Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Org Lett ; 26(15): 3054-3059, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38557107

ABSTRACT

While pentacyclic triterpenoids have a rich history in chemistry and biology, the challenges associated with their asymmetric synthesis contribute to the current reality that medicinal exploration in the area is largely constrained to natural product derivatization. To address this deficiency, a function-oriented synthesis of pentacyclic triterpenoids was pursued. Overall, we report a divergent synthesis of 26-norgermanicol and 26-norlupeol and we have identified a new class of androgen receptor antagonist that is ∼6× more potent than lupeol.


Subject(s)
Biological Products , Triterpenes , Pentacyclic Triterpenes , Triterpenes/pharmacology , Androgen Receptor Antagonists/pharmacology , Biological Products/pharmacology
3.
Front Oncol ; 12: 857590, 2022.
Article in English | MEDLINE | ID: mdl-35574319

ABSTRACT

Background: Among women, breast cancer is the leading cause of cancer-related death worldwide. Estrogen receptor α-positive (ERα+) breast cancer accounts for 70% of all breast cancer subtypes. Although ERα+ breast cancer initially responds to estrogen deprivation or blockade, the emergence of resistance compels the use of more aggressive therapies. While ERα is a driver in ERα+ breast cancer, ERß plays an inhibitory role in several different cancer types. To date, the lack of highly selective ERß agonists without ERα activity has limited the exploration of ERß activation as a strategy for ERα+ breast cancer. Methods: We measured the expression levels of ESR1 and ESR2 genes in immortalized mammary epithelial cells and different breast cancer cell lines. The viability of ERα+ breast cancer cell lines upon treatments with specific ERß agonists, including OSU-ERb-12 and LY500307, was assessed. The specificity of the ERß agonists, OSU-ERb-12 and LY500307, was confirmed by reporter assays. The effects of ERß agonists on cell proliferation, cell cycle, apoptosis, colony formation, cell migration, and expression of tumor suppressor proteins were analyzed. The expression of ESR2 and genes containing ERE-AP1 composite response elements was examined in ERα+ human breast cancer samples to determine the correlation between ESR2 expression and overall survival and that of putative ESR2-regulated genes. Results: In this study, we demonstrate the efficacy of highly selective ERß agonists in ERα+ breast cancer cell lines and drug-resistant derivatives. ERß agonists blocked cell proliferation, migration, and colony formation and induced apoptosis and S and/or G2/M cell-cycle arrest of ERα+ breast cancer cell lines. Also, increases in the expression of the key tumor suppressors FOXO1 and FOXO3a were noted. Importantly, the strong synergy between ERß agonists and ERα antagonists suggested that the efficacy of ERß agonists is maximized by combination with ERα blockade. Lastly, ESR2 (ERß gene) expression was negatively correlated with ESR1 (ERα gene) and CCND1 RNA expression in human metastatic ERα+/HER2- breast cancer samples. Conclusion: Our results demonstrate that highly selective ERß agonists attenuate the viability of ERα+ breast cancer cell lines in vitro and suggest that this therapeutic strategy merits further evaluation for ERα+ breast cancer.

4.
J Med Chem ; 64(13): 9330-9353, 2021 07 08.
Article in English | MEDLINE | ID: mdl-34181409

ABSTRACT

Selective agonism of the estrogen receptor (ER) subtypes, ERα and ERß, has historically been difficult to achieve due to the high degree of ligand-binding domain structural similarity. Multiple efforts have focused on the use of classical organic scaffolds to model 17ß-estradiol geometry in the design of ERß selective agonists, with several proceeding to various stages of clinical development. Carborane scaffolds offer many unique advantages including the potential for novel ligand/receptor interactions but remain relatively unexplored. We synthesized a series of para-carborane estrogen receptor agonists revealing an ERß selective structure-activity relationship. We report ERß agonists with low nanomolar potency, greater than 200-fold selectivity for ERß over ERα, limited off-target activity against other nuclear receptors, and only sparse CYP450 inhibition at very high micromolar concentrations. The pharmacological properties of our para-carborane ERß selective agonists measure favorably against clinically developed ERß agonists and support further evaluation of carborane-based selective estrogen receptor modulators.


Subject(s)
Boron Compounds/pharmacology , Estrogen Receptor beta/agonists , Estrogens/pharmacology , Boron Compounds/chemical synthesis , Boron Compounds/chemistry , Dose-Response Relationship, Drug , Estrogens/chemical synthesis , Estrogens/chemistry , HEK293 Cells , Humans , Molecular Structure , Structure-Activity Relationship
5.
Elife ; 92020 10 19.
Article in English | MEDLINE | ID: mdl-33073769

ABSTRACT

As part of the Reproducibility Project: Cancer Biology, we published a Registered Report (Phelps et al., 2016) that described how we intended to replicate selected experiments from the paper 'Coding-independent regulation of the tumor suppressor PTEN by competing endogenous mRNAs' (Tay et al., 2011). Here, we report the results. We found depletion of putative PTEN competing endogenous mRNAs (ceRNAs) in DU145 cells did not impact PTEN 3'UTR regulation using a reporter, while the original study reported decreased activity when SERINC1, VAPA, and CNOT6L were depleted (Figure 3C; Tay et al., 2011). Using the same reporter, we found decreased activity when ceRNA 3'UTRs were overexpressed, while the original study reported increased activity (Figure 3D; Tay et al., 2011). In HCT116 cells, ceRNA depletion resulted in decreased PTEN protein levels, a result similar to the findings reported in the original study (Figure 3G,H; Tay et al., 2011); however, while the original study reported an attenuated ceRNA effect in microRNA deficient (DicerEx5) HCT116 cells, we observed increased PTEN protein levels. Further, we found depletion of the ceRNAs VAPA or CNOT6L did not statistically impact DU145, wild-type HCT116, or DicerEx5 HCT116 cell proliferation. The original study reported increased DU145 and wild-type HCT116 cell proliferation when these ceRNAs were depleted, which was attenuated in the DicerEx5 HCT116 cells (Figure 5B; Tay et al., 2011). Differences between the original study and this replication attempt, such as variance between biological repeats, are factors that might have influenced the results. Finally, we report meta-analyses for each result.


Subject(s)
Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , PTEN Phosphohydrolase/genetics , RNA, Messenger/genetics , HCT116 Cells , Humans , Male , MicroRNAs/genetics , MicroRNAs/metabolism , PTEN Phosphohydrolase/metabolism , RNA, Messenger/metabolism
6.
J Biol Chem ; 292(46): 18924-18936, 2017 11 17.
Article in English | MEDLINE | ID: mdl-28900037

ABSTRACT

The transcription factor C/EBPα is essential for myeloid differentiation and is frequently dysregulated in acute myeloid leukemia. Although studied extensively, the precise regulation of its gene by upstream factors has remained largely elusive. Here, we investigated its transcriptional activation during myeloid differentiation. We identified an evolutionarily conserved octameric sequence, CCCAGCAG, ∼100 bases upstream of the CEBPA transcription start site, and demonstrated through mutational analysis that this sequence is crucial for C/EBPα expression. This sequence is present in the genes encoding C/EBPα in humans, rodents, chickens, and frogs and is also present in the promoters of other C/EBP family members. We identified that ZNF143, the human homolog of the Xenopus transcriptional activator STAF, specifically binds to this 8-bp sequence to activate C/EBPα expression in myeloid cells through a mechanism that is distinct from that observed in liver cells and adipocytes. Altogether, our data suggest that ZNF143 plays an important role in the expression of C/EBPα in myeloid cells.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha/genetics , Myeloid Cells/cytology , Promoter Regions, Genetic , Trans-Activators/metabolism , Transcriptional Activation , Base Sequence , Cell Line , Conserved Sequence , Gene Expression Regulation, Developmental , Hematopoiesis , Humans , Myeloid Cells/metabolism , Protein Binding
7.
Expert Rev Mol Diagn ; 16(3): 277-84, 2016.
Article in English | MEDLINE | ID: mdl-26671731

ABSTRACT

Multiple myeloma (MM) is a hematologic malignancy of plasma cells (PCs). In the United States, MM accounts for approximately 1% of all diagnoses and 2% of all cancer-related deaths. Although MM is a treatable disease, most patients eventually relapse, and despite the development of numerous treatment options it is still considered incurable. Mechanisms of communication between MM-PCs and bone marrow microenvironment, including cell-cell contacts and release of pro-survival factors, promote cancer cell survival and drug resistance. Recently, the importance of extracellular vesicles (EVs) as mechanisms of communication between MM cells and other cells in the microenvironment has been reported. In this review, the authors provide the update on the biology and clinical aspects of EVs in MM.


Subject(s)
Biomarkers, Tumor/metabolism , Extracellular Vesicles/pathology , Multiple Myeloma/pathology , Animals , Extracellular Vesicles/metabolism , Humans , Multiple Myeloma/metabolism , Tumor Microenvironment
8.
Oncotarget ; 6(31): 31134-50, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26429859

ABSTRACT

Multiple myeloma (MM) is a hematological malignancy of plasma cells in the bone marrow. Despite multiple treatment options, MM is inevitably associated with drug resistance and poor outcomes. Histone deacetylase inhibitors (HDACi's) are promising novel chemotherapeutics undergoing evaluation in clinical trials for the potential treatment of patients with MM. Although in preclinical studies HDACi's have proven anti-myeloma activity, but in the clinic single-agent HDACi treatments have been limited due to low tolerability. Improved clinical outcomes were reported only when HDACi's were combined with other drugs. Here, we show that a novel pan-HDACi AR-42 downregulates CD44, a glycoprotein that has been associated with lenalidomide and dexamethasone resistance in myeloma both in vitro and in vivo. We also show that this CD44 downregulation is in part mediated by miR-9-5p, targeting insulin-like growth factor 2 mRNA binding protein 3 (IGF2BP3), which directly binds to CD44 mRNA and increases its stability. Importantly, we also demonstrate that AR-42 enhances anti-myeloma activity of lenalidomide in primary MM cells isolated from lenalidomide resistant patients and in in vivo MM mouse model. Thus, our findings shed light on potential novel combinatorial therapeutic approaches modulating CD44 expression, which may help overcome lenalidomide resistance in myeloma patients.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Drug Resistance, Neoplasm/drug effects , Hyaluronan Receptors/metabolism , Multiple Myeloma/drug therapy , Phenylbutyrates/pharmacology , Thalidomide/analogs & derivatives , Animals , Apoptosis , Blotting, Western , Cell Proliferation , Drug Therapy, Combination , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Gene Expression Profiling , Histone Deacetylase Inhibitors/pharmacology , Humans , Hyaluronan Receptors/genetics , Immunoenzyme Techniques , Lenalidomide , Mice , Mice, Nude , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Thalidomide/pharmacology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
J Clin Endocrinol Metab ; 100(10): E1370-7, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26274343

ABSTRACT

CONTEXT: We previously showed that a long noncoding RNA gene, PTCSC3, located close to the variant rs944289 that predisposes to papillary thyroid carcinoma (PTC) might target the S100A4 gene. OBJECTIVE: The aim was to investigate the impact of PTCSC3 on S100A4 expression and its role in cancer development. DESIGN: S100A4 abundance was analyzed by quantitative PCR (qPCR) in unaffected and tumor tissue from n = 73 PTC patients. The expression of PTCSC3 and S100A4 was studied in BCPAP and TPC-1 cell lines with forced expression of PTCSC3 by qPCR. Expression of S100A4 target genes (VEGF and MMP-9) was studied in the BCPAP cell line with forced expression of PTCSC3 by qPCR, reverse transcriptase PCR, and Western blot. The impact of PTCSC3 on BCPAP motility and invasiveness was analyzed by the Transwell and Matrigel assays, respectively. SETTING: This was a laboratory-based study using cells from clinical samples and thyroid cancer cell lines. MAIN OUTCOME AND MEASURE: We aimed to find evidence for a link between the expression of PTCSC3 and thyroid carcinogenesis. RESULTS: Expression data from PTC cell lines pinpointed S100A4 as the most significantly downregulated gene in the presence of PTCSC3. S100A4 was upregulated in tumor tissue (P = 9.33 × 10(-7)) while PTCSC3 was strongly downregulated (P = 2.2 × 10(-16)). S100A4 transcription was moderately correlated with PTCSC3 expression in unaffected thyroid tissue (r = 0.429, P = .0001), and strongly in unaffected tissue of patients with the risk allele of rs944289 (r = 0.685, P = 7.88 × 10(-5)). S100A4, VEGF, and MMP-9 were suppressed in the presence of PTCSC3 (P = .0051, P = .0090, and P =.0037, respectively). PTC cells expressing PTCSC3 showed reduction in motility and invasiveness (P = 4.52 × 10(-5) and P = 1.0 × 10(-4), respectively). CONCLUSIONS: PTCSC3 downregulates S100A4, leading to a reduction in cell motility and invasiveness. We propose that PTCSC3 impacts PTC predisposition and carcinogenesis through the S100A4 pathway.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Papillary/genetics , Cell Proliferation/genetics , Neoplasm Invasiveness/genetics , RNA, Untranslated/genetics , S100 Proteins/genetics , Thyroid Neoplasms/genetics , Carcinogenesis/pathology , Carcinoma, Papillary/pathology , Cell Line, Tumor , Down-Regulation , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Genotype , Humans , Matrix Metalloproteinase 9/genetics , Neoplasm Invasiveness/pathology , S100 Calcium-Binding Protein A4 , Thyroid Gland/pathology , Thyroid Neoplasms/pathology , Vascular Endothelial Growth Factor A/genetics
10.
J Biomol Screen ; 20(9): 1150-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26109609

ABSTRACT

Recent progress of genetic studies has dramatically unveiled pathogenesis of acute myeloid leukemia (AML). However, overall survival of AML still remains unsatisfactory, and development of novel therapeutics is required. CCAAT/enhancer binding protein α (C/EBPα) is one of the crucial transcription factors that induce granulocytic differentiation, and its activity is perturbed in human myeloid leukemias. As its reexpression can induce differentiation and subsequent apoptosis of leukemic cells in vitro, we hypothesized that chemical compounds that restore C/EBPα expression and/or activity would lead to myeloid differentiation of leukemic cells. Using a cell-based high-throughput screening, we identified 2-[(E)-2-(3,4-dihydroxyphenyl)vinyl]-3-(2-methoxyphenyl)-4(3H)-quinazolinone as a potent inducer of C/EBPα and myeloid differentiation. Leukemia cell lines and primary blast cells isolated from human patients with AML treated with ICCB280 demonstrated evidence of morphological and functional differentiation, as well as massive apoptosis. We performed conformational analyses of the high-throughput screening hit compounds to postulate the spatial requirements for high potency. Our results warrant a development of novel differentiation therapies and significantly affect care of patients with AML with unfavorable prognosis in the near future.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Differentiation/drug effects , Leukemia, Myeloid, Acute/drug therapy , Apoptosis , Base Sequence , CCAAT-Enhancer-Binding Proteins/physiology , Drug Screening Assays, Antitumor , Gene Expression Regulation, Leukemic/drug effects , Genes, Reporter , HL-60 Cells , High-Throughput Screening Assays , Humans , Transcriptional Activation/drug effects , Tretinoin/pharmacology
11.
Haematologica ; 99(4): 697-705, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24162792

ABSTRACT

C/EPBα proteins, encoded by the CCAAT-enhancer-binding protein α gene, play a crucial role in granulocytic development, and defects in this transcription factor have been reported in acute myeloid leukemia. Here, we defined the C/EBPα signature characterized by a set of genes up-regulated upon C/EBPα activation. We analyzed expression of the C/EBPα signature in a cohort of 525 patients with acute myeloid leukemia and identified a subset characterized by low expression of this signature. We referred to this group of patients as the C/EBPα dysfunctional subset. Remarkably, a large percentage of samples harboring C/EBPα biallelic mutations clustered within this subset. We hypothesize that re-activation of the C/EBPα signature in the C/EBPα dysfunctional subset could have therapeutic potential. In search for small molecules able to reverse the low expression of the C/EBPα signature we applied the connectivity map. This analysis predicted positive connectivity between the C/EBPα activation signature and histone deacetylase inhibitors. We showed that these inhibitors reactivate expression of the C/EBPα signature and promote granulocytic differentiation of primary samples from the C/EBPα dysfunctional subset harboring biallelic C/EBPα mutations. Altogether, our study identifies histone deacetylase inhibitors as potential candidates for the treatment of certain leukemias characterized by down-regulation of the C/EBPα signature.


Subject(s)
Antineoplastic Agents/pharmacology , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Gene Expression Regulation, Leukemic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Transcriptome , CCAAT-Enhancer-Binding Protein-alpha/genetics , Cell Differentiation , Cell Line, Tumor , Cluster Analysis , Gene Expression Profiling , Humans , Mutation/drug effects , Mutation/genetics , Transcriptional Activation
12.
Blood ; 121(1): 159-69, 2013 Jan 03.
Article in English | MEDLINE | ID: mdl-23100311

ABSTRACT

Recently, we showed that increased miR-181a expression was associated with improved outcomes in cytogenetically normal acute myeloid leukemia (CN-AML). Interestingly, miR-181a expression was increased in CN-AML patients harboring CEBPA mutations, which are usually biallelic and associate with better prognosis. CEBPA encodes the C/EBPα transcription factor. We demonstrate here that the presence of N-terminal CEBPA mutations and miR-181a expression are linked. Indeed, the truncated C/EBPα-p30 isoform, which is produced from the N-terminal mutant CEBPA gene or from the differential translation of wild-type CEBPA mRNA and is commonly believed to have no transactivation activity, binds to the miR-181a-1 promoter and up-regulates the microRNA expression. Furthermore, we show that lenalidomide, a drug approved for myelodysplastic syndromes and multiple myeloma, enhances translation of the C/EBPα-p30 isoform, resulting in higher miR-181a levels. In xenograft mouse models, ectopic miR-181a expression inhibits tumor growth. Similarly, lenalidomide exhibits antitumorigenic activity paralleled by increased miR-181a expression. This regulatory pathway may explain an increased sensitivity to apoptosis-inducing chemotherapy in subsets of AML patients. Altogether, our data provide a potential explanation for the improved clinical outcomes observed in CEBPA-mutated CN-AML patients, and suggest that lenalidomide treatment enhancing the C/EBPα-p30 protein levels and in turn miR-181a may sensitize AML blasts to chemotherapy.


Subject(s)
CCAAT-Enhancer-Binding Proteins/physiology , Gene Expression Regulation, Leukemic/drug effects , Immunologic Factors/pharmacology , Leukemia, Myeloid, Acute/drug therapy , MicroRNAs/biosynthesis , Neoplasm Proteins/biosynthesis , RNA, Neoplasm/biosynthesis , Thalidomide/analogs & derivatives , Adult , Animals , Antimetabolites, Antineoplastic/pharmacology , CCAAT-Enhancer-Binding Proteins/biosynthesis , CCAAT-Enhancer-Binding Proteins/genetics , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Cell Line, Tumor/transplantation , Cytarabine/pharmacology , Frameshift Mutation , Humans , Immunologic Factors/therapeutic use , K562 Cells , Lenalidomide , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , MicroRNAs/genetics , Neoplasm Proteins/genetics , Point Mutation , Promoter Regions, Genetic/genetics , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Protein Isoforms/physiology , Protein Structure, Tertiary , RNA, Neoplasm/genetics , Recombinant Fusion Proteins/physiology , Thalidomide/pharmacology , Thalidomide/therapeutic use , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
13.
J Clin Invest ; 122(12): 4490-504, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23160200

ABSTRACT

C/EBPs are a family of transcription factors that regulate growth control and differentiation of various tissues. We found that C/EBPγ is highly upregulated in a subset of acute myeloid leukemia (AML) samples characterized by C/EBPα hypermethylation/silencing. Similarly, C/EBPγ was upregulated in murine hematopoietic stem/progenitor cells lacking C/EBPα, as C/EBPα mediates C/EBPγ suppression. Studies in myeloid cells demonstrated that CEBPG overexpression blocked neutrophilic differentiation. Further, downregulation of Cebpg in murine Cebpa-deficient stem/progenitor cells or in human CEBPA-silenced AML samples restored granulocytic differentiation. In addition, treatment of these leukemias with demethylating agents restored the C/EBPα-C/EBPγ balance and upregulated the expression of myeloid differentiation markers. Our results indicate that C/EBPγ mediates the myeloid differentiation arrest induced by C/EBPα deficiency and that targeting the C/EBPα-C/EBPγ axis rescues neutrophilic differentiation in this unique subset of AMLs.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Cell Differentiation , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/metabolism , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , CCAAT-Enhancer-Binding Proteins/metabolism , Cells, Cultured , Chromatin Immunoprecipitation , DNA Methylation , DNA Modification Methylases/antagonists & inhibitors , Decitabine , Epigenesis, Genetic , Genes, Reporter , Granulocyte Colony-Stimulating Factor/physiology , Granulocytes , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Luciferases, Renilla/biosynthesis , Luciferases, Renilla/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/metabolism , Neutrophils/metabolism , Neutrophils/physiology , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Protein Binding , Stem Cells/metabolism , Stem Cells/physiology , Up-Regulation
14.
J Clin Invest ; 122(8): 2955-66, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22797303

ABSTRACT

Mutations that activate the fms-like tyrosine kinase 3 (FLT3) receptor are among the most prevalent mutations in acute myeloid leukemias. The oncogenic role of FLT3 mutants has been attributed to the abnormal activation of several downstream signaling pathways, such as STAT3, STAT5, ERK1/2, and AKT. Here, we discovered that the cyclin-dependent kinase 1 (CDK1) pathway is also affected by internal tandem duplication mutations in FLT3. Moreover, we also identified C/EBPα, a granulopoiesis-promoting transcription factor, as a substrate for CDK1. We further demonstrated that CDK1 phosphorylates C/EBPα on serine 21, which inhibits its differentiation-inducing function. Importantly, we found that inhibition of CDK1 activity relieves the differentiation block in cell lines with mutated FLT3 as well as in primary patient-derived peripheral blood samples. Clinical trials with CDK1 inhibitors are currently under way for various malignancies. Our data strongly suggest that targeting the CDK1 pathway might be applied in the treatment of FLT3ITD mutant leukemias, especially those resistant to FLT3 inhibitor therapies.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha/metabolism , CDC2 Protein Kinase/antagonists & inhibitors , Leukemia, Myeloid, Acute/metabolism , fms-Like Tyrosine Kinase 3/metabolism , Animals , Base Sequence , CCAAT-Enhancer-Binding Protein-alpha/chemistry , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Granulocytes/drug effects , Granulocytes/pathology , HEK293 Cells , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Models, Biological , Mutation , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/genetics , Serine/chemistry , Signal Transduction , Transfection , fms-Like Tyrosine Kinase 3/genetics
15.
Proc Natl Acad Sci U S A ; 109(22): 8646-51, 2012 May 29.
Article in English | MEDLINE | ID: mdl-22586128

ABSTRACT

A genome-wide association study of papillary thyroid carcinoma (PTC) pinpointed two independent SNPs (rs944289 and rs965513) located in regions containing no annotated genes (14q13.3 and 9q22.33, respectively). Here, we describe a unique, long, intergenic, noncoding RNA gene (lincRNA) named Papillary Thyroid Carcinoma Susceptibility Candidate 3 (PTCSC3) located 3.2 kb downstream of rs944289 at 14q.13.3 and the expression of which is strictly thyroid specific. By quantitative PCR, PTCSC3 expression was strongly down-regulated (P = 2.84 × 10(-14)) in thyroid tumor tissue of 46 PTC patients and the risk allele (T) was associated with the strongest suppression (genotype [TT] (n = 21) vs. [CT] (n = 19), P = 0.004). In adjacent unaffected thyroid tissue, the genotype [TT] was associated with up-regulation of PTCSC3 ([TT] (n = 21) vs. [CT] (n = 19), P = 0.034). The SNP rs944289 was located in a binding site for the CCAAT/enhancer binding proteins (C/EBP) α and ß. The risk allele destroyed the binding site in silico. Both C/EBPα and C/EBPß activated the PTCSC3 promoter in reporter assays (P = 0.0009 and P = 0.0014, respectively) and the risk allele reduced the activation compared with the nonrisk allele (C) (P = 0.026 and P = 0.048, respectively). Restoration of PTCSC3 expression in PTC cell line cells (TPC-1 and BCPAP) inhibited cell growth (P = 0.002 and P = 0.019, respectively) and affected the expression of genes involved in DNA replication, recombination and repair, cellular movement, tumor morphology, and cell death. Our data suggest that SNP rs944289 predisposes to PTC through a previously uncharacterized, long intergenic noncoding RNA gene (PTCSC3) that has the characteristics of a tumor suppressor.


Subject(s)
Carcinoma, Papillary/genetics , Polymorphism, Single Nucleotide , RNA, Untranslated/genetics , Thyroid Neoplasms/genetics , Animals , Binding Sites/genetics , Blotting, Northern , CCAAT-Enhancer-Binding Protein-beta/metabolism , COS Cells , Carcinoma, Papillary/pathology , Cell Line, Tumor , Cell Proliferation , Chlorocebus aethiops , Chromosomes, Human, Pair 14/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Genetic Predisposition to Disease/genetics , Genotype , HEK293 Cells , Humans , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Thyroid Neoplasms/pathology
16.
Blood ; 119(21): 4878-88, 2012 May 24.
Article in English | MEDLINE | ID: mdl-22474248

ABSTRACT

The transcription factor C/EBPα is a critical mediator of myeloid differentiation and is often functionally impaired in acute myeloid leukemia. Recent studies have suggested that oncogenic FLT3 activity disrupts wild-type C/EBPα function via phosphorylation on serine 21 (S21). Despite the apparent role of pS21 as a negative regulator of C/EBPα transcription activity, the mechanism by which phosphorylation tips the balance between transcriptionally competent and inhibited forms remains unresolved. In the present study, we used immuno-affinity purification combined with quantitative mass spectrometry to delineate the proteins associated with C/EBPα on chromatin. We identified DEK, a protein with genetic links to leukemia, as a member of the C/EBPα complexes, and demonstrate that this association is disrupted by S21 phosphorylation. We confirmed that DEK is recruited specifically to chromatin with C/EBPα to enhance GCSFR3 promoter activation. In addition, we demonstrated that genetic depletion of DEK reduces the ability of C/EBPα to drive the expression of granulocytic target genes in vitro and disrupts G-CSF-mediated granulocytic differentiation of fresh human BM-derived CD34(+) cells. Our data suggest that C/EBPα and DEK coordinately activate myeloid gene expression and that S21 phosphorylation on wild-type C/EBPα mediates protein interactions that regulate the differentiation capacity of hematopoietic progenitors.


Subject(s)
CCAAT-Enhancer-Binding Proteins/physiology , Cell Differentiation/genetics , Chromosomal Proteins, Non-Histone/physiology , Myeloid Cells/physiology , Oncogene Proteins/physiology , Antibodies/pharmacology , CCAAT-Enhancer-Binding Proteins/antagonists & inhibitors , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , HEK293 Cells , Hematopoiesis/drug effects , Hematopoiesis/genetics , Humans , K562 Cells , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Phosphorylation/drug effects , Phosphorylation/physiology , Poly-ADP-Ribose Binding Proteins , Protein Binding/drug effects , Protein Kinase Inhibitors/pharmacology
17.
EMBO J ; 30(19): 4059-70, 2011 Aug 26.
Article in English | MEDLINE | ID: mdl-21873977

ABSTRACT

The transcription factor RUNX1 is essential to establish the haematopoietic gene expression programme; however, the mechanism of how it activates transcription of haematopoietic stem cell (HSC) genes is still elusive. Here, we obtained novel insights into RUNX1 function by studying regulation of the human CD34 gene, which is expressed in HSCs. Using transgenic mice carrying human CD34 PAC constructs, we identified a novel downstream regulatory element (DRE), which is bound by RUNX1 and is necessary for human CD34 expression in long-term (LT)-HSCs. Conditional deletion of Runx1 in mice harbouring human CD34 promoter-DRE constructs abrogates human CD34 expression. We demonstrate by chromosome conformation capture assays in LT-HSCs that the DRE physically interacts with the human CD34 promoter. Targeted mutagenesis of RUNX binding sites leads to perturbation of this interaction and decreased human CD34 expression in LT-HSCs. Overall, our in vivo data provide novel evidence about the role of RUNX1 in mediating interactions between distal and proximal elements of the HSC gene CD34.


Subject(s)
Antigens, CD34/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , Animals , Bone Marrow Transplantation , Chromatin/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Fetal Blood/cytology , Genotype , HL-60 Cells , Humans , Mice , Mice, Transgenic , Models, Biological , Regulatory Sequences, Nucleic Acid/genetics
18.
Exp Hematol ; 35(2): 230-9, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17258072

ABSTRACT

OBJECTIVE: We investigated the role of CCAAT enhancer-binding protein-alpha (C/EBPalpha) during zebrafish embryonic blood development. METHODS: Whole-mount mRNA in situ hybridization was performed to determine the spatio-temporal expression pattern of zebrafish cebpa in developing hematopoietic progenitors. A deletion mutation of cebpa (zD420), which mimics the human dominant-negative mutations of C/EBPalpha, was transfected into CV1 cell line to evaluate its transcriptional activity in vitro and injected into zebrafish embryos at the one- to two-cell stage to examine its effects on primitive hematopoiesis during early zebrafish development. RESULTS: Zebrafish cebpa is expressed in the anterior and posterior lateral plate mesoderm at 12 hours postfertilization, along with scl, pu.1, and gata1 in developing hematopoietic progenitors. In vitro, the deletion mutation of cebpa (zD420) prevents expression of the full-length protein, allowing the expression of truncated isoforms from internal translational initiation sites. As in the human, the truncated zebrafish C/EBPalpha proteins did not activate the expression of known target granulocytic genes, and in fact suppressed transactivation that was induced in vitro by the full-length protein. Forced expression of the zD420 mRNA in zebrafish embryos led to an expansion of primitive erythropoiesis, without a discernible effect on granulopoiesis. CONCLUSION: Expression of the truncated isoforms of cebpa alters the developmental pattern of hematopoietic progenitor cells during embryogenesis.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha/genetics , Erythropoiesis/genetics , Gene Expression Regulation, Developmental , Genes, Dominant , Zebrafish/genetics , Animals , Base Sequence , Blood Vessels/embryology , Blood Vessels/metabolism , CCAAT-Enhancer-Binding Protein-alpha/metabolism , DNA, Complementary/genetics , Embryonic Development/genetics , Embryonic Development/physiology , Gene Deletion , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Molecular Sequence Data , Mutation , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/physiology , RNA, Messenger , Reverse Transcriptase Polymerase Chain Reaction/methods , Sequence Analysis, DNA , Translocation, Genetic/genetics , Translocation, Genetic/physiology , Transplantation, Heterologous , Zebrafish/embryology , Zebrafish/metabolism
19.
Int J Oncol ; 30(2): 349-55, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17203216

ABSTRACT

Standard chemotherapy is not curative for many patients with acute myeloid leukemia (AML). New treatment strategies combining demethylating agents, such as decitabine, and drugs that induce myelomonocytic differentiation (i.e. Vitamin D3) may re-establish functional hematopoiesis in these patients. We studied the effects of decitabine alone or in combination with Vitamin D3 (VD3) on U937 cells and AML blasts. Preincubation with decitabine (0.1-1 microM) and subsequent exposure to VD3 (3 nM) synergistically induced monocytic differentiation. To elucidate the mechanisms of decitabine- and VD3-induced monocytic differentiation, we investigated the effects of the two drugs on transcription factors implicated in monocytic differentiation. Northern and Western blotting showed that decitabine induced transcription of c-jun but not PU.1, while VD3 increased PU.1, IRF8, and C/EBPbeta but not c-jun. Using electromobility shift assays, we demonstrated increased DNA binding of nuclear proteins from decitabine- and VD3-induced U937 cells to the CD11b promoter. In addition, we investigated whether the myeloid transcription factor Sp1 played a role in decitabine- and VD3-induced CD14 expression. Indeed, we found that mithramycin A, a specific inhibitor of Sp1, inhibited both VD3- and decitabine-induced upregulation of CD14, which is in line with previous data showing that Sp1 is critical for CD14 promoter activity. Induction of CD11b and/or CD14 by decitabine and/or VD3 was confirmed in primary AML patient samples at the time of diagnosis. In conclusion, decitabine synergizes with Vitamin D3 to induce CD11b and CD14 expression, likely by enhancing PU.1/c-jun and Sp1 transcriptional activity.


Subject(s)
Azacitidine/analogs & derivatives , Cholecalciferol/biosynthesis , Monocytes/cytology , Transcription, Genetic , Azacitidine/pharmacokinetics , CD11b Antigen/biosynthesis , Cell Differentiation , Decitabine , Humans , Lipopolysaccharide Receptors/biosynthesis , Models, Biological , Monocytes/metabolism , Plicamycin/pharmacology , Sp1 Transcription Factor/metabolism , Transcription Factors/metabolism , U937 Cells
20.
BMC Cancer ; 6: 158, 2006 Jun 14.
Article in English | MEDLINE | ID: mdl-16774685

ABSTRACT

BACKGROUND: C/EBPalpha is a transcription factor essential for terminal differentiation of several cell types. It has not known if C/EBPalpha protein is expressed and functions in the prostate gland. METHODS: The presence of C/EBPalpha in normal and cancerous prostate epithelium was examined by immunochemistry. Over expression of C/EBPalpha in LNCaP cells was conducted with retrovirus-mediated transduction. PSA expression was examined by RT-PCR and western blot and PSA promoter activity by luciferase reporter assay. RESULTS: In normal prostate C/EBPalpha was expressed in the basal layer of the epithelium. In prostate cancer C/EBPalpha was detected at low levels throughout the cancers and in advanced prostate cancer C/EBPalpha expression was associated with decreased expression of AR and PSA. Overexpression of C/EBPalpha inhibited epigenetically PSA expression and was accompanied by the loss of expression of AR. Transient increase of C/EBPalpha inhibited the PSA promoter/enhancer activity independently of expression of AR. CONCLUSION: In LNCaP cells C/EBPalpha over expression inhibits expression of PSA by AR -dependent and independent mechanisms and by extinguishing AR expression provides a model for hormonal independent cell growth.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha/metabolism , Down-Regulation/genetics , Gene Expression Regulation, Neoplastic , Promoter Regions, Genetic/genetics , Prostate-Specific Antigen/genetics , Receptors, Androgen/metabolism , Animals , Cell Line, Tumor , Humans , Immunohistochemistry , Male , Prostate/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rats , Transcription, Genetic/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...