Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Biochemistry ; 62(7): 1321-1329, 2023 04 04.
Article in English | MEDLINE | ID: mdl-36883372

ABSTRACT

The Myb transcription factor is involved in the proliferation of hematopoietic cells, and deregulation of its expression can lead to cancers such as leukemia. Myb interacts with various proteins, including the histone acetyltransferases p300 and CBP. Myb binds to a small domain of p300, the KIX domain (p300KIX), and inhibiting this interaction is a potential new drug discovery strategy in oncology. The available structures show that Myb binds to a very shallow pocket of the KIX domain, indicating that it might be challenging to identify inhibitors of this interaction. Here, we report the design of Myb-derived peptides which interact with p300KIX. We show that by mutating only two Myb residues that bind in or near a hotspot at the surface of p300KIX, it is possible to obtain single-digit nanomolar peptidic inhibitors of the Myb/p300KIX interaction that bind 400-fold tighter to p300KIX than wildtype Myb. These findings suggest that it might also be possible to design potent low molecular-weight compounds to disrupt the Myb/p300KIX interaction.


Subject(s)
E1A-Associated p300 Protein , Peptides , Proto-Oncogene Proteins c-myb , Peptides/pharmacology , Protein Binding , Proto-Oncogene Proteins c-myb/antagonists & inhibitors , Proto-Oncogene Proteins c-myb/chemistry , E1A-Associated p300 Protein/antagonists & inhibitors , E1A-Associated p300 Protein/chemistry
2.
Curr Drug Discov Technol ; 17(2): 183-190, 2020.
Article in English | MEDLINE | ID: mdl-30848204

ABSTRACT

BACKGROUND: Tubulin is the biochemical target for several clinically used anticancer drugs as it helps in the formation of mitotic spindle during mitosis stage of cell division. Many of the anti-cancer drugs are known to interact with tubulin and microtubules including some plant alkaloids, such as paclitaxel, etoposide and topotecan. In silico drug design of these molecules were performed prior to testing these drugs in vitro. In silico drug design of these anti-cancer drugs becomes a challenge due to the complex structure of target protein. This challenge was overcome by predicting the structure of the target protein (tubulin) by homology modeling. METHODS: In this study, computer aided drug designing approach was applied to predict the suitable docking site in target protein and the interaction of tubulin protein with paclitaxel, etoposide and topotecan was explored by molecular docking using Schrödinger software. Docking score and glide energy were determined with ligands to validate their anticancer properties. RESULTS: The results indicate that etoposide is the best drug for tubulin with a docking score of - 4.916 and glide energy of -46.470 kcal/mol compared to paclitaxel and topotecan. CONCLUSION: The testing of these drugs in silico provides an alternate to in vitro testing of these molecules on cancer cell lines which is a time and cost intensive process. The in silico study of parameters, such as docking score and glide energy, will help pharmacists in developing new molecules as targets for cancers in a time and cost-effective manner.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Drug Design , Gene Expression Regulation, Neoplastic/drug effects , Neoplasms/drug therapy , Proto-Oncogene Proteins c-myb/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/therapeutic use , Etoposide/chemistry , Etoposide/pharmacology , Etoposide/therapeutic use , Humans , Lactones/chemistry , Lactones/pharmacology , Lactones/therapeutic use , Ligands , Molecular Docking Simulation , Neoplasms/genetics , Paclitaxel/chemistry , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Proto-Oncogene Proteins c-myb/metabolism , Proto-Oncogene Proteins c-myb/ultrastructure , Sesquiterpenes/chemistry , Sesquiterpenes/pharmacology , Sesquiterpenes/therapeutic use , Structure-Activity Relationship , Topotecan/chemistry , Topotecan/pharmacology , Topotecan/therapeutic use
3.
J Exp Med ; 215(10): 2673-2685, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30209067

ABSTRACT

Pluripotent cells have been used to probe developmental pathways that are involved in genetic diseases and oncogenic events. To find new therapies that would target MYB-driven tumors, we developed a pluripotent zebrafish blastomere culture system. We performed a chemical genetic screen and identified retinoic acid agonists as suppressors of c-myb expression. Retinoic acid treatment also decreased c-myb gene expression in human leukemia cells. Translocations that drive overexpression of the oncogenic transcription factor MYB are molecular hallmarks of adenoid cystic carcinoma (ACC), a malignant salivary gland tumor with no effective therapy. Retinoic acid agonists inhibited tumor growth in vivo in ACC patient-derived xenograft models and decreased MYB binding at translocated enhancers, thereby potentially diminishing the MYB positive feedback loop driving ACC. Our findings establish the zebrafish pluripotent cell culture system as a method to identify modulators of tumor formation, particularly establishing retinoic acid as a potential new effective therapy for ACC.


Subject(s)
Blastomeres/immunology , Carcinoma, Adenoid Cystic/drug therapy , Proto-Oncogene Proteins c-myb/antagonists & inhibitors , Salivary Gland Neoplasms/drug therapy , Tretinoin/pharmacology , Zebrafish Proteins/antagonists & inhibitors , Zebrafish/immunology , Animals , Blastomeres/pathology , Carcinoma, Adenoid Cystic/genetics , Carcinoma, Adenoid Cystic/immunology , Carcinoma, Adenoid Cystic/pathology , Humans , Mice , Mice, Nude , Proto-Oncogene Proteins c-myb/genetics , Proto-Oncogene Proteins c-myb/immunology , Salivary Gland Neoplasms/genetics , Salivary Gland Neoplasms/immunology , Salivary Gland Neoplasms/pathology , U937 Cells , Xenograft Model Antitumor Assays , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/immunology
4.
Sci Rep ; 8(1): 13159, 2018 09 03.
Article in English | MEDLINE | ID: mdl-30177851

ABSTRACT

The transcription factor MYB plays key roles in hematopoietic cells and has been implicated the development of leukemia. MYB has therefore emerged as an attractive target for drug development. Recent work has suggested that targeting MYB by small-molecule inhibitors is feasible and that inhibition of MYB has potential as a therapeutic approach against acute myeloid leukemia. To facilitate the identification of small-molecule MYB inhibitors we have re-designed and improved a previously established cell-based screening assay and have employed it to screen a natural product library for potential inhibitors. Our work shows that teniposide and etoposide, chemotherapeutic agents causing DNA-damage by inhibiting topoisomerase II, potently inhibit MYB activity and induce degradation of MYB in AML cell lines. MYB inhibition is suppressed by caffeine, suggesting that MYB is inhibited indirectly via DNA-damage signalling. Importantly, ectopic expression of an activated version of MYB in pro-myelocytic NB4 cells diminished the anti-proliferative effects of teniposide, suggesting that podophyllotoxins disrupt the proliferation of leukemia cells not simply by inducing general DNA-damage but that their anti-proliferative effects are boosted by inhibition of MYB. Teniposide and etoposide therefore act like double-edged swords that might be particularly effective to inhibit tumor cells with deregulated MYB.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Etoposide/pharmacology , Gene Expression Regulation, Leukemic , Podophyllotoxin/pharmacology , Proto-Oncogene Proteins c-myb/antagonists & inhibitors , Teniposide/pharmacology , Cell Line, Tumor , DNA Damage , Dose-Response Relationship, Drug , High-Throughput Screening Assays , Humans , Leukocytes/drug effects , Leukocytes/metabolism , Leukocytes/pathology , Proto-Oncogene Proteins c-myb/genetics , Proto-Oncogene Proteins c-myb/metabolism , Signal Transduction , Small Molecule Libraries/pharmacology
6.
Biochem Biophys Res Commun ; 503(4): 2807-2813, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30103947

ABSTRACT

The regulation of natural killer (NK) cell activity is an important research goal for the development of immunotherapies. In this study, we identified transcription factors affecting NK cell activity. In particular, we screened transcription factors affected by interleukin-2 (IL-2) and transforming growth factor-beta (TGF-ß) by protein/DNA arrays using primary NK cells. We found that celastrol, a c-Myb inhibitor, inhibited NK-92 cells more strongly than any other inhibitors of transcription factor candidates. In addition, c-Myb and c-Myb-related signaling molecules, e.g., Nemo-like kinase (NLK) and c-Myc, were regulated by the activation status of NK cells, suggesting that c-Myb is a key regulator of NK cell activity. We also found that celastrol inhibits NK-92-cell-mediated cytotoxicity via the downregulation of NKG2D and granzyme B. Knockdown studies also showed that c-Myb is important for NK cell activation. In particular, the knockdown of c-Myb did not significantly affect NK cell proliferation and survival but decreased the secretion of IFN-γ and the cytotoxicity of NK cells. Our data demonstrate that c-Myb plays a critical role in the activation of NK cells and therefore is a therapeutic target for cancer and viral diseases.


Subject(s)
Cytotoxicity, Immunologic/drug effects , Immunologic Factors/pharmacology , Killer Cells, Natural/drug effects , Proto-Oncogene Proteins c-myb/genetics , Triterpenes/pharmacology , Cell Line , Cell Proliferation/drug effects , Gene Expression Regulation , Granzymes/genetics , Granzymes/immunology , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-2/pharmacology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Lymphocyte Activation/drug effects , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/immunology , Pentacyclic Triterpenes , Primary Cell Culture , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Proto-Oncogene Proteins c-myb/antagonists & inhibitors , Proto-Oncogene Proteins c-myb/immunology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/immunology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Transforming Growth Factor beta/pharmacology
8.
PLoS One ; 13(2): e0190934, 2018.
Article in English | MEDLINE | ID: mdl-29394256

ABSTRACT

Myb is a key regulator of hematopoietic progenitor cell proliferation and differentiation and has emerged as a potential target for the treatment of acute leukemia. Using a myeloid cell line with a stably integrated Myb-inducible reporter gene as a screening tool we have previously identified Celastrol, a natural compound with anti-tumor activity, as a potent Myb inhibitor that disrupts the interaction of Myb with the co-activator p300. We showed that Celastrol inhibits the proliferation of acute myeloid leukemia (AML) cells and prolongs the survival of mice in an in vivo model of AML, demonstrating that targeting Myb with a small-molecule inhibitor is feasible and might have potential as a therapeutic approach against AML. Recently we became aware that the reporter system used for Myb inhibitor screening also responds to inhibition of C/EBPß, a transcription factor known to cooperate with Myb in myeloid cells. By re-investigating the inhibitory potential of Celastrol we have found that Celastrol also strongly inhibits the activity of C/EBPß by disrupting its interaction with the Taz2 domain of p300. Together with previous studies our work reveals that Celastrol independently targets Myb and C/EBPß by disrupting the interaction of both transcription factors with p300. Myb, C/EBPß and p300 cooperate in myeloid-specific gene expression and, as shown recently, are associated with so-called super-enhancers in AML cells that have been implicated in the maintenance of the leukemia. We hypothesize that the ability of Celastrol to disrupt the activity of a transcriptional Myb-C/EBPß-p300 module might explain its promising anti-leukemic activity.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , CCAAT-Enhancer-Binding Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-myb/antagonists & inhibitors , Triterpenes/pharmacology , p300-CBP Transcription Factors/antagonists & inhibitors , 3T3-L1 Cells , Amino Acid Sequence , Animals , CCAAT-Enhancer-Binding Proteins/chemistry , CCAAT-Enhancer-Binding Proteins/genetics , Cell Line , Cell Proliferation/drug effects , Chickens , Cysteine/chemistry , Gene Expression/drug effects , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Pentacyclic Triterpenes , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins c-myb/chemistry , Quail , p300-CBP Transcription Factors/chemistry , p300-CBP Transcription Factors/genetics
9.
Curr Top Med Chem ; 17(30): 3256-3268, 2018 Feb 09.
Article in English | MEDLINE | ID: mdl-29231144

ABSTRACT

BACKGROUND: Protein c-Myb is a therapeutic target. Some sesquiterpene lactones suppress Myb-dependent gene expression, which results in their potential anti-cancer activity. MATERIAL & METHODS: Database ChEMBL is a representative of lactones for physicochemical and physiochemical properties. Data presented for 31 natural lactones are discussed in terms of quantitative structureactivity relationships with the objective to predict inhibitors of Myb-induced gene expression. Several constitutional descriptors are related to structure-activity. α-Methylene-γ-lactone groups enhance while OH functions worsen potency. The latter feature is in agreement with the fact that the more lipophilic the lactone, the greater the cytotoxicity because of the ability to cross lipoidal biomembranes. In general, numbers of π-systems and atoms, and polarizability enhance activity. Linear and nonlinear structure-activity models are developed, between lactones of a great structural diversity, to predict inhibitors of Myb-induced gene expression. Four variables (ML, UNC, TCO+OCOR, UNC+UNA) related to ATOM show a positive correlation because of the partial anionic and H-acceptor characters of O-atom. In most, CO group is conjugated. RESULT AND CONCLUSION: Term OH shows negative coefficients because of the partial cationic quality of H-atom and because OH forms H-bonds with CO, causing them to be less H-acceptor. s-trans-s-trans-Germacranolide structure is the most active. Coefficients standard errors result acceptable in almost all equations. After cross-validation, linear equations for lactones, pseudoguaianolides and germacranolides are the most predictive. Most descriptors are constitutional variables.


Subject(s)
Biological Products/pharmacology , Gene Expression Regulation/drug effects , Lactones/pharmacology , Proto-Oncogene Proteins c-myb/antagonists & inhibitors , Quantitative Structure-Activity Relationship , Sesquiterpenes/pharmacology , Biological Products/chemistry , Dose-Response Relationship, Drug , Humans , Lactones/chemistry , Models, Molecular , Molecular Structure , Proto-Oncogene Proteins c-myb/genetics , Sesquiterpenes/chemistry
10.
J Natl Cancer Inst ; 109(9)2017 09 01.
Article in English | MEDLINE | ID: mdl-28954282

ABSTRACT

Background: Adenoid cystic carcinoma (ACC) is an aggressive cancer with no curative treatment for patients with recurrent/metastatic disease. The MYB-NFIB gene fusion is the main genomic hallmark and a potential therapeutic target. Methods: Oncogenic signaling pathways were studied in cultured cells and/or tumors from 15 ACC patients. Phospho-receptor tyrosine kinase (RTK) arrays were used to study the activity of RTKs. Effects of RTK inhibition on cell proliferation were analyzed with AlamarBlue, sphere assays, and two ACC xenograft models (n = 4-9 mice per group). The molecular effects of MYB-NFIB knockdown and IGF1R inhibition were studied with quantitative polymerase chain reaction, immunoblot, and gene expression microarrays. All statistical tests were two-sided. Results: The MYB-NFIB fusion drives proliferation of ACC cells and is crucial for spherogenesis. Intriguingly, the fusion is regulated through AKT-dependent signaling induced by IGF1R overexpression and is downregulated upon IGF1R-inhibition (% expression of control ± SD = 27.2 ± 1.3, P < .001). MYB-NFIB regulates genes involved in cell cycle control, DNA replication/repair, and RNA processing. The transcriptional program induced by MYB-NFIB affects critical oncogenic mediators normally controlled by MYC and is reversed by pharmacological inhibition of IGF1R. Co-activation of epidermal growth factor receptor (EGFR) and MET promoted proliferation of ACC cells, and combined targeting of IGFR1/EGFR/MET induced differentiation and synergistically inhibited the growth of patient-derived xenografted ACCs (ACCX5M1, % growth of control ± SD = 34.9 ± 20.3, P = .006; ACCX6, % growth of control ± SD = 24.1 ± 17.5, P = .04). Conclusions: MYB-NFIB is an oncogenic driver and a key therapeutic target in ACC that is regulated by AKT-dependent IGF1R signaling. Our studies uncover a new strategy to target an oncogenic transcriptional master regulator and provide new important insights into the biology and treatment of ACC.


Subject(s)
Carcinoma, Adenoid Cystic/genetics , Carcinoma, Adenoid Cystic/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-myb/metabolism , Receptors, Somatomedin/metabolism , Signal Transduction , Adult , Aged , Aged, 80 and over , Animals , Apoptosis , Biomarkers, Tumor , Carcinoma, Adenoid Cystic/drug therapy , Carcinoma, Adenoid Cystic/pathology , Cell Cycle , Cell Proliferation/genetics , Cluster Analysis , Disease Models, Animal , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Insulin-Like Growth Factor II/pharmacology , Male , Middle Aged , Neoplasm Grading , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-myb/antagonists & inhibitors , Proto-Oncogene Proteins c-myb/genetics , Receptor, IGF Type 1 , Signal Transduction/drug effects , Transcription, Genetic , Xenograft Model Antitumor Assays
11.
Leukemia ; 31(10): 2057-2064, 2017 10.
Article in English | MEDLINE | ID: mdl-28260788

ABSTRACT

Oncogenic driver mutations are those that provide a proliferative or survival advantage to neoplastic cells, resulting in clonal selection. Although most cancer-causing mutations have been detected in the protein-coding regions of the cancer genome; driver mutations have recently also been discovered within noncoding genomic sequences. Thus, a current challenge is to gain precise understanding of how these unique genomic elements function in cancer pathogenesis, while clarifying mechanisms of gene regulation and identifying new targets for therapeutic intervention. Here we report a C-to-T single nucleotide transition that occurs as a somatic mutation in noncoding sequences 4 kb upstream of the transcriptional start site of the LMO1 oncogene in primary samples from patients with T-cell acute lymphoblastic leukaemia. This single nucleotide alteration conforms to an APOBEC-like cytidine deaminase mutational signature, and generates a new binding site for the MYB transcription factor, leading to the formation of an aberrant transcriptional enhancer complex that drives high levels of expression of the LMO1 oncogene. Since APOBEC-signature mutations are common in a broad spectrum of human cancers, we suggest that noncoding nucleotide transitions such as the one described here may activate potent oncogenic enhancers not only in T-lymphoid cells but in other cell lineages as well.


Subject(s)
APOBEC Deaminases/metabolism , DNA-Binding Proteins/biosynthesis , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Leukemic/genetics , LIM Domain Proteins/biosynthesis , Neoplasm Proteins/biosynthesis , Point Mutation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Transcription Factors/biosynthesis , Transcriptome , 5' Untranslated Regions/genetics , Base Sequence , Binding Sites , Cell Line, Tumor , Child , Chromatin Immunoprecipitation , DNA, Neoplasm/genetics , DNA-Binding Proteins/genetics , Genes, myb , Humans , Jurkat Cells , LIM Domain Proteins/genetics , Neoplasm Proteins/genetics , Polymorphism, Single Nucleotide , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogene Proteins c-myb/antagonists & inhibitors , Proto-Oncogene Proteins c-myb/genetics , Proto-Oncogene Proteins c-myb/metabolism , RNA Interference , RNA, Small Interfering/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
12.
Sci Rep ; 7: 41094, 2017 01 23.
Article in English | MEDLINE | ID: mdl-28112219

ABSTRACT

c-Myb is a transcription factor that plays a key role in cell proliferation, differentiation, and apoptosis. It has been reported that c-Myb is expressed within the chicken otic placode, but whether c-Myb exists in the mammalian cochlea, and how it exerts its effects, has not been explored yet. Here, we investigated the expression of c-Myb in the postnatal mouse cochlea and HEI-OC1 cells and found that c-Myb was expressed in the hair cells (HCs) of mouse cochlea as well as in cultured HEI-OC1 cells. Next, we demonstrated that c-Myb expression was decreased in response to neomycin treatment in both cochlear HCs and HEI-OC1 cells, suggesting an otoprotective role for c-Myb. We then knocked down c-Myb expression with shRNA transfection in HEI-OC1 cells and found that c-Myb knockdown decreased cell viability, increased expression of pro-apoptotic factors, and enhanced cell apoptosis after neomycin insult. Mechanistic studies revealed that c-Myb knockdown increased cellular levels of reactive oxygen species and decreased Bcl-2 expression, both of which are likely to be responsible for the increased sensitivity of c-Myb knockdown cells to neomycin. This study provides evidence that c-Myb might serve as a new target for the prevention of aminoglycoside-induced HC loss.


Subject(s)
Hair Cells, Auditory/pathology , Mitochondria/drug effects , Protein Synthesis Inhibitors/pharmacology , Proto-Oncogene Proteins c-myb/genetics , Aminoglycosides/pharmacology , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation/drug effects , Hair Cells, Auditory/drug effects , Mice , Mitochondria/metabolism , Neomycin/pharmacology , Proto-Oncogene Proteins c-myb/antagonists & inhibitors , Reactive Oxygen Species/metabolism
13.
Exp Hematol ; 47: 31-35, 2017 03.
Article in English | MEDLINE | ID: mdl-28017646

ABSTRACT

The transcription factor Myb is a key regulator of hematopoietic cell proliferation, differentiation, and survival and has been implicated in the development of leukemia and several other human cancers. Pharmacological inhibition of Myb is therefore emerging as a potential therapeutic strategy. Recently, the first low-molecular-weight compounds that show Myb inhibitory activity have been identified. Characterization of these compounds suggests disruption of the protein-protein-interaction of Myb and the coactivator p300 as a suitable strategy to inhibit Myb.


Subject(s)
Antineoplastic Agents/pharmacology , Molecular Targeted Therapy , Proto-Oncogene Proteins c-myb/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Gene Expression Regulation, Leukemic/drug effects , Humans , Leukemia/drug therapy , Leukemia/etiology , Leukemia/metabolism , Protein Binding , Proto-Oncogene Proteins c-myb/chemistry , Proto-Oncogene Proteins c-myb/genetics , Proto-Oncogene Proteins c-myb/metabolism , Signal Transduction/drug effects , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation
14.
Oncotarget ; 7(52): 86300-86312, 2016 Dec 27.
Article in English | MEDLINE | ID: mdl-27863435

ABSTRACT

SETBP1 missense mutations have been frequently identified in multiple myeloid neoplasms; however, their oncogenic potential remains unclear. Here we show that expression of Setbp1 mutants carrying two such mutations in mouse bone marrow progenitors efficiently induced development of acute myeloid leukemias (AMLs) in irradiated recipient mice with significantly shorter latencies and greater penetrance than expression of wild-type Setbp1, suggesting that these mutations are highly oncogenic. The increased oncogenicity of Setbp1 missense mutants could be due in part to their capability to drive significantly higher target gene transcription. We further identify Myb as a critical mediator of Setbp1-induced self-renewal as its knockdown caused efficient differentiation of myeloid progenitors immortalized by wild-type Setbp1 and Setbp1 missense mutants. Interestingly, Myb is also a direct transcriptional target of Setbp1 and Setbp1 missense mutants as they directly bind to the Myb locus in immortalized cells and dramatically activate a critical enhancer/promoter region of Myb in luciferase reporter assays. Furthermore, Myb knockdown in Setbp1 and Setbp1 missense mutations-induced AML cells also efficiently induced their differentiation in culture and significantly prolonged the survival of their secondary recipient mice, suggesting that targeting MYB pathway could be a promising strategy for treating human myeloid neoplasms with SETBP1 activation.


Subject(s)
Carrier Proteins/physiology , Leukemia, Myeloid, Acute/etiology , Nuclear Proteins/physiology , Proto-Oncogene Proteins c-myb/physiology , Animals , Carrier Proteins/genetics , Female , Homeobox A10 Proteins , Homeodomain Proteins/genetics , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred C57BL , Mutation, Missense , Nuclear Proteins/genetics , Proto-Oncogene Proteins c-myb/antagonists & inhibitors
15.
Mol Cancer Ther ; 15(12): 2905-2915, 2016 12.
Article in English | MEDLINE | ID: mdl-27707899

ABSTRACT

The transcription factor c-Myb is essential for the proliferation of hematopoietic cells and has been implicated in the development of leukemia and other human cancers. Pharmacologic inhibition of Myb is therefore emerging as a potential therapeutic strategy for these diseases. By using a Myb reporter cell line, we have identified plumbagin and several naphthoquinones as potent low-molecular weight Myb inhibitors. We demonstrate that these compounds inhibit c-Myb by binding to the c-Myb transactivation domain and disrupting the cooperation of c-Myb with the coactivator p300, a major driver of Myb activity. Naphthoquinone-induced inhibition of c-Myb suppresses Myb target gene expression and induces the differentiation of the myeloid leukemia cell line HL60. We demonstrate that murine and human primary acute myeloid leukemia cells are more sensitive to naphthoquinone-induced inhibition of clonogenic proliferation than normal hematopoietic progenitor cells. Overall, our work demonstrates for the first time the potential of naphthoquinones as small-molecule Myb inhibitors that may have therapeutic potential for the treatment of leukemia and other tumors driven by deregulated Myb. Mol Cancer Ther; 15(12); 2905-15. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , E1A-Associated p300 Protein/metabolism , Leukemia, Myeloid, Acute/metabolism , Proto-Oncogene Proteins c-myb/metabolism , Antineoplastic Agents/chemistry , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Leukemic/drug effects , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Naphthoquinones/chemistry , Naphthoquinones/pharmacology , Protein Binding/drug effects , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins c-myb/antagonists & inhibitors , Proto-Oncogene Proteins c-myb/chemistry , Reactive Oxygen Species/metabolism
16.
Sci Rep ; 6: 28446, 2016 06 29.
Article in English | MEDLINE | ID: mdl-27354262

ABSTRACT

We have recently demonstrated that the transcription factor MYB can modulate several cancer-associated phenotypes in pancreatic cancer. In order to understand the molecular basis of these MYB-associated changes, we conducted deep-sequencing of transcriptome of MYB-overexpressing and -silenced pancreatic cancer cells, followed by in silico pathway analysis. We identified significant modulation of 774 genes upon MYB-silencing (p < 0.05) that were assigned to 25 gene networks by in silico analysis. Further analyses placed genes in our RNA sequencing-generated dataset to several canonical signalling pathways, such as cell-cycle control, DNA-damage and -repair responses, p53 and HIF1α. Importantly, we observed downregulation of the pancreatic adenocarcinoma signaling pathway in MYB-silenced pancreatic cancer cells exhibiting suppression of EGFR and NF-κB. Decreased expression of EGFR and RELA was validated by both qPCR and immunoblotting and they were both shown to be under direct transcriptional control of MYB. These observations were further confirmed in a converse approach wherein MYB was overexpressed ectopically in a MYB-null pancreatic cancer cell line. Our findings thus suggest that MYB potentially regulates growth and genomic stability of pancreatic cancer cells via targeting complex gene networks and signaling pathways. Further in-depth functional studies are warranted to fully understand MYB signaling in pancreatic cancer.


Subject(s)
Gene Regulatory Networks/genetics , Proto-Oncogene Proteins c-myb/metabolism , Signal Transduction/genetics , Cell Line, Tumor , Down-Regulation , ErbB Receptors/genetics , ErbB Receptors/metabolism , Genomic Instability , High-Throughput Nucleotide Sequencing , Humans , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-myb/antagonists & inhibitors , Proto-Oncogene Proteins c-myb/genetics , RNA Interference , RNA, Small Interfering/metabolism , Sequence Analysis, DNA , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Up-Regulation
17.
Bioorg Med Chem Lett ; 26(4): 1276-8, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26774656

ABSTRACT

Antisense oligonucleotides (ASOs) have a great therapeutic potential for the modulation of gene expression because of the high specificity. The major obstacles for clinical application are enzymatic degradation and low uptake into cells in vivo. In this study, we prepared the conjugate comprising Tat peptide and ASO with phosphorothioate linkages in a simple manner; azide alkyne Huisgen cycloaddition using a copper catalyst. The obtained conjugate showed a high stability in serum, compared with the conjugate with phosphodiester linkages. The conjugates with antisense for c-myb that is transcriptional factor concerning cell growth inhibited the cell proliferation in a dose dependent manner sequence-specifically. These findings suggest Tat-mediated ASOs delivery is useful for the treatment of various diseases.


Subject(s)
Gene Products, tat/metabolism , Oligonucleotides, Antisense/chemistry , Peptides/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Click Chemistry , Gene Silencing , Humans , Phosphates/chemistry , Proto-Oncogene Proteins c-myb/antagonists & inhibitors , Proto-Oncogene Proteins c-myb/genetics , Proto-Oncogene Proteins c-myb/metabolism
19.
Cell Death Differ ; 22(12): 1906-21, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25857263

ABSTRACT

The transcription factor MYB has a key role in hematopoietic progenitor cells (HPCs) lineage choice, by enhancing erythropoiesis at the expense of megakaryopoiesis. We previously demonstrated that MYB controls erythroid versus megakaryocyte lineage decision by transactivating KLF1 and LMO2 expression. To further unravel the molecular mechanisms through which MYB affects lineage fate decision, we performed the integrative analysis of miRNA and mRNA changes in MYB-silenced human primary CD34+ HPCs. Among the miRNAs with the highest number of predicted targets, we focused our studies on hsa-miR-486-3p by demonstrating that MYB controls miR-486-3p expression through the transactivation of its host gene, ankyrin-1 (ANK1) and that miR-486-3p affects HPCs commitment. Indeed, overexpression and knockdown experiments demonstrated that miR-486-3p supports the erythropoiesis while restraining the megakaryopoiesis. Of note, miR-486-3p also favors granulocyte differentiation while repressing the macrophage differentiation. To shed some light on the molecular mechanisms through which miR-486-3p affects HPCs lineage commitment, we profiled the gene expression changes upon miR-486-3p overexpression in CD34+ cells. Among the genes downregulated in miR-486-3p-overexpressing HPCs and computationally predicted to be miR-486-3p targets, we identified MAF as a miR-486-3p target by 3'UTR luciferase reporter assay. Noteworthy, MAF overexpression was able to partially reverse the effects of miR-486-3p overexpression on erythroid versus megakaryocyte lineage choice. Moreover, the MYB/MAF co-silencing constrained the skewing of erythroid versus megakaryocyte lineage commitment in MYB-silenced CD34+ cells, by restraining the expansion of megakaryocyte lineage while partially rescuing the impairment of erythropoiesis. Therefore, our data collectively demonstrate that MYB favors erythropoiesis and restrains megakaryopoiesis through the transactivation of miR-486-3p expression and the subsequent downregulation of MAF. As a whole, our study uncovers the MYB/miR-486-3p/MAF axis as a new mechanism underlying the MYB-driven control of erythroid versus megakaryocyte lineage fate decision.


Subject(s)
MicroRNAs/metabolism , Proto-Oncogene Proteins c-maf/metabolism , Proto-Oncogene Proteins c-myb/metabolism , 3' Untranslated Regions , Ankyrins/genetics , Ankyrins/metabolism , Antigens, CD34/metabolism , Cell Differentiation , Cell Lineage , Cells, Cultured , Chondrogenesis , Down-Regulation , Erythroid Cells/cytology , Erythroid Cells/metabolism , Genes, Reporter , Genetic Loci , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Megakaryocytes/cytology , Megakaryocytes/metabolism , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Oligonucleotides, Antisense/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins c-maf/antagonists & inhibitors , Proto-Oncogene Proteins c-maf/genetics , Proto-Oncogene Proteins c-myb/antagonists & inhibitors , Proto-Oncogene Proteins c-myb/genetics , RNA Interference
20.
Mol Cancer Ther ; 14(6): 1276-85, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25740244

ABSTRACT

The transcription factor c-Myb is highly expressed in hematopoietic progenitor cells and controls the transcription of genes important for lineage determination, cell proliferation, and differentiation. Deregulation of c-Myb has been implicated in the development of leukemia and certain other types of human cancer. c-Myb activity is highly dependent on the interaction of the c-Myb with the KIX domain of the coactivator p300, making the disruption of this interaction a reasonable strategy for the development of Myb inhibitors. Here, we have used bacterial Autodisplay to develop an in vitro binding assay that mimics the interaction of Myb and the KIX domain of p300. We have used this binding assay to investigate the potential of Naphthol AS-E phosphate, a compound known to bind to the KIX domain, to disrupt the interaction between Myb and p300. Our data show that Naphthol AS-E phosphate interferes with the Myb-KIX interaction in vitro and inhibits Myb activity in vivo. By using several human leukemia cell lines, we demonstrate that Naphthol AS-E phosphate suppresses the expression of Myb target genes and induces myeloid differentiation and apoptosis. Our work identifies Naphthol AS-E phosphate as the first low molecular weight compound that inhibits Myb activity by disrupting its interaction with p300, and suggests that inhibition of the Myb-KIX interaction might be a useful strategy for the treatment of leukemia and other tumors caused by deregulated c-Myb.


Subject(s)
E1A-Associated p300 Protein/metabolism , Naphthols/pharmacology , Organophosphates/pharmacology , Proto-Oncogene Proteins c-myb/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Binding Sites/genetics , Blotting, Western , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , E1A-Associated p300 Protein/genetics , Gene Expression Regulation, Leukemic/genetics , HL-60 Cells , Humans , Microscopy, Fluorescence , Naphthols/metabolism , Organophosphates/metabolism , Protein Binding/drug effects , Proto-Oncogene Proteins c-myb/antagonists & inhibitors , Proto-Oncogene Proteins c-myb/genetics , Reverse Transcriptase Polymerase Chain Reaction , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...