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.
Acta Pharmacol Sin ; 40(12): 1568-1577, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31197245

ABSTRACT

c-Maf is a critical oncogenic transcription factor that contributes to myelomagenesis. Our previous studies demonstrated that the deubiquitinase USP5 stabilizes c-Maf and promotes myeloma cell proliferation and survival; therefore, the USP5/c-Maf axis could be a potential target for myeloma therapy. As a concept of principle, the present study established a USP5/c-Maf-based luciferase system that was used to screen an FDA-approved drug library. It was found that mebendazole, a typical anthelmintic drug, preferentially induced apoptosis in c-Maf-expressing myeloma cells. Moreover, oral administration of mebendazole delayed the growth of human myeloma xenografts in nude mice but did not show overt toxicity. Further studies showed that the selective antimyeloma activity of mebendazole was associated with the inhibition of the USP5/c-Maf axis. Mebendazole downregulated USP5 expression and disrupted the interaction between USP5 and c-Maf, thus leading to increased levels of c-Maf ubiquitination and subsequent c-Maf degradation. Mebendazole inhibited c-Maf transcriptional activity, as confirmed by both luciferase assays and expression measurements of c-Maf downstream genes. In summary, this study identified mebendazole as a USP5/c-Maf inhibitor that could be developed as a novel antimyeloma agent.


Subject(s)
Antineoplastic Agents/therapeutic use , Mebendazole/therapeutic use , Multiple Myeloma/drug therapy , Proto-Oncogene Proteins c-maf/metabolism , Ubiquitin-Specific Proteases/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cyanoacrylates/therapeutic use , Drug Repositioning , Drug Synergism , Female , HEK293 Cells , Humans , Mice, Inbred BALB C , Mice, Nude , Multiple Myeloma/metabolism , Proof of Concept Study , Protein Binding/drug effects , Proto-Oncogene Proteins c-maf/chemistry , Pyridines/therapeutic use , Ubiquitin-Specific Proteases/chemistry , Ubiquitination/drug effects , Xenograft Model Antitumor Assays
2.
J Clin Invest ; 128(9): 3779-3793, 2018 08 31.
Article in English | MEDLINE | ID: mdl-30059018

ABSTRACT

SUMOylation is involved in the development of several inflammatory diseases, but the physiological significance of SUMO-modulated c-Maf in autoimmune diabetes is not completely understood. Here, we report that an age-dependent attenuation of c-Maf SUMOylation in CD4+ T cells is positively correlated with the IL-21-mediated diabetogenesis in NOD mice. Using 2 strains of T cell-specific transgenic NOD mice overexpressing wild-type c-Maf (Tg-WTc) or SUMOylation site-mutated c-Maf (Tg-KRc), we demonstrated that Tg-KRc mice developed diabetes more rapidly than Tg-WTc mice in a CD4+ T cell-autonomous manner. Moreover, SUMO-defective c-Maf preferentially transactivated Il21 to promote the development of CD4+ T cells with an extrafollicular helper T cell phenotype and expand the numbers of granzyme B-producing effector/memory CD8+ T cells. Furthermore, SUMO-defective c-Maf selectively inhibited recruitment of Daxx/HDAC2 to the Il21 promoter and enhanced histone acetylation mediated by CREB-binding protein (CBP) and p300. Using pharmacological interference with CBP/p300, we illustrated that CBP30 treatment ameliorated c-Maf-mediated/IL-21-based diabetogenesis. Taken together, our results show that the SUMOylation status of c-Maf has a stronger regulatory effect on IL-21 than the level of c-Maf expression, through an epigenetic mechanism. These findings provide new insights into how SUMOylation modulates the pathogenesis of autoimmune diabetes in a T cell-restricted manner and on the basis of a single transcription factor.


Subject(s)
Diabetes Mellitus, Type 1/etiology , Interleukins/genetics , Proto-Oncogene Proteins c-maf/metabolism , Sumoylation , Amino Acid Substitution , Animals , Benzimidazoles/pharmacology , Binding Sites/genetics , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Epigenesis, Genetic , Interleukins/biosynthesis , Isoxazoles/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic , Models, Biological , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins c-maf/chemistry , Proto-Oncogene Proteins c-maf/genetics , Transcriptional Activation , p300-CBP Transcription Factors/antagonists & inhibitors
3.
Am J Med Genet A ; 164A(5): 1272-6, 2014 May.
Article in English | MEDLINE | ID: mdl-24664492

ABSTRACT

Congenital cataracts are the most important cause of severe visual impairment in infants. Genetic factors contribute to the disease development and 29 genes are known to cause congenital cataracts. Identifying the genetic cause of congenital cataracts can be difficult because of genetic heterogeneity. V-maf avian musculoaponeurotic fibrosarcoma oncogene homolog (MAF) encodes a basic region/leucine zipper transcription factor that plays a key role as a regulator of embryonic lens fiber cell development. MAF mutations have been reported to cause juvenile-onset pulverulent cataract, microcornea, iris coloboma, and other anterior segment dysgenesis. We report on six patients in a family who have congenital cataracts were identified MAF mutation by whole exome sequencing (WES). The heterozygous MAF mutation Q303L detected in the present family occurs in a well conserved glutamine residue at the basic region of the DNA-binding domain. All affected members showed congenital cataracts. Three of the six members showed microcornea and one showed iris coloboma. Congenital cataracts with MAF mutation exhibited phenotypically variable cataracts within the family. Review of the patients with MAF mutations supports the notion that congenital cataracts caused by MAF mutations could be accompanied by microcornea and/or iris coloboma. WES is a useful tool for detecting disease-causing mutations in patients with genetically heterogeneous conditions.


Subject(s)
Asian People/genetics , Cataract/congenital , Cataract/genetics , Mutation, Missense , Proto-Oncogene Proteins c-maf/genetics , Alleles , Amino Acid Sequence , Cataract/diagnosis , Child, Preschool , DNA Mutational Analysis , Exome , Female , Genetic Association Studies , Genotype , High-Throughput Nucleotide Sequencing , Humans , Infant , Japan , Male , Molecular Sequence Data , Pedigree , Phenotype , Proto-Oncogene Proteins c-maf/chemistry , Sequence Alignment
4.
Eur J Immunol ; 40(4): 1174-84, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20127678

ABSTRACT

The function of transcription factors can be critically regulated by SUMOylation. c-Maf, the cellular counterpart of v-maf oncogene, is a potent transactivator of the IL-4 gene in Th2 cells. We found in a yeast two-hybrid screen that c-Maf can interact with Ubc9 and PIAS1, two key enzymes of the SUMOylation pathway. In this study, we report that c-Maf co-localized with these two SUMO (small ubiquitin-like modifier) ligases in the nucleus and that c-Maf can be SUMOylated in vitro and also in primary Th2 cells. We also demonstrated that lysine-33 is the dominant, if not the only, SUMO acceptor site of c-Maf. SUMOylation of c-Maf attenuated its transcriptional activity. Reciprocally, a SUMOylation resistant c-Maf was more potent than WT-c-Maf in driving IL-4 production in c-Maf-deficient Th2 cells. Furthermore, we showed that ablation of the SUMO site did not alter the subcellular localization or the stability of c-Maf protein but instead enhanced its recruitment to the Il4-promoter. We conclude that SUMOylation at lysine-33 is a functionally critical post-translational modification event of c-Maf in Th cells.


Subject(s)
Interleukin-4/biosynthesis , Protein Inhibitors of Activated STAT/physiology , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-maf/physiology , Small Ubiquitin-Related Modifier Proteins/physiology , Th2 Cells/metabolism , Ubiquitin-Conjugating Enzymes/physiology , Amino Acid Sequence , Animals , Cell Line , Cells, Cultured/metabolism , Humans , Interleukin-4/genetics , Kidney , Lysine/chemistry , Mice , Molecular Sequence Data , Protein Inhibitors of Activated STAT/chemistry , Protein Inhibitors of Activated STAT/isolation & purification , Protein Interaction Mapping , Proto-Oncogene Proteins c-maf/chemistry , Recombinant Fusion Proteins/physiology , Sequence Alignment , Sequence Homology, Amino Acid , Small Ubiquitin-Related Modifier Proteins/chemistry , Small Ubiquitin-Related Modifier Proteins/genetics , Small Ubiquitin-Related Modifier Proteins/isolation & purification , Transcription, Genetic , Two-Hybrid System Techniques , Ubiquitin-Conjugating Enzymes/chemistry
5.
J Biol Chem ; 282(46): 33681-33690, 2007 Nov 16.
Article in English | MEDLINE | ID: mdl-17875642

ABSTRACT

Nrf2-small Maf heterodimer activates the transcription of many cytoprotective genes through the antioxidant response element and serves as a key factor in xenobiotic and oxidative stress responses. Our surface plasmon resonance-microarray binding analysis revealed that both Nrf2-MafG heterodimer and MafG homodimer bind to the consensus Maf recognition element with high affinity but bind differentially to the suboptimal binding sequences degenerated from the consensus. We examined the molecular basis distinguishing the binding profile of Nrf2-MafG heterodimer from that of MafG homodimer and found that the Ala-502 residue in the basic region of Nrf2 is a critical determinant of its binding specificity. In Maf proteins, a tyrosine resides in the position corresponding to Ala-502 in Nrf2. We prepared a mutant Nrf2 molecule in which Ala-502 was replaced with tyrosine. In surface plasmon resonance-microarray analysis, heterodimer of Nrf2(A502Y) and MafG displayed a binding specificity similar to that of MafG homodimer. The target genes activated by mutant Nrf2(A502Y)-small Maf heterodimer were largely different, albeit with some overlap, from those activated by wild-type Nrf2-small Maf, indicating that the array of target genes regulated by Nrf2-small Maf heterodimer differs substantially from that regulated by Maf homodimer in vivo. These results suggest that the distinct DNA binding profile of Nrf2-Maf heterodimer is biologically significant for Nrf2 to function as a key regulator of cytoprotective genes. Our contention is supported that the differential DNA binding specificity between Maf homodimers and Nrf2-Maf heterodimers establishes the differential gene regulation by these dimer-forming transcription factors.


Subject(s)
DNA/chemistry , Gene Expression Regulation , NF-E2-Related Factor 2/chemistry , Proto-Oncogene Proteins c-maf/chemistry , Amino Acid Sequence , Dimerization , Humans , Kinetics , MafG Transcription Factor/chemistry , Models, Molecular , Molecular Sequence Data , Mutation , NF-E2-Related Factor 2/metabolism , Oligonucleotide Array Sequence Analysis , Protein Binding , Proto-Oncogene Proteins c-maf/metabolism , Repressor Proteins/chemistry , Sequence Homology, Amino Acid , Surface Plasmon Resonance , Tyrosine/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...