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.
J Biol Chem ; 294(38): 14135-14148, 2019 09 20.
Article in English | MEDLINE | ID: mdl-31366726

ABSTRACT

The tumor suppressor CYLD is a deubiquitinating enzyme that suppresses polyubiquitin-dependent signaling pathways, including the proinflammatory and cell growth-promoting NF-κB pathway. Missense mutations in the CYLD gene are present in individuals with syndromes such as multiple familial trichoepithelioma (MFT), but the pathogenic roles of these mutations remain unclear. Recent studies have shown that CYLD interacts with a RING finger domain protein, mind bomb homologue 2 (MIB2), in the regulation of NOTCH signaling. However, whether MIB2 is an E3 ubiquitin ligase that acts on CYLD is unknown. Here, using the cell-free-based AlphaScreen and pulldown assays to detect protein-protein interactions, along with immunofluorescence assays and murine Mib2 knockout cells and animals, we demonstrate that MIB2 promotes proteasomal degradation of CYLD and enhances NF-κB signaling. Of note, arthritic inflammation was suppressed in Mib2-deficient mice. We further observed that the ankyrin repeat in MIB2 interacts with the third CAP domain in CYLD and that MIB2 catalyzes Lys-48-linked polyubiquitination of CYLD at Lys-338 and Lys-530. MIB2-dependent CYLD degradation activated NF-κB signaling via tumor necrosis factor alpha (TNFα) stimulation and the linear ubiquitination assembly complex (LUBAC). Mib2-knockout mice had reduced serum interleukin-6 (IL-6) and exhibited suppressed inflammatory responses in the K/BxN serum-transfer arthritis model. Interestingly, MIB2 significantly enhanced the degradation of a CYLDP904L variant identified in an individual with MFT, although the molecular pathogenesis of the disease was not clarified here. Together, these results suggest that MIB2 enhances NF-κB signaling in inflammation by promoting the ubiquitin-dependent degradation of CYLD.


Subject(s)
Deubiquitinating Enzyme CYLD/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cysteine Endopeptidases/metabolism , Deubiquitinating Enzymes/metabolism , Female , HEK293 Cells , HeLa Cells , Humans , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Polyubiquitin/metabolism , Proteasome Endopeptidase Complex/metabolism , Signal Transduction/physiology , Transcription Factor RelA , Tumor Necrosis Factor-alpha/metabolism , Ubiquitin/metabolism , Ubiquitination
2.
Biochem Biophys Res Commun ; 495(3): 2289-2295, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29284118

ABSTRACT

Nuclear factor-κB (NF-κB) proteins are transcription factors that play key roles in regulating most immune responses and cell death. Constitutively active NF-κB has been shown to exhibit chemoresistance by inducing anti-apoptosis in tumor cells. Multiple myeloma is known as a constitutive NF-κB activating disease, and the proteasome inhibitor bortezomib is used to treat multiple myeloma and mantle cell lymphoma. We demonstrate here that DANFIN (N,N'-bis-(2,4-dimethyl-phenyl)-ethane-1,2-diamine) functions as an inhibitor of the p65 family proteins and induces chemosensitization to bortezomib in multiple myeloma. DANFIN was found to be an inhibitor of interactions between p65 and IκBα without the inhibition of the DNA binding activity of the p65 protein. In addition, DANFIN affected the IκBα binding region in Rel Homology Domain (RHD) and suppressed the nuclear translocalization of the p65 protein in cells. Furthermore, in multiple myeloma cells, DANFIN suppressed the expression level of NF-κB target genes and induced apoptosis. The combination therapy of DANFIN with bortezomib dramatically enhanced the apoptosis of multiple myeloma cells and indicated a remarkable anti-tumor effect in a multiple-myeloma xenograft mouse model.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Apoptosis/drug effects , Bortezomib/administration & dosage , Diamines/administration & dosage , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , NF-kappa B/metabolism , Animals , Cell Line, Tumor , Drug Synergism , Female , Mice , Mice, Inbred BALB C , Mice, Nude , Multiple Myeloma/pathology , Transcription Factors/metabolism , Treatment Outcome
3.
PLoS One ; 11(6): e0156718, 2016.
Article in English | MEDLINE | ID: mdl-27249653

ABSTRACT

Ubiquitination is a key post-translational modification in the regulation of numerous biological processes in eukaryotes. The primary roles of ubiquitination are thought to be the triggering of protein degradation and the regulation of signal transduction. During protein ubiquitination, substrate specificity is mainly determined by E3 ubiquitin ligase (E3). Although more than 600 genes in the human genome encode E3, the E3s of many target proteins remain unidentified owing to E3 diversity and the instability of ubiquitinated proteins in cell. We demonstrate herein a novel biochemical analysis for the identification of E3s targeting specific proteins. Using wheat cell-free protein synthesis system, a protein array containing 227 human and 23 mouse recombinant E3s was synthesized. To establish the high-throughput binding assay using AlphaScreen technology, we selected MDM2 and p53 as the model combination of E3 and its target protein. The AlphaScreen assay specifically detected the binding of p53 and MDM2 in a crude translation mixture. Then, a comprehensive binding assay using the E3 protein array was performed. Eleven of the E3s showed high binding activity, including four previously reported E3s (e.g., MDM2, MDM4, and WWP1) targeting p53. This result demonstrated the reliability of the assay. Another interactors, RNF6 and DZIP3-which there have been no report to bind p53-were found to ubiquitinate p53 in vitro. Further analysis showed that RNF6 decreased the amount of p53 in H1299 cells in E3 activity-dependent manner. These results suggest the possibility that the RNF6 ubiquitinates and degrades p53 in cells. The novel in vitro screening system established herein is a powerful tool for finding novel E3s of a target protein.


Subject(s)
Plant Proteins/metabolism , Triticum/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cell-Free System , High-Throughput Screening Assays , Humans , Mice , Models, Theoretical , Protein Binding , Substrate Specificity
4.
PLoS One ; 11(6): e0156716, 2016.
Article in English | MEDLINE | ID: mdl-27271343

ABSTRACT

Polypeptide tag technology is widely used for protein detection and affinity purification. It consists of two fundamental elements: a peptide sequence and a binder which specifically binds to the peptide tag. In many tag systems, antibodies have been used as binder due to their high affinity and specificity. Recently, we obtained clone Ra48, a high-affinity rabbit monoclonal antibody (mAb) against dopamine receptor D1 (DRD1). Here, we report a novel tag system composed of Ra48 antibody and its epitope sequence. Using a deletion assay, we identified EEAAGIARP in the C-terminal region of DRD1 as the minimal epitope of Ra48 mAb, and we named this sequence the "AGIA" tag, based on its central sequence. The tag sequence does not include the four amino acids, Ser, Thr, Tyr, or Lys, which are susceptible to post-translational modification. We demonstrated performance of this new tag system in biochemical and cell biology applications. SPR analysis demonstrated that the affinity of the Ra48 mAb to the AGIA tag was 4.90 × 10-9 M. AGIA tag showed remarkably high sensitivity and specificity in immunoblotting. A number of AGIA-fused proteins overexpressed in animal and plant cells were detected by anti-AGIA antibody in immunoblotting and immunostaining with low background, and were immunoprecipitated efficiently. Furthermore, a single amino acid substitution of the second Glu to Asp (AGIA/E2D) enabled competitive dissociation of AGIA/E2D-tagged protein by adding wild-type AGIA peptide. It enabled one-step purification of AGIA/E2D-tagged recombinant proteins by peptide competition under physiological conditions. The sensitivity and specificity of the AGIA system makes it suitable for use in multiple methods for protein analysis.


Subject(s)
Antibodies, Monoclonal/metabolism , Chromatography, Affinity/methods , Peptide Fragments/immunology , Receptors, Dopamine D1/chemistry , Receptors, Dopamine D1/metabolism , Amino Acid Sequence , Animals , Antibody Affinity , Cell Line, Tumor , Epitopes/immunology , HEK293 Cells , HeLa Cells , Humans , MCF-7 Cells , Mice , NIH 3T3 Cells , Protein Binding , Rabbits , Receptors, Dopamine D1/analysis , Receptors, Dopamine D1/immunology
5.
FEBS Lett ; 588(17): 3030-7, 2014 Aug 25.
Article in English | MEDLINE | ID: mdl-24931367

ABSTRACT

The Cdk-related protein kinase Pctaire1/Cdk16 is abundantly expressed in brain, testis and skeletal muscle. Functional roles of Pctaire1 such as regulation of neuron migration and neurite outgrowth thus far have been mainly elucidated in the field of nervous system development. Although these regulations based on cytoskeletal rearrangements evoke a possible role of Pctaire1 in the development of skeletal muscle, little is known in this regard. In this study, we demonstrated that myogenic differentiation and subsequent fusion is promoted in Pctaire1 overexpressing cells, and conversely, is inhibited in the knockdown cells. Furthermore, our findings suggest that Pctaire1 exerts promyogenic effects by regulating myoblast migration and process formation during skeletal myogenesis.


Subject(s)
Cell Movement , Cyclin-Dependent Kinases/metabolism , Muscle Development , Muscle, Skeletal/cytology , Muscle, Skeletal/growth & development , Myoblasts/cytology , Animals , Cell Differentiation , Cell Fusion , Cell Line , Cyclin-Dependent Kinases/deficiency , Cyclin-Dependent Kinases/genetics , Gene Knockdown Techniques , Humans , Mice , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...