Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
BMB Rep ; 53(2): 94-99, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31234958

ABSTRACT

Brain cytoplasmic 200 RNA (BC200 RNA) is proposed to act as a local translational modulator by inhibiting translation after being targeted to neuronal dendrites. However, the mechanism by which BC200 RNA inhibits translation is not fully understood. Although a detailed functional analysis of RNA motifs is essential for understanding the BC200 RNA-mediated translation-inhibition mechanism, there is little relevant research on the subject. Here, we performed a systematic domain-dissection analysis of BC200 RNA to identify functional RNA motifs responsible for its translationalinhibition activity. Various RNA variants were assayed for their ability to inhibit translation of luciferase mRNA in vitro. We found that the 111-200-nucleotide region consisting of part of the Alu domain as well as the A/C-rich domain (consisting of both the A-rich and C-rich domains) is most effective for translation inhibition. Surprisingly, we also found that individual A-rich, A/C-rich, and Alu domains can enhance translation but at different levels for each domain, and that these enhancing effects manifest as cap-dependent translation. [BMB Reports 2020; 53(2): 94-99].


Subject(s)
Brain/metabolism , Protein Biosynthesis/genetics , RNA, Long Noncoding/chemistry , RNA, Long Noncoding/metabolism , Alu Elements/genetics , Animals , Gene Expression Regulation , Genes, Reporter/genetics , Humans , In Vitro Techniques , Luciferases/genetics , Nucleotide Motifs , RNA, Long Noncoding/genetics , Rabbits , Reticulocytes/metabolism
2.
Sci Rep ; 9(1): 10003, 2019 07 10.
Article in English | MEDLINE | ID: mdl-31292507

ABSTRACT

Elevated expression of transmembrane serine protease 4 (TMPRSS4) correlates with poor prognosis in non-small cell lung cancer, gastric cancer, colorectal cancer, prostate cancer, and other cancer patients. Previously, we demonstrated that TMPRSS4 mediates tumor cell invasion, migration, proliferation, and metastasis. In addition, we reported novel 2-hydroxydiarylamide derivatives, IMD-0354 and KRT1853, as TMPRSS4 serine protease inhibitors. Here, we further evaluated the effects of the representative derivatives on TMPRSS4-mediated cellular function and signaling. IMD-0354 and KRT1853 inhibited cancer cell invasion, migration, and proliferation in TMPRSS4-expressing prostate, colon, and lung cancer cells. Both compounds suppressed TMPRSS4-mediated induction of Sp1/3, AP-1, and NF-κB transcription factors. Furthermore, TMPRSS4 promoted cancer cell survival and drug resistance, and both compounds enhanced anoikis sensitivity as well as reduced bcl-2 and survivin levels. Importantly, KRT1853 efficiently reduced tumor growth in prostate and colon cancer xenograft models. These results strongly recommend KRT1853 for further development as a novel anti-cancer agent.


Subject(s)
Benzamides/administration & dosage , Colorectal Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Prostatic Neoplasms/drug therapy , Serine Proteinase Inhibitors/administration & dosage , Animals , Benzamides/chemistry , Benzamides/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/metabolism , Female , HCT116 Cells , HeLa Cells , Humans , Lung Neoplasms/metabolism , Male , Membrane Proteins/antagonists & inhibitors , Prostatic Neoplasms/metabolism , Serine Endopeptidases , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/pharmacology , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
3.
Mol Cells ; 42(6): 495-500, 2019 Jun 30.
Article in English | MEDLINE | ID: mdl-31250622

ABSTRACT

Brain cytoplasmic 200 RNA (BC200 RNA), originally identified as a neuron-specific non-coding RNA, is also observed in various cancer cells that originate from non-neural cells. Studies have revealed diverse functions of BC200 RNA in cancer cells. Accordingly, we hypothesized that BC200 RNA might be modified in cancer cells to generate cancerous BC200 RNA responsible for its cancer-specific functions. Here, we report that BC200 RNA sequences are highly heterogeneous in cancer cells by virtue of multiple adenine nucleotide insertions in the internal A-rich region. The insertion of adenine nucleotides enhances BC200 RNAmediated translation inhibition, possibly by increasing the binding affinity of BC200 RNA for eIF4A (eukaryotic translation initiation factor 4A).


Subject(s)
Adenine Nucleotides , RNA, Long Noncoding/chemistry , Brain/metabolism , DEAD-box RNA Helicases/antagonists & inhibitors , DEAD-box RNA Helicases/metabolism , Eukaryotic Initiation Factor-4A/antagonists & inhibitors , Eukaryotic Initiation Factor-4A/metabolism , HeLa Cells , Humans , MCF-7 Cells , Mutagenesis, Insertional , Protein Biosynthesis , RNA, Long Noncoding/metabolism
4.
Sci Rep ; 7(1): 6884, 2017 07 31.
Article in English | MEDLINE | ID: mdl-28761139

ABSTRACT

Brain cytoplasmic 200 RNA (BC200 RNA), a neuron-specific non-coding RNA, is also highly expressed in a number of tumors of non-neuronal origin. However, the biosynthesis of BC200 RNA remains poorly understood. In this study, we show that the efficient transcription of BC200 RNA requires both internal and upstream promoter elements in cancer cells. The transcription complex seems to interact with a broad range of sequences within the upstream 100-bp region. The cellular levels and half-lives of BC200 RNA were found to differ across various cancer cell types, but there was no significant correlation between these parameters. Exogenously expressed BC200 RNA had a shorter half-life than that observed for the endogenous version in cancer cells, suggesting that BC200 RNA might be protected by some limiting factor(s) in cancer cells. Transient transfection experiments showed that the transcriptional activity of the exogenous BC200 RNA promoter element varied depending on the cancer cell type. However, the promoter activities together with the half-life data could not explain the differences in the levels of BC200 RNA among different cell types, suggesting that there is another level of transcriptional regulation beyond that detected by our transient transfection experiments.


Subject(s)
Neoplasms/genetics , Promoter Regions, Genetic , RNA, Long Noncoding/chemistry , RNA, Long Noncoding/genetics , Transcription, Genetic , Up-Regulation , Binding Sites , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Half-Life , HeLa Cells , Humans , MCF-7 Cells , Models, Molecular , Nucleic Acid Conformation , Organ Specificity , RNA Stability , RNA, Long Noncoding/metabolism , TATA-Box Binding Protein/metabolism , Transfection
5.
RNA Biol ; 14(10): 1418-1430, 2017 10 03.
Article in English | MEDLINE | ID: mdl-28277927

ABSTRACT

Although BC200 RNA is best known as a neuron-specific non-coding RNA, it is overexpressed in various cancer cells. BC200 RNA was recently shown to contribute to metastasis in several cancer cell lines, but the underlying mechanism was not understood in detail. To examine this mechanism, we knocked down BC200 RNA in cancer cells, which overexpress the RNA, and examined cell motility, profiling of ribosome footprints, and the correlation between cell motility changes and genes exhibiting altered ribosome profiles. We found that BC200 RNA knockdown reduced cell migration and invasion, suggesting that BC200 RNA promotes cell motility. Our ribosome profiling analysis identified 29 genes whose ribosomal occupations were altered more than 2-fold by BC200 RNA knockdown. Many (> 30%) of them were directly or indirectly related to cancer progression. Among them, we focused on S100A11 (which showed a reduced ribosome footprint) because its expression was previously shown to increase cellular motility. S100A11 was decreased at both the mRNA and protein levels following knockdown of BC200 RNA. An actinomycin-chase experiment showed that BC200 RNA knockdown significantly decreased the stability of the S100A11 mRNA without changing its transcription rate, suggesting that the downregulation of S100A11 was mainly caused by destabilization of its mRNA. Finally, we showed that the BC200 RNA-knockdown-induced decrease in cell motility was mainly mediated by S100A11. Together, our results show that BC200 RNA promotes cell motility by stabilizing S100A11 transcripts.


Subject(s)
Neoplasms/genetics , RNA, Long Noncoding/genetics , S100 Proteins/chemistry , S100 Proteins/genetics , Cell Line, Tumor , Cell Movement , Gene Knockdown Techniques , HeLa Cells , Humans , MCF-7 Cells , Neoplasm Invasiveness , RNA Stability , RNA, Messenger/chemistry , S100 Proteins/metabolism
6.
BMB Rep ; 50(6): 318-322, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28042783

ABSTRACT

Brain cytoplasmic 200 RNA (BC200 RNA) is a neuron-specific non-coding RNA, implicated in the inhibition of local synaptodendritic protein synthesis, and is highly expressed in some cancer cells. Although BC200 RNA has been shown to inhibit translation in vitro, the cellular location of this inhibition is unknown. In this study, we used a BC200 RNA-recognizing antibody to identify the cellular locations of BC200 RNA in HeLa cervical carcinoma cells. We observed punctate signals in both the cytoplasm and nucleus, and further discovered that BC200 RNA co-localized with the p-body decapping enzyme, DCP1A, and the heterogeneous nuclear ribonucleoprotein E2 (hnRNP E2). The latter is a known BC200 RNA-binding partner protein and a constituent of p-bodies. This suggests that BC200 RNA is localized to p-bodies via hnRNP E2. [BMB Reports 2017; 50(6): 318-322].


Subject(s)
Brain/metabolism , Cell Nucleus/metabolism , Cytoplasm/metabolism , RNA-Binding Proteins/metabolism , RNA/metabolism , Endoribonucleases/genetics , Endoribonucleases/metabolism , Female , HeLa Cells , Humans , Plasmids/genetics , RNA/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism
7.
FEBS Lett ; 591(2): 393-405, 2017 01.
Article in English | MEDLINE | ID: mdl-28027391

ABSTRACT

The long noncoding RNA BC200 (brain cytoplasmic RNA, 200 nucleotides) acts as a translational modulator of local protein synthesis at dendrites. BC200 RNA has been shown to inhibit translation in vitro, but it remains unknown how this translation inhibition might be controlled in a cell. Here, we performed yeast three-hybrid screening and identified hnRNP E1 and hnRNP E2 as BC200 RNA-interacting proteins. We found that: these hnRNA proteins could restore BC200 RNA-inhibited translation; BC200 RNA interacts with hnRNP E1 and E2 mainly through its unique 3' C-rich domain; and the RNA binding specificities and modes of the two proteins differed somewhat. Our results offer new insights into the regulation of BC200 RNA-mediated translation inhibition.


Subject(s)
Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Protein Biosynthesis , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/metabolism , Base Sequence , DNA-Binding Proteins , Eukaryotic Initiation Factor-4A/metabolism , HeLa Cells , Humans , Nucleic Acid Conformation , Protein Binding
8.
Mol Biol Rep ; 43(10): 1019-25, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27406387

ABSTRACT

The steroid receptor RNA activator (SRA) is a long non-coding RNA (lncRNA) that acts as a putative coactivator for steroid receptor-mediated transcription. A recent study showed that SRA RNA can be structurally dissected into four domains comprising various secondary structures, but the contribution of each domain to the coactivation ability of SRA RNA was previously unknown. Here, we assessed the functional contributions of the various domains of SRA. We examined the effects of each domain on the coactivation of estrogen receptor-α (ERα)-mediated transcription of a luciferase reporter gene in HeLa cells. Then the detailed domain analysis was focused on domain III (D3) not only with the reporter gene in HeLa cells, but also with ERα-responsive genes in MCF7 breast cancer cells. Domain deletion analysis showed that the deletion of any domain decreased the luciferase activity, and that deletion of D3 caused the largest decrease. This D3 deletion effect was not recovered by co-expression of D3 alone; moreover, the expression of D3 fragments (particularly helices H15-H18, which are highly conserved across vertebrates) inhibited luciferase expression in HeLa cells. Moreover, a fragment containing helices H15-H18 reduced ERα-responsive gene expression in MCF7 breast cancer cells. Our findings indicate that D3 inhibited ERα-mediated transcription of a reporter gene in HeLa cells and that helices H15-H18, as a core element responsible for the D3-driven inhibition, reduced expression of ERα-responsive genes in breast cancer cells.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/genetics , Estrogen Receptor alpha/metabolism , Gene Expression Regulation , Transcription, Genetic , HeLa Cells , Humans , MCF-7 Cells , Nucleic Acid Conformation , Sequence Deletion , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...