. CC cells with ELK4 knockdown had lower viability and proliferation than the control cells. ELK4 knockdown blocked the progression of the cell cycle from G1 to S phase. ELK4 knockdown suppressed the stem cell-like characteristics of the HPV+ CC cells. ELK4 bonded with the FBXO22 promoter, inhibiting the levels of phosphatase and tensin homolog (PTEN). Conclusion: ELK4 facilitated cell cycle progression and stem cell-like characteristics by regulating the FBXO22/PTEN axis. Thus, ELK4 could be a potential therapeutic target to arrest the progress of HPV-associated CC.


Subject(s)
F-Box Proteins , Papillomavirus Infections , Uterine Cervical Neoplasms , Female , Humans , Uterine Cervical Neoplasms/metabolism , Human Papillomavirus Viruses , Cell Line, Tumor , Cell Proliferation , Stem Cells/metabolism , Stem Cells/pathology , Cell Cycle , PTEN Phosphohydrolase/metabolism , PTEN Phosphohydrolase/pharmacology , ets-Domain Protein Elk-4/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/pharmacology , F-Box Proteins/pharmacology
3.
J Hematol Oncol ; 15(1): 53, 2022 05 07.
Article in English | MEDLINE | ID: mdl-35526007

ABSTRACT

BACKGROUND: Transfer RNA-derived fragments (tRFs) are a new class of small non-coding RNAs. Recent studies suggest that tRFs participate in some pathological processes. However, the biological functions and mechanisms of tRFs in non-small cell lung cancer (NSCLC) are largely unknown. METHODS: Differentially expressed tRFs were identified by tRF and tiRNA sequencing using 9 pairs of pre- and post-operation plasma from patients with NSCLC. Quantitative real-time PCR (qRT-PCR) and fluorescence in situ hybridization (FISH) were used to determine the levels of tRF in tissues, plasma, and cells. Gain- and loss-of-function experiments were implemented to investigate the oncogenic effects of tRF on NSCLC cells in vitro and in vivo. Chromatin immunoprecipitation (ChIP), luciferase reporter, RNA pulldown, mass spectrum, RNA immunoprecipitation (RIP), Western blot, co-immunoprecipitation (Co-IP) assays, and rescue experiments were performed to explore the regulatory mechanisms of tRF in NSCLC. RESULTS: AS-tDR-007333 was an uncharacterized tRF and significantly up-regulated in NSCLC tissues, plasma, and cells. Clinically, AS-tDR-007333 overexpression could distinguish NSCLC patients from healthy controls and associated with poorer prognosis of NSCLC patients. Functionally, overexpression of AS-tDR-007333 enhanced proliferation and migration of NSCLC cells, whereas knockdown of AS-tDR-007333 resulted in opposite effects. Mechanistically, AS-tDR-007333 promoted the malignancy of NSCLC cells by activating MED29 through two distinct mechanisms. First, AS-tDR-007333 bound to and interacted with HSPB1, which activated MED29 expression by enhancing H3K4me1 and H3K27ac in MED29 promoter. Second, AS-tDR-007333 stimulated the expression of transcription factor ELK4, which bound to MED29 promoter and increased its transcription. Therapeutically, inhibition of AS-tDR-007333 suppressed NSCLC cell growth in vivo. CONCLUSIONS: Our study identifies a new oncogenic tRF and uncovers a novel mechanism that AS-tDR-007333 promotes NSCLC malignancy through the HSPB1-MED29 and ELK4-MED29 axes. AS-tDR-007333 is a potential diagnostic or prognostic marker and therapeutic target for NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/genetics , Gene Expression Regulation , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , In Situ Hybridization, Fluorescence , Lung Neoplasms/genetics , Molecular Chaperones , RNA, Transfer/genetics , RNA, Transfer/metabolism , ets-Domain Protein Elk-4/genetics , ets-Domain Protein Elk-4/metabolism
4.
Cell Death Dis ; 12(11): 957, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34663788

ABSTRACT

Long non-coding RNAs (lncRNAs) play important regulatory roles in the initiation and progression of various cancers. However, the biological roles and the potential mechanisms of lncRNAs in gastric cancers remain unclear. Here, we report that the expression of lncRNA SNHG22 (small nucleolar RNA host gene 22) was significantly increased in GC (Gastric Cancer) tissues and cells, which confers poor prognosis of patients. Knockdown of SNHG22 inhibited the proliferation and invasion ability of GC cells. Moreover, we identified that the transcriptional factor, ELK4 (ETS transcription factor ELK4), could promote SNHG22 expression in GC cells. In addition, using RNA pull-down followed MS assay, we found that SNHG22 directly bound to EZH2 (enhancer of zeste 2 polycomb repressive complex 2 subunit) to suppress the expression of tumor suppressor genes. At the same time, SNHG22 sponged miR-200c-3p to increase Notch1 (notch receptor 1) expression. Taken together, our findings demonstrated the role of SNHG22 on promoting proliferation and invasion of GC cells. And we revealed a new regulatory mechanism of SNHG22 in GC cells. SNHG22 is a promising lncRNA biomarker for diagnosis and prognosis and a potential target for GC treatment.


Subject(s)
Disease Progression , Enhancer of Zeste Homolog 2 Protein/metabolism , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Receptor, Notch1/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , ets-Domain Protein Elk-4/metabolism , Animals , Base Sequence , Binding Sites , Cell Line, Tumor , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Genes, Tumor Suppressor , Histones/metabolism , Humans , Lysine/metabolism , Methylation , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Neoplasm Invasiveness , Promoter Regions, Genetic/genetics , Protein Binding , RNA, Long Noncoding/genetics , Up-Regulation/genetics
5.
Inflammation ; 44(5): 2106-2119, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34091806

ABSTRACT

Systemic lupus erythematosus (SLE) is a prototypic autoimmune disease and a common complication of SLE is lupus nephritis (LN) during which lupus autoantibodies and proinflammatory cytokines attack the kidney and cause renal dysfunction. The current treatments to LN are limited due to a poor understanding of the pathogenesis. Here, we studied the molecular mechanisms of LN by investigating the function of circELK4/miR-27b-3p axis. MRL/lpr mice and LPS-treated HK-2 cells were used as the mouse model and cell model of LN, respectively. Blood samples were collected from LN patients. qRT-PCR and western blot were used to measure expression levels of circELK4, miR-27b-3p, apoptosis-related proteins, cytokines, and STING/IRF-3/IFN-I signaling. ELISA was performed to examine levels of cytokines including IL-6 and TNF-α. H&E staining was used to examine kidney morphology. TUNEL staining and flow cytometry were used to determine cell apoptosis. Dual luciferase activity assay and RNA pull down were employed to validate the interactions of circELK4/miR-27b-3p and miR-27b-3p/STING. CircELK4 was elevated in LN mice, patients, and LPS-treated HK-2 cells. Knockdown of circELK4 attenuated renal injury in LN mice and LPS-induced HK-2 cell injury. CircELK4 directly bound to miR-27b-3p while miR-27b-3p targeted STING. Moreover, overexpression of circELK4 could partially reverse the effects of miR-27b-3p mimics on cell apoptosis and inflammation. Furthermore, circELK4/miR-27b-3p regulated renal cell damage via modulating STING/IRF3/IFN-I signaling. CircELK4 contributes to renal injury by promoting inflammation and cell apoptosis via acting as a miR-27b-3p sponge to modulate STING/IRF3/IFN-I signaling in LN.


Subject(s)
Interferon Regulatory Factor-3/metabolism , Interferon Type I/metabolism , Lupus Nephritis/metabolism , Membrane Proteins/metabolism , ets-Domain Protein Elk-4/metabolism , Adult , Animals , Cell Line , Female , HEK293 Cells , Humans , Lupus Nephritis/genetics , Lupus Nephritis/pathology , Male , Mice , Mice, Inbred MRL lpr , MicroRNAs/metabolism , Middle Aged , Signal Transduction/physiology
6.
Oncogene ; 39(24): 4666-4680, 2020 06.
Article in English | MEDLINE | ID: mdl-32398865

ABSTRACT

Aberrant expression of laminin-332 promotes tumour growth and metastasis in multiple cancers. However, the dysregulated expression and mechanism of action of LAMB3, which encodes the ß3 subunit of laminin-332, and the mechanism underlying dysregulated LAMB3 expression in CRC remain obscure. Here, we show that LAMB3 is overexpressed in CRC and that this overexpression is correlated with tumour metastasis and poor prognosis. Overexpression of LAMB3 promoted cell proliferation and cell migration in vitro and tumour growth and metastasis in vivo, while knockdown of LAMB3 elicited opposing effects. LAMB3 inhibited the tumour suppressive function of FOXO3/4 by activating AKT in CRC. Both the BET inhibitor JQ1 and the MEK inhibitor U0126 decreased the mRNA level of LAMB3 in multiple CRC cells. Mechanistically, ELK4 cooperated with BRD2 to regulate the transcription of LAMB3 in CRC by directly binding to the ETS binding motifs in the LAMB3 promoter. ELK4 was as acetylated at K125, which enhanced the interaction between ELK4 and BRD2. JQ1 disrupted the interaction between ELK4 and BRD2, resulting in decreased binding of BRD2 to the LAMB3 promoter and downregulation of LAMB3 transcription. Both ELK4 and BRD2 expression was associated with LAMB3 expression in CRC. LAMB3 expression was also negatively correlated with FOXO3/4 in CRC. Our study reveals the pro-tumorigenic role of LAMB3 through the AKT-FOXO3/4 axis and the transcriptional mechanism of LAMB3 in CRC, demonstrating that LAMB3 is a potential therapeutic target that can be targeted by BET inhibitors and MEK inhibitors.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Cycle Proteins/metabolism , Colorectal Neoplasms/metabolism , Forkhead Box Protein O3/metabolism , Forkhead Transcription Factors/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Transcription Factors/metabolism , ets-Domain Protein Elk-4/metabolism , Acetylation , Animals , Cell Adhesion Molecules/genetics , Cell Cycle Proteins/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Female , Forkhead Box Protein O3/genetics , Forkhead Transcription Factors/genetics , Humans , Male , Mice , Proto-Oncogene Proteins c-akt/genetics , Transcription Factors/genetics , ets-Domain Protein Elk-4/genetics , Kalinin
7.
J Biol Chem ; 292(32): 13296-13311, 2017 08 11.
Article in English | MEDLINE | ID: mdl-28655758

ABSTRACT

Sirtuin 7 (SIRT7), a member of the NAD+-dependent class III histone deacetylases, is involved in the regulation of various cellular processes and in resisting various stresses, such as hypoxia, low glucose levels, and DNA damage. Interestingly, SIRT7 is linked to the control of glycolysis, suggesting a role in glucose metabolism. Given the important roles of SIRT7, it is critical to clarify how SIRT7 activity is potentially regulated. It has been reported that some transcriptional and post-transcriptional regulatory mechanisms are involved. However, little is known how SIRT7 is regulated by the post-translational modifications. Here, we identified ubiquitin-specific peptidase 7 (USP7), a deubiquitinase, as a negative regulator of SIRT7. We showed that USP7 interacts with SIRT7 both in vitro and in vivo, and we further demonstrated that SIRT7 undergoes endogenous Lys-63-linked polyubiquitination, which is removed by USP7. Although the USP7-mediated deubiquitination of SIRT7 had no effect on its stability, the deubiquitination repressed its enzymatic activity. We also showed that USP7 coordinates with SIRT7 to regulate the expression of glucose-6-phosphatase catalytic subunit (G6PC), a gluconeogenic gene. USP7 depletion by RNA interference increased both G6PC expression and SIRT7 enzymatic activity. Moreover, SIRT7 targeted the G6PC promoter through the transcription factor ELK4 but not through forkhead box O1 (FoxO1). In summary, SIRT7 is a USP7 substrate and has a novel role as a regulator of gluconeogenesis. Our study may provide the basis for new clinical approaches to treat metabolic disorders related to glucose metabolism.


Subject(s)
Gene Expression Regulation, Enzymologic , Glucose-6-Phosphatase/metabolism , Promoter Regions, Genetic , Protein Processing, Post-Translational , Sirtuins/metabolism , Ubiquitin Thiolesterase/metabolism , ets-Domain Protein Elk-4/metabolism , Amino Acid Substitution , Cell Line, Tumor , Gene Deletion , Gluconeogenesis , Glucose-6-Phosphatase/antagonists & inhibitors , Glucose-6-Phosphatase/genetics , HEK293 Cells , Humans , Hydrolysis , Lysine/metabolism , Mutation , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Protein Subunits/genetics , Protein Subunits/metabolism , RNA Interference , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sirtuins/antagonists & inhibitors , Sirtuins/genetics , Substrate Specificity , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin Thiolesterase/genetics , Ubiquitin-Specific Peptidase 7 , Ubiquitination , ets-Domain Protein Elk-4/genetics
8.
Biochem J ; 474(9): 1509-1528, 2017 04 19.
Article in English | MEDLINE | ID: mdl-28275114

ABSTRACT

The adapter protein Dok-4 (downstream of kinase-4) has been reported as both an activator and inhibitor of Erk and Elk-1, but lack of knowledge about the identity of its partner molecules has precluded any mechanistic insight into these seemingly conflicting properties. We report that Dok-4 interacts with the transactivation domain of Elk-4 through an atypical phosphotyrosine-binding domain-mediated interaction. Dok-4 possesses a nuclear export signal and can relocalize Elk-4 from nucleus to cytosol, whereas Elk-4 possesses two nuclear localization signals that restrict interaction with Dok-4. The Elk-4 protein, unlike Elk-1, is highly unstable in the presence of Dok-4, through both an interaction-dependent mechanism and a pleckstrin homology domain-dependent but interaction-independent mechanism. This is reversed by proteasome inhibition, depletion of endogenous Dok-4 or lysine-to-arginine mutation of putative Elk-4 ubiquitination sites. Finally, Elk-4 transactivation is potently inhibited by Dok-4 overexpression but enhanced by Dok-4 knockdown in MDCK renal tubular cells, which correlates with increased basal and EGF-induced expression of Egr-1, Fos and cylcinD1 mRNA, and cell proliferation despite reduced Erk activation. Thus, Dok-4 can target Elk-4 activity through multiple mechanisms, including binding of the transactivation domain, nuclear exclusion and protein destabilization, without a requirement for inhibition of Erk.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Nuclear Export Signals/genetics , Nuclear Localization Signals/genetics , ets-Domain Protein Elk-4/genetics , Active Transport, Cell Nucleus/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Animals , COS Cells , Cell Proliferation/genetics , Dogs , Gene Expression Regulation , HEK293 Cells , Humans , Immunoblotting , Madin Darby Canine Kidney Cells , Mice , Microscopy, Confocal , Protein Binding , RNA Interference , Sequence Homology, Amino Acid , Two-Hybrid System Techniques , ets-Domain Protein Elk-4/metabolism
9.
Genomics ; 109(2): 75-82, 2017 03.
Article in English | MEDLINE | ID: mdl-28189763

ABSTRACT

DNA adenine methyltransferase identification (DamID) is an enzymatic technology for detecting DNA regions targeted by chromatin-associated proteins. Proteins are fused to bacterial DNA adenine methyltransferase (Dam) and expressed in cultured cells or whole organisms. Here, we used DamID to detect DNA regions bound by the cardiac-restricted transcription factors (TFs) NKX2-5 and SRF, and ubiquitously-expressed co-factors ELK1 and ELK4. We compared targets bound by these TFs as N- and C-terminal fusions with Dam, for both wild type (WT) NKX2-5 and mutant proteins mimicking those found in congenital heart disease. Overall, DamID is highly robust: while the orientation of WT Dam fusions can affect the size of the target sets, their signatures remained largely reproducible. Furthermore, a severe NKX2-5 mutant lacking the homeodomain showed strong steric effects negatively impacting target discovery. The extent of steric effect is likely to be dependent on the protein in question and the orientation of Dam fusion.


Subject(s)
Chromatin/metabolism , Gene Expression Regulation , Genetic Techniques , Heart Defects, Congenital/metabolism , Site-Specific DNA-Methyltransferase (Adenine-Specific) , Animals , DNA/metabolism , Heart Defects, Congenital/genetics , Homeobox Protein Nkx-2.5/metabolism , Humans , Mice , Serum Response Factor/metabolism , ets-Domain Protein Elk-1/metabolism , ets-Domain Protein Elk-4/metabolism
10.
Pharmazie ; 72(11): 687-693, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-29442044

ABSTRACT

We aimed to elucidate the roles and regulatory mechanism of miR-3188 in oxidized low-density lipoprotein (ox-LDL)-induced cell injury in THP-1 derived macrophages, thus providing a new insight for the treatment of atherosclerosis (AS). A total of 85 AS patients and 45 healthy controls were enrolled. The levels of miR-3188 and lipoprotein-associated phospholipase A2 (Lp-PLA2) in AS patients and healthy controls were detected. Then ox-LDL was used to treat human THP-1 derived macrophages. The effects of overexpression and suppression of miR-3188 on regulating ox-LDL-induced cell injury in THP-1 derived macrophages were investigated. Additionally, the potential target of miR-3188 was identified, which was verified by luciferase reporter assay. Besides, the relationship between miR-3188 and RhoA/ROCK pathway was explored. miR-3188 was downregulated in AS patients, while the levels of Lp-PLA2 in AS patients were increased. Ox-LDL significantly induced cell injury by decreasing cell viability, inducing cell apoptosis and increasing the production of inflammatory cytokines, including IL-1ß, IL-6, MCP-1 and TNF-α. In addition, miR-3188 was significantly downregulated after ox-LDL treatment. Overexpression of miR-3188 alleviated ox-LDL-induced cell injury, while inhibition of miR-3188 had opposite effects. ETS-domain protein 4 (ELK4) was a target of miR-3188. The effects of miR-3188 inhibition on ox-LDL-induced cell injury were markedly reversed by knockdown of ELK4. Besides, inhibition of miR-3188 enhanced ox-LDL-activated RhoA/ROCK pathway, while knockdown of ELK4 suppressed this pathway. Downregulation of miR-3188 may contribute to AS development via negatively regulating Lp-PLA2, targeting ELK4 and activating RhoA/ROCK pathway. miR-3188 may serve as a target for AS treatment.


Subject(s)
Atherosclerosis/pathology , MicroRNAs/physiology , ets-Domain Protein Elk-4/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , 1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , Apoptosis/physiology , Atherosclerosis/genetics , Case-Control Studies , Cell Survival/physiology , Cytokines/metabolism , Down-Regulation , Female , Gene Knockdown Techniques , Humans , Lipoproteins, LDL/metabolism , Macrophages/pathology , Male , MicroRNAs/genetics , Middle Aged , ets-Domain Protein Elk-4/genetics
11.
PLoS One ; 11(3): e0150382, 2016.
Article in English | MEDLINE | ID: mdl-26938874

ABSTRACT

The chimeric RNA, SLC45A3-ELK4, was found to be a product of cis-splicing between the two adjacent genes (cis-SAGe). Despite the biological and clinical significance of SLC45A3-ELK4, its generating mechanism has not been elucidated. It was shown in one cell line that the binding of transcription factor CTCF to the insulators located at or near the gene boundaries, inversely correlates with the level of the chimera. To investigate the mechanism of such cis-SAGe events, we sequenced potential regions that may play a role in such transcriptional read-through. We could not detect mutations at the transcription termination site, insulator sites, splicing sites, or within CTCF itself in LNCaP cells, thus suggesting a "soft-wired" mechanism in regulating the cis-SAGe event. To investigate the role CTCF plays in regulating the chimeric RNA expression, we compared the levels of CTCF binding to the insulators in different cell lines, as well as clinical samples. Surprisingly, we did not find an inverse correlation between CTCF level, or its bindings to the insulators and SLC45A3-ELK4 expression among different samples. However, in three prostate cancer cell lines, different environmental factors can cause the expression levels of the chimeric RNA to change, and these changes do inversely correlate with CTCF level, and/or its bindings to the insulators. We thus conclude that CTCF and its bindings to the insulators are not the primary reasons for differential SLC45A3-ELK4 expression in different cell lines, or clinical cases. However, they are the likely mechanism for the same cells to respond to different environmental cues, in order to regulate the expression of SLC45A3-ELK4 chimeric RNA. This response to different environmental cues is not general to other cis-SAGe events, as we only found one out of 16 newly identified chimeric RNAs showing a pattern similar to SLC45A3-ELK4.


Subject(s)
Gene Expression Regulation, Neoplastic , Membrane Transport Proteins/metabolism , RNA/genetics , Repressor Proteins/metabolism , ets-Domain Protein Elk-4/metabolism , Base Sequence , Binding Sites , CCCTC-Binding Factor , Cell Line, Tumor , Chromatin Immunoprecipitation , HEK293 Cells , Humans , Male , Molecular Sequence Data , Monosaccharide Transport Proteins , Mutation , Point Mutation , Prostatic Neoplasms/metabolism , Repressor Proteins/genetics , Sequence Homology, Nucleic Acid
12.
OMICS ; 18(10): 615-24, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25188740

ABSTRACT

Chromosomal rearrangements and fusion genes play important roles in tumor development and progression. Four high-frequency prostate cancer-specific fusion genes were recently reported in Chinese cases. We attempted to confirm one of the fusion genes, USP9Y-TTTY15, by reverse transcription PCR, but detected the presence of the USP9Y-TTTY15 fusion transcript in cancer samples, nonmalignant prostate tissues, and normal tissues from other organs, demonstrating that it is a transcription-induced chimeric RNA, which is commonly produced in normal tissues. In 105 prostate cancer samples and case-matched adjacent nonmalignant tissues, we determined the expression level of USP9Y-TTTY15 and a previously reported transcription-induced chimeric RNA, SLC45A3-ELK4. The expression levels of both chimeric RNAs vary greatly in cancer and normal cells. USP9Y-TTTY15 expression is neither higher in cancer than adjacent normal tissues, nor correlated with features of advanced prostate cancer. Although the expression level of SLC45A3-ELK4 is higher in cancer than normal cells, and a dramatic increase in its expression from normal to cancer cells is correlated with advanced disease, its expression level in cancer samples alone is not correlated with any clinical parameters. These data show that both chimeric RNAs contribute less to prostate carcinogenesis than previously reported.


Subject(s)
Oncogene Proteins, Fusion/genetics , Prostatic Neoplasms/metabolism , RNA, Untranslated/genetics , Aged , Humans , Male , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Minor Histocompatibility Antigens , Monosaccharide Transport Proteins , Neoplasm Staging , Oncogene Proteins, Fusion/metabolism , Prostate/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA, Untranslated/metabolism , Transcription, Genetic , Transcriptome , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , ets-Domain Protein Elk-4/genetics , ets-Domain Protein Elk-4/metabolism
13.
BMC Genomics ; 15: 301, 2014 Apr 23.
Article in English | MEDLINE | ID: mdl-24758171

ABSTRACT

BACKGROUND: Serum response factor (SRF) is a widely expressed transcription factor involved in multiple regulatory programs. It is believed that SRF can toggle between disparate programs of gene expression through association with different cofactors. However, the direct evidence as to how these factors function on a genome-wide level is still lacking. RESULTS: In the present study, I explored the functions of SRF and its representative cofactors, megakaryoblastic leukemia 1/2 (MKL1/2) and ETS-domain protein 4 (ELK4), during fungal infection challenge in macrophages. The knockdown study, combined with gene expression array analysis, revealed that MKL1/2 regulated SRF-dependent genes were related to actin cytoskeleton organization, while ELK4 regulated SRF-dependent genes were related to external stimulus responses. Subsequent chromatin immunoprecipitation coupled with massively parallel sequencing (ChIP-seq) suggested that many of these regulations were mediated directly in cis. CONCLUSIONS: I conclude that SRF utilizes MKL1/2 to fulfill steady state cellular functions, including cytoskeletal organization, and utilizes ELK4 to facilitate acute responses to external infection. Together, these findings indicate that SRF, along with its two cofactors, are important players in both cellular homeostasis and stress responses in macrophages.


Subject(s)
Gene Expression Regulation , Macrophages/metabolism , Serum Response Factor/genetics , Trans-Activators/metabolism , Transcription Factors/metabolism , Transcription, Genetic , ets-Domain Protein Elk-4/metabolism , Animals , Binding Sites , Cell Line , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Macrophages/drug effects , Male , Mice , Nucleotide Motifs , Position-Specific Scoring Matrices , Promoter Regions, Genetic , Protein Binding , Protein Transport , Reproducibility of Results , Trans-Activators/genetics , Transcription Factors/genetics , Zymosan/pharmacology , ets-Domain Protein Elk-4/genetics
14.
Am J Hum Genet ; 92(4): 489-503, 2013 Apr 04.
Article in English | MEDLINE | ID: mdl-23540573

ABSTRACT

Analysis of 4,405 variants in 89,050 European subjects from 41 case-control studies identified three independent association signals for estrogen-receptor-positive tumors at 11q13. The strongest signal maps to a transcriptional enhancer element in which the G allele of the best candidate causative variant rs554219 increases risk of breast cancer, reduces both binding of ELK4 transcription factor and luciferase activity in reporter assays, and may be associated with low cyclin D1 protein levels in tumors. Another candidate variant, rs78540526, lies in the same enhancer element. Risk association signal 2, rs75915166, creates a GATA3 binding site within a silencer element. Chromatin conformation studies demonstrate that these enhancer and silencer elements interact with each other and with their likely target gene, CCND1.


Subject(s)
Breast Neoplasms/genetics , Chromosomes, Human, Pair 11/genetics , Cyclin D1/genetics , Enhancer Elements, Genetic/genetics , Polymorphism, Single Nucleotide/genetics , Binding Sites , Case-Control Studies , Cell Line, Tumor , Chromatin/chemistry , Chromatin/genetics , Chromatin Immunoprecipitation , Cyclin D1/metabolism , Electrophoretic Mobility Shift Assay , Female , GATA3 Transcription Factor/antagonists & inhibitors , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic , Humans , Luciferases/metabolism , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Silencer Elements, Transcriptional/genetics , ets-Domain Protein Elk-4/antagonists & inhibitors , ets-Domain Protein Elk-4/genetics , ets-Domain Protein Elk-4/metabolism
15.
Science ; 339(6122): 959-61, 2013 Feb 22.
Article in English | MEDLINE | ID: mdl-23348503

ABSTRACT

Cutaneous melanoma occurs in both familial and sporadic forms. We investigated a melanoma-prone family through linkage analysis and high-throughput sequencing and identified a disease-segregating germline mutation in the promoter of the telomerase reverse transcriptase (TERT) gene, which encodes the catalytic subunit of telomerase. The mutation creates a new binding motif for Ets transcription factors and ternary complex factors (TCFs) near the transcription start and, in reporter gene assays, caused up to twofold increase in transcription. We then screened the TERT promoter in sporadic melanoma and observed recurrent ultraviolet signature somatic mutations in 125 of 168 (74%) of human cell lines derived from metastatic melanomas, 45 of 53 corresponding metastatic tumor tissues (85%), and 25 of 77 (33%) primary melanomas. The majority of those mutations occurred at two positions in the TERT promoter and also generated binding motifs for Ets/TCF transcription factors.


Subject(s)
Gene Expression Regulation, Neoplastic , Germ-Line Mutation , Melanoma/genetics , Promoter Regions, Genetic , Skin Neoplasms/genetics , Telomerase/genetics , Binding Sites , Cell Line, Tumor , Female , High-Throughput Nucleotide Sequencing , Humans , Male , Melanoma/secondary , Pedigree , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins c-ets/metabolism , Sequence Analysis, DNA , Skin Neoplasms/pathology , Telomerase/chemistry , Telomerase/metabolism , Transcription Initiation Site , Transcription, Genetic , ets-Domain Protein Elk-1/metabolism , ets-Domain Protein Elk-4/metabolism
16.
Cell Res ; 23(1): 10-2, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22847743

ABSTRACT

Sirtuins are NAD-dependent deacetylases that are conserved from yeast to mammals. A new report sheds light on the function of SIRT7, the least understood member of the Sirtuin family by identifying its locus-specific H3K18 deacetylase activity, and linking it to maintenance of cellular transformation in malignancies.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Sirtuins/metabolism , Acetylation , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Histones/genetics , Histones/metabolism , Humans , Promoter Regions, Genetic , Ribosomal Proteins/metabolism , ets-Domain Protein Elk-4/metabolism
17.
Nature ; 487(7405): 114-8, 2012 Jul 05.
Article in English | MEDLINE | ID: mdl-22722849

ABSTRACT

Sirtuin proteins regulate diverse cellular pathways that influence genomic stability, metabolism and ageing. SIRT7 is a mammalian sirtuin whose biochemical activity, molecular targets and physiological functions have been unclear. Here we show that SIRT7 is an NAD(+)-dependent H3K18Ac (acetylated lysine 18 of histone H3) deacetylase that stabilizes the transformed state of cancer cells. Genome-wide binding studies reveal that SIRT7 binds to promoters of a specific set of gene targets, where it deacetylates H3K18Ac and promotes transcriptional repression. The spectrum of SIRT7 target genes is defined in part by its interaction with the cancer-associated E26 transformed specific (ETS) transcription factor ELK4, and comprises numerous genes with links to tumour suppression. Notably, selective hypoacetylation of H3K18Ac has been linked to oncogenic transformation, and in patients is associated with aggressive tumour phenotypes and poor prognosis. We find that deacetylation of H3K18Ac by SIRT7 is necessary for maintaining essential features of human cancer cells, including anchorage-independent growth and escape from contact inhibition. Moreover, SIRT7 is necessary for a global hypoacetylation of H3K18Ac associated with cellular transformation by the viral oncoprotein E1A. Finally, SIRT7 depletion markedly reduces the tumorigenicity of human cancer cell xenografts in mice. Together, our work establishes SIRT7 as a highly selective H3K18Ac deacetylase and demonstrates a pivotal role for SIRT7 in chromatin regulation, cellular transformation programs and tumour formation in vivo.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Histone Deacetylases/metabolism , Histones/metabolism , Lysine/metabolism , Sirtuins/metabolism , Acetylation , Adenovirus E1A Proteins/genetics , Adenovirus E1A Proteins/metabolism , Animals , Base Sequence , Binding Sites , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Chromatin/metabolism , Contact Inhibition , Disease Progression , Humans , Mice , Neoplasm Transplantation , Nucleotide Motifs , Phenotype , Promoter Regions, Genetic , Repressor Proteins/metabolism , Sirtuins/deficiency , Sirtuins/genetics , Transcription, Genetic , Transplantation, Heterologous , ets-Domain Protein Elk-4/metabolism
18.
Hum Pathol ; 43(11): 1910-6, 2012 11.
Article in English | MEDLINE | ID: mdl-22569213

ABSTRACT

ETS gene rearrangements are frequently found in prostate cancer. Several studies have assessed the rearrangement status of the most commonly found ETS rearranged gene ERG, and the less frequent genes, ETV-1, ETV-4, ETV-5, and ELK-4 in primary prostate cancer. However, frequency in metastatic disease is not well investigated. Recently, we have assessed the ERG rearrangement status in both primary and corresponding lymph node metastases and observed that ERG rearrangement in primary prostate cancer transfers into lymph node metastases, suggesting it to be a clonal expansion event during prostate cancer progression. As a continuation, we investigated in this study whether this observation is valid for the less frequent ETS rearranged genes. Using dual-color break-apart fluorescent in situ hybridization assays, we evaluated the status of all less frequent ETS gene rearrangements for the first time on tissue microarrays constructed from a large cohort of 86 patients with prostate cancer and composed of primary and corresponding lymph node metastases, as well as in a second cohort composed of 43 distant metastases. ETV-1, ETV-4, ETV-5, and ELK-4 rearrangements were found in 8 (10%) of 81, 5 (6%) of 85, 1 (1%) of 85, and 2 (2%) of 86 of primary prostate cancer, respectively, and in 6 (8%) of 73, 4 (6%) of 72, 1 (1%) of 75, and 1 (1%) of 78 of corresponding lymph node metastases, respectively. ETV-1 and ETV-5 rearrangements were not found in the distant metastases cases, whereas ETV-4 and ELK-4 rearrangements were found in 1 (4%) of 25 and 1 (4%) of 24, respectively. Our findings suggest that rearrangement of the less frequent ETS genes is a clonal event during prostate cancer progression.


Subject(s)
Adenocarcinoma/genetics , Adenovirus E1A Proteins/genetics , DNA-Binding Proteins/genetics , Gene Rearrangement , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Transcription Factors/genetics , ets-Domain Protein Elk-4/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/secondary , Adenovirus E1A Proteins/metabolism , Bone Neoplasms/genetics , Bone Neoplasms/secondary , Brain Neoplasms/genetics , Brain Neoplasms/secondary , Clone Cells , DNA-Binding Proteins/metabolism , Disease Progression , Humans , In Situ Hybridization, Fluorescence , Male , Prognosis , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ets , Transcription Factors/metabolism , ets-Domain Protein Elk-4/metabolism
19.
Neuro Oncol ; 13(11): 1202-12, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21846680

ABSTRACT

Glioma is the most common adult primary brain tumor. Its most malignant form, glioblastoma multiforme (GBM), is almost invariably fatal, due in part to the intrinsic resistance of GBM to radiation- and chemotherapy-induced apoptosis. We analyzed B-cell leukemia-2 (Bcl-2) anti-apoptotic proteins in GBM and found myeloid cell leukemia-1 (Mcl-1) to be the highest expressed in the majority of malignant gliomas. Mcl-1 was functionally important, as neutralization of Mcl-1 induced apoptosis and increased chemotherapy-induced apoptosis. To determine how Mcl-1 was regulated in glioma, we analyzed the promoter and identified a novel functional single nucleotide polymorphism in an uncharacterized E26 transformation-specific (ETS) binding site. We identified the ETS transcription factor ELK4 as a critical regulator of Mcl-1 in glioma, since ELK4 downregulation was shown to reduce Mcl-1 and increase sensitivity to apoptosis. Importantly the presence of the single nucleotide polymorphism, which ablated ELK4 binding in gliomas, was associated with lower Mcl-1 levels and a greater dependence on Bcl-xL. Furthermore, in vivo, ELK4 downregulation reduced tumor formation in glioblastoma xenograft models. The critical role of ELK4 in Mcl-1 expression and protection from apoptosis in glioma defines ELK4 as a novel potential therapeutic target for GBM.


Subject(s)
Apoptosis , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Glioblastoma/prevention & control , Proto-Oncogene Proteins c-bcl-2/metabolism , ets-Domain Protein Elk-4/metabolism , Adult , Animals , Base Sequence , Blotting, Western , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/prevention & control , Cell Line, Tumor , Chromatin Immunoprecipitation , Down-Regulation , Electrophoretic Mobility Shift Assay , Glioblastoma/metabolism , Humans , Luciferases/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Molecular Sequence Data , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Grading , Polymorphism, Single Nucleotide/genetics , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Transfection , bcl-X Protein/genetics , bcl-X Protein/metabolism , ets-Domain Protein Elk-4/antagonists & inhibitors , ets-Domain Protein Elk-4/genetics
20.
Int J Mol Med ; 28(1): 89-93, 2011 07.
Article in English | MEDLINE | ID: mdl-21491078

ABSTRACT

The ETS family of transcription factors plays important roles in both normal and neoplastic cells for different biological processes such as proliferation, differentiation, development, transformation, apoptosis, migration, invasion and angiogenesis. The 27 ETS factors are probably a part of complex regulatory networks including interactions among family members. In human prostate cancer, rearrangements have been found in several genes of the ETS family resulting in chimeric oncoproteins. In a previous study we found that the ETS family prototype, Ets-1 affects biological properties of PC3 prostate cancer cells. In a first effort to understand the cooperative interactions between different ETS factors in prostate cancer, in the present study we examined the expression pattern of all 27 ETS members using quantitative RT-PCR (qRT-PCR) in the androgen-sensitive VCaP and LNCaP, and the androgen-insensitive PC3 and DU-145 prostate cancer cell lines as well as in human prostate cancer tissue samples. We further investigated whether the ETS family prototype, Ets-1, regulates other ETS family members by examining the effect of Ets-1 blockade in PC3 cells on their expression. We found an expression specificity of various ETS family members in the prostate cancer cell lines which might reflect their different biological properties. In human prostate samples only 3 among the 27 ETS family members (Ehf, Elk-4 and Ets-2) showed significant expression differences between normal and cancerous prostate glands. We finally demonstrate that the family prototype, Ets-1, regulates the family members Elf-1, Elf-2, Elk-1, Etv-5 and Spi-1 in PC3 prostate cancer cells. Chimeric oncoproteins containing ETS family members arising due to frequent translocations in prostate cancer are probably part of a regulatory network involving other ETS family members as well.


Subject(s)
Prostatic Neoplasms/metabolism , Proto-Oncogene Protein c-ets-1/metabolism , Proto-Oncogene Proteins c-ets/biosynthesis , Aged , Androgens/metabolism , Cell Line, Tumor , Gene Expression Profiling , Gene Regulatory Networks , Humans , Male , Middle Aged , Oncogene Proteins, Fusion/genetics , Prostatic Neoplasms/genetics , Proto-Oncogene Protein c-ets-1/genetics , Proto-Oncogene Protein c-ets-2/genetics , Proto-Oncogene Protein c-ets-2/metabolism , Proto-Oncogene Proteins c-ets/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Translocation, Genetic , ets-Domain Protein Elk-4/genetics , ets-Domain Protein Elk-4/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...