A chemical biology screen identifies glucocorticoids that regulate c-maf expression by increasing its proteasomal degradation through up-regulation of ubiquitin

Blood ◽  
2007 ◽  
Vol 110 (12) ◽  
pp. 4047-4054 ◽  
Author(s):  
Xinliang Mao ◽  
A. Keith Stewart ◽  
Rose Hurren ◽  
Alessandro Datti ◽  
Xuegong Zhu ◽  
...  

AbstractThe oncogene c-maf is frequently overexpressed in multiple myeloma cell lines and patient samples and contributes to increased cellular proliferation in part by inducing cyclin D2 expression. To identify regulators of c-maf, we developed a chemical screen in NIH3T3 cells stably overexpressing c-maf and the cyclin D2 promoter driving luciferase. From a screen of 2400 off-patent drugs and chemicals, we identified glucocorticoids as c-maf–dependent inhibitors of cyclin D2 transactivation. In multiple myeloma cell lines, glucocorticoids reduced levels of c-maf protein without influencing corresponding mRNA levels. Subsequent studies demonstrated that glucocorticoids increased ubiquitination-dependent degradation of c-maf and up-regulated ubiquitin C mRNA. Moreover, ectopic expression of ubiquitin C recapitulated the effects of glucocorticoids, demonstrating regulation of c-maf protein through the abundance of the ubiquitin substrate. Thus, using a chemical biology approach, we identified a novel mechanism of action of glucocorticoids and a novel mechanism by which levels of c-maf protein are regulated by the abundance of the ubiquitin substrate.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 368-368
Author(s):  
Xinliang Mao ◽  
A. Keith Stewart ◽  
Rose Hurren ◽  
Alessandro Datti ◽  
Chang Xin Shi ◽  
...  

Abstract The oncogene c-maf is frequently over-expressed in multiple myeloma cell lines and patient samples and contributes to increased cellular proliferation in part by inducing cyclin D2 expression. Therefore, small molecules that inhibit c-maf and its targets could be useful chemical probes to better understand the role and regulation of this protein. We developed a high throughput chemical screen in NIH 3T3 cells stably over-expressing the promoter of the c-maf target cyclin D2 driving firefly luciferase. From a screen of 2400 off-patent drugs and chemicals, we identified 32 compounds that preferentially reduced cyclin D2 transactivation. Of these, 24 of the 32 hits belonged to the corticosteroid family of drugs. Indeed, the screen identified 24 of the 26 corticosteroids in the library. The most potent inhibitors were glucocorticoids such as dexamethasone. Mineralocorticoids such as fludrocortisone were weak hits, reflecting their weak glucocorticoid activity. The 24 glucocorticoids identified in this screen preferentially reduced cyclin D2 transactivation in the presence of c-maf. For example, the IC50 of dexamethasone was 11 ± 0.7 nM and >50 uM in NIH 3T3 cell with and without c-maf, respectively. Given the effects of glucocorticoids on c-maf in NIH 3T3 cells, we extended our investigation to multiple myeloma cell lines and demonstrated that nanomolar concentrations of the glucocorticoid dexamethasone reduced levels of c-maf protein and its target cyclin D2 within 6 hours of treatment. C-Maf was down regulated in isogenic MM1.S but not MM1.R cells respectively sensitive and resistant to dexamethasone. We also observed reductions in another c-maf target, beta integrin. Compared to cell lines RPMI 8226 and OCIMY5 that harbor the t(14;16) c-maf translocation, the concentration of dexamethasone required to reduce c-maf was approximately 50-fold lower in cell lines such as LP1 and OPM1 that lack the translocation but over-express c-maf. While dexamethasone reduced c-maf protein, no changes in levels of c-maf mRNA were detected. In both multiple myeloma and NIH3T3 cells, dexamethasone increased the ubiquitin-dependent destruction of c-maf. Finally, we linked glucocorticoids to c-maf ubiquitination by demonstrating that dexamethasone upregulated ubiquitin C mRNA at concentrations associated with the ubiquitination of c-maf. Moreover, ectopic expression of ubiquitin cDNA recapitulated the effects of dexamethasone and reduced levels of c-maf, suggesting that increased expression of ubiquitin C by dexamethasone is functionally important for dexamethasone’s effects on c-maf levels. Conclusion: a chemical biology screen identified glucocorticoids as c-maf dependent inhibitors of cyclin D2 transactivation. Glucocorticoids reduce c-maf by promoting its ubiquitination via the upregulation of ubiquitin C mRNA. This work provides new insights into the regulation of c-maf and has identified a novel mechanism by which glucocorticoids exert an anti-myeloma effect.


eLife ◽  
2016 ◽  
Vol 5 ◽  
Author(s):  
Andrew R Conery ◽  
Richard C Centore ◽  
Adrianne Neiss ◽  
Patricia J Keller ◽  
Shivangi Joshi ◽  
...  

Pharmacological inhibition of chromatin co-regulatory factors represents a clinically validated strategy to modulate oncogenic signaling through selective attenuation of gene expression. Here, we demonstrate that CBP/EP300 bromodomain inhibition preferentially abrogates the viability of multiple myeloma cell lines. Selective targeting of multiple myeloma cell lines through CBP/EP300 bromodomain inhibition is the result of direct transcriptional suppression of the lymphocyte-specific transcription factor IRF4, which is essential for the viability of myeloma cells, and the concomitant repression of the IRF4 target gene c-MYC. Ectopic expression of either IRF4 or MYC antagonizes the phenotypic and transcriptional effects of CBP/EP300 bromodomain inhibition, highlighting the IRF4/MYC axis as a key component of its mechanism of action. These findings suggest that CBP/EP300 bromodomain inhibition represents a viable therapeutic strategy for targeting multiple myeloma and other lymphoid malignancies dependent on the IRF4 network.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4769-4769
Author(s):  
Susan B. Ingersoll ◽  
Natalie D. Thoni ◽  
Farhana Ahmed ◽  
Kimberly A. Monahan ◽  
Lizette Caballero ◽  
...  

Abstract Aberrant methylation of promoter regions of tumor suppressor genes (TSG) has recently become recognized as an important epigenetic event in cancer and tumor progression including multiple myeloma. Interleukin-6 (IL-6), which is known to play a significant role in the pathophysiology of myeloma, regulates DNA methylation by inducing expression of FLI-1, a transcription factor of DNA methyltransferase-1 (DNMT-1), resulting in over-expression of DNMT-1 and thus hypermethylation of TSG. Despite this understanding, attempts to bring IL-6 blockade to the clinic have had limited success. We hypothesize that IL-6 regulation of epigenetic events (hypermethylation) may be an important pathway that could eventually allow rational chemotherapeutic/anit-IL-6 combinations. We have studied the correlation of IL-6 expression and dependence in myeloma cell lines and correlated that to the methylation profile of TSG, DcR1 and CDH1. Using three well-established multiple myeloma cell lines: U266B1 (U266), RPMI 8226 (RPMI), and KAS6/1 (KAS), we have confirmed that KAS is IL-6-dependent (exogenous IL-6 is needed) whereas the U266 and RPMI are IL-6 independent. To determine if blockade of the IL-6 pathway would inhibit the growth of the myeloma cell lines, these cells were grown in the presence of an anti-IL-6 antibody (B-E8; Cell Sciences, Canton, MA) that is known to block IL-6 signaling in the cells. We found that blocking IL-6 (by B-E8, 200 ng/ml) inhibited the growth of U266 (36% inhibition; n≥3, p<0.01) and KAS (68% inhibition; n≥3, p<0.001) cells, but not RPMI cells. We examined IL-6 expression in these three cell lines by RT-PCR and found that U266 expresses IL-6 mRNA, but RPMI and KAS cells do not. This IL-6 mRNA expression pattern correlates well with the anti-IL-6 cellular proliferation findings. Since RPMI is not dependent on the addition of exogenous IL-6 and blocking the IL-6 pathway had no effect on cell growth; therefore, we would not expect the cells to express IL-6. The U266 cells are not dependent on exogenous IL-6 but cellular proliferation can be blocked with an anti-IL-6 antibody; therefore, we expected that the cells would endogenously express IL-6. Furthermore, KAS cells are dependent on exogenous IL-6 and cell growth can be inhibited by anti-IL-6 antibody; therefore, we would not expect the cells to express IL-6. To determine if IL-6 sensitivity correlated with hypermethylation of TSG, we investigated the methylation status of the DcR1(tumor necrosis factor-related apoptosis inducing ligand decoy receptor 1) and CDH1 (Cadherin 1) loci. We have shown that CDH1 is methylated in U266 cells and un-methylated in RPMI cells. Experiments are underway to determine the methylation status in KAS cells and gene expression in all cell-lines for CDH1. Finally, we found that the RPMI and KAS cells are un-methylated and U266 cells are methylated at the DcR1 locus. We have found that in the RPMI and U266 cell lines that methylation of target TSG correlates with anti-IL-6 sensitivity. These data support our hypothesis that an IL-6-dependent pathway may regulate hypermethylation of important TSG in multiple myeloma. Newer chemotherapeutic agents that may affect methylation pathways are being studied in combination with IL-6 blockade.


2000 ◽  
Vol 111 (4) ◽  
pp. 1118-1121 ◽  
Author(s):  
A. Bellahcene ◽  
I. Van Riet ◽  
C. de Greef ◽  
N. Antoine ◽  
M. F. Young ◽  
...  

2008 ◽  
Vol 49 (7) ◽  
pp. 1374-1383 ◽  
Author(s):  
Antonino Neri ◽  
Sandra Marmiroli ◽  
Pierfrancesco Tassone ◽  
Luigia Lombardi ◽  
Lucia Nobili ◽  
...  

2004 ◽  
Vol 52 (5) ◽  
pp. 335-344 ◽  
Author(s):  
Naomi Gronich ◽  
Liat Drucker ◽  
Hava Shapiro ◽  
Judith Radnay ◽  
Shai Yarkoni ◽  
...  

BackgroundAccumulating reports indicate that statins widely prescribed for hypercholesteromia have antineoplastic activity. We hypothesized that because statins inhibit farnesylation of Ras that is often mutated in multiple myeloma (MM), as well as the production of interleukin (IL)-6, a key cytokine in MM, they may have antiproliferative and/or proapoptotic effects in this malignancy.MethodsU266, RPMI 8226, and ARH77 were treated with simvastatin (0-30 μM) for 5 days. The following aspects were evaluated: viability (IC50), cell cycle, cell death, cytoplasmic calcium ion levels, supernatant IL-6 levels, and tyrosine kinase activity.ResultsExposure of all cell lines to simvastatin resulted in reduced viability with IC50s of 4.5 μM for ARH77, 8 μM for RPMI 8226, and 13 μM for U266. The decreased viability is attributed to cell-cycle arrest (U266, G1; RPMI 8226, G2M) and cell death. ARH77 underwent apoptosis, whereas U266 and RPMI 8226 displayed a more necrotic form of death. Cytoplasmic calcium levels decreased significantly in all treated cell lines. IL-6 secretion from U266 cells was abrogated on treatment with simvastatin, whereas total tyrosine phosphorylation was unaffected.ConclusionsSimvastatin displays significant antimyeloma activity in vitro. Further research is warranted for elucidation of the modulated molecular pathways and clinical relevance.


Blood ◽  
2000 ◽  
Vol 95 (3) ◽  
pp. 1039-1046 ◽  
Author(s):  
G. Teoh ◽  
Y.-T. Tai ◽  
M. Urashima ◽  
S. Shirahama ◽  
M. Matsuzaki ◽  
...  

It has been reported that the activation of multiple myeloma (MM) cells by CD40 induces proliferation, growth arrest, and apoptosis. To determine whether the biologic sequelae of CD40 activation in MM cells depends on p53 function, we identified temperature-sensitive p53 mutations in the RPMI 8226 (tsp53E285K) and the HS Sultan (tsp53Y163H) MM cell lines. These cells were then used as a model system of inducible wtp53-like function because wild-type-like p53 is induced at permissive (30°C) but not at restrictive (37°C) temperatures. Using p21-luciferase reporter assays, we confirmed that CD40 induces p53 transactivation in RPMI 8226 and HS Sultan cells cultured under permissive, but not restrictive, conditions. Furthermore, CD40 activation of these MM cells under permissive, but not restrictive, temperatures increased the expression of p53 and p21 mRNA and protein. Importantly, CD40 activation induced the proliferation of RPMI 8226 and HS Sultan cells at restrictive temperatures and growth arrest and increased subG1 phase cells at permissive temperatures. These data confirmed that CD40 activation might have distinct biologic sequelae in MM cells, depending on their p53 status.


Sign in / Sign up

Export Citation Format

Share Document