Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Immunol ; 45(1): 260-72, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25308712

ABSTRACT

T-cell functions must be tightly controlled to keep the balance between vital proinflammatory activity and detrimental overactivation. MicroRNA-146a (miR-146a) has been identified as a key negative regulator of T-cell responses in mice. Its role in human T cells and its relevance to human inflammatory disease, however, remains poorly defined. In this study, we have characterized miR-146a-driven pathways in primary human T cells. Our results identify miR-146a as a critical gatekeeper of Th1-cell differentiation processes acting via molecular mechanisms not uncovered so far. MiR-146a targets protein kinase C epsilon (PRKCε), which is part of a functional complex consisting of PRKCε and signal transducer and activator of transcription 4 (STAT4). Within this complex, PRKCε phosphorylates STAT4, which in turn is capable of promoting Th1-cell differentiation processes in human CD4(+) T lymphocytes. In addition, we observed that T cells of sepsis patients had reduced levels of miR-146a and an increased PRKCε expression in the initial hyperinflammatory phase of the disease. Collectively, our results identify miR-146a as a potent inhibitor of Th1-cell differentiation in human T cells and suggest that dysregulation of miR-146a contributes to the pathogenesis of sepsis.


Subject(s)
MicroRNAs/genetics , Protein Kinase C-epsilon/genetics , STAT4 Transcription Factor/genetics , Sepsis/genetics , Th1 Cells/immunology , Cell Differentiation , Gene Expression Regulation , Humans , MicroRNAs/immunology , Phosphorylation , Primary Cell Culture , Protein Kinase C-epsilon/immunology , STAT4 Transcription Factor/immunology , Sepsis/immunology , Sepsis/pathology , Signal Transduction , Th1 Cells/pathology
2.
Acta Neuropathol ; 125(5): 671-81, 2013 May.
Article in English | MEDLINE | ID: mdl-23340988

ABSTRACT

Favorable outcome after chemotherapy of glioblastomas cannot unequivocally be linked to promoter hypermethylation of the O6-methylguanine-DNA methyltransferase (MGMT) gene encoding a DNA repair enzyme associated with resistance to alkylating agents. This indicates that molecular mechanisms determining MGMT expression have not yet been fully elucidated. We here show that glioblastomas are capable to downregulate MGMT expression independently of promoter methylation by elongation of the 3'-UTR of the mRNA, rendering the alternatively polyadenylated transcript susceptible to miRNA-mediated suppression. While the elongated transcript is poorly expressed in normal brain, its abundance in human glioblastoma specimens is inversely correlated with MGMT mRNA expression. Using a bioinformatically guided experimental approach, we identified miR-181d, miR-767-3p, and miR-648 as significant post-transcriptional regulators of MGMT in glioblastomas; the first two miRNAs induce MGMT mRNA degradation, the latter affects MGMT protein translation. A regression model including the two miRNAs influencing MGMT mRNA expression and the MGMT methylation status reliably predicts The Cancer Genome Atlas MGMT expression data. Responsivity of MGMT expressing T98G glioma cells to temozolomide was significantly enhanced after transfection of miR-181d, miR-767-3p, and miR-648. Taken together, our results uncovered alternative polyadenylation of the MGMT 3'-UTR and miRNA targeting as new mechanisms of MGMT silencing.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/genetics , Glioblastoma/genetics , MicroRNAs/drug effects , O(6)-Methylguanine-DNA Methyltransferase/genetics , Polyadenylation/drug effects , 3' Untranslated Regions , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Culture Techniques , Cell Line, Tumor , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing/drug effects , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , MicroRNAs/metabolism , O(6)-Methylguanine-DNA Methyltransferase/drug effects , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Promoter Regions, Genetic , Temozolomide
3.
PLoS One ; 6(9): e25341, 2011.
Article in English | MEDLINE | ID: mdl-21980429

ABSTRACT

In order to generate genomic signals, the androgen receptor (AR) has to be transported into the nucleus upon androgenic stimuli. However, there is evidence from in vitro experiments that in castration-resistant prostate cancer (CRPC) cells the AR is able to translocate into the nucleus in a ligand-independent manner. The recent finding that inhibition of the glycogen-synthase-kinase 3ß (GSK-3ß) induces a rapid nuclear export of the AR in androgen-stimulated prostate cancer cells prompted us to analyze the effects of a GSK-3ß inhibition in the castration-resistant LNCaP sublines C4-2 and LNCaP-SSR. Both cell lines exhibit high levels of nuclear AR in the absence of androgenic stimuli. Exposure of these cells to the maleimide SB216763, a potent GSK-3ß inhibitor, resulted in a rapid nuclear export of the AR even under androgen-deprived conditions. Moreover, the ability of C4-2 and LNCaP-SSR cells to grow in the absence of androgens was diminished after pharmacological inhibition of GSK-3ß in vitro. The ability of SB216763 to modulate AR signalling and function in CRPC in vivo was additionally demonstrated in a modified chick chorioallantoic membrane xenograft assay after systemic delivery of SB216763. Our data suggest that inhibition of GSK-3ß helps target the AR for export from the nucleus thereby diminishing the effects of mislocated AR in CRPC cells. Therefore, inhibition of GSK-3ß could be an interesting new strategy for the treatment of CRPC.


Subject(s)
Glycogen Synthase Kinase 3/antagonists & inhibitors , Orchiectomy , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Protein Kinase Inhibitors/pharmacology , Receptors, Androgen/metabolism , Active Transport, Cell Nucleus/drug effects , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Gene Silencing , Glycogen Synthase Kinase 3/chemistry , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Indoles/pharmacology , Karyopherins/metabolism , Ligands , Male , Maleimides/pharmacology , Phosphorylation/drug effects , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction/drug effects , Tyrosine/metabolism , Up-Regulation/drug effects , Exportin 1 Protein
4.
J Cell Biochem ; 109(6): 1192-200, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20127713

ABSTRACT

The androgen receptor (AR) is a ligand-dependent transcription factor belonging to the steroid hormone receptor superfamily. Under normal conditions, in the absence of a ligand, the AR is localized to the cytoplasm and is actively transported into the nucleus upon binding of androgens. In advanced prostate cancer (PCa) cell lines, an increased sensitivity to dihydrotestosterone (DHT), enabling the cells to proliferate under sub-physiological levels of androgens, has been associated with increased stability and nuclear localization of the AR. There is experimental evidence that the glycogen synthase kinase-3beta (GSK-3beta), a multifunctional serine/threonine kinase is involved in estrogen and AR stability. As demonstrated in the following study by immunoprecipitation analysis, GSK-3beta binds to the AR forming complexes in the cytoplasm and in the nucleus. Furthermore, inhibition of GSK-3beta activity by pharmacological inhibitors like the maleimide SB216761, the chloromethyl-thienyl-ketone GSK-3 inhibitor VI or the aminopyrazol GSK-3 inhibitor XIII in cells grown in the presence of DHT triggered a rapid nuclear export of endogenous AR as well as of green fluorescent AR-EosFP. The nuclear export of AR following GSK-3beta inhibition could be blocked by leptomycin B suggesting a CRM1-dependent export mechanism. This assumption is supported by the localization of a putative CRM1 binding site at the C-terminus of the AR protein. The results suggest that GSK-3beta is an important element not only in AR stability but also significantly alters nuclear translocation of the AR, thereby modulating the androgenic response of human PCa cells.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Karyopherins/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Active Transport, Cell Nucleus/drug effects , Binding Sites , Blotting, Western , Cell Line, Tumor , Dihydrotestosterone/pharmacology , Enzyme Inhibitors/pharmacology , Fatty Acids, Unsaturated/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Humans , Immunoprecipitation , Karyopherins/antagonists & inhibitors , Male , Microscopy, Fluorescence , Protein Binding/drug effects , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Exportin 1 Protein
5.
Neoplasia ; 10(6): 624-34, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18516299

ABSTRACT

The glycogen synthase kinase 3 (GSK-3) is a serine/threonine kinase widely expressed in mammalian tissues. Initially identified by its ability to modulate glycogen synthesis, GSK-3 turned out to be a multifunctional enzyme, able to phosphorylate many proteins, including members of the steroid receptor superfamily. Although GSK-3 was shown to phosphorylate the androgen receptor (AR), its effects on AR transcriptional activity remain controversial. Analysis of short hairpin RNA (shRNA)-mediated downmodulation of GSK-3 proteins in prostate cancer cells showed a reduction in AR transcriptional activity and AR protein levels. Pharmacological GSK-3 inhibitors such as the maleimide SB216763 or the aminopyrazole GSK inhibitor XIII inhibited AR-dependent reporter gene activity and AR expression in vitro. Analysis of androgen-induced nuclear translocation of the AR was performed in PC3 cells transfected with pAR-t1EosFP coding for EosAR, a green fluorescent AR fusion protein. When grown in presence of androgens, EosAR was predominantly nuclear. Incubation with SB216763 before and after androgen treatment almost completely reduced nuclear EosAR. In contrast, the thiazole-containing urea compound AR-A014418 increased rather than decreased AR-expression/function. Although not all GSK inhibitors affected AR-stability/function, our observations suggest a potential new therapeutic application for some of these compounds in prostate cancer.


Subject(s)
Androgens/metabolism , Glycogen Synthase Kinase 3/metabolism , Prostatic Neoplasms/metabolism , Active Transport, Cell Nucleus , Antineoplastic Agents/pharmacology , Cell Proliferation , Enzyme Inhibitors/pharmacology , Green Fluorescent Proteins/metabolism , Humans , Indoles/pharmacology , Male , Maleimides/pharmacology , RNA, Small Interfering/metabolism , Receptors, Androgen/metabolism , Signal Transduction , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...