Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Cells ; 9(11)2020 10 29.
Article in English | MEDLINE | ID: mdl-33138093

ABSTRACT

NF-Y is a transcription factor (TF) comprising three subunits (NF-YA, NF-YB, NF-YC) that binds with high specificity to the CCAAT sequence, a widespread regulatory element in gene promoters of prosurvival, cell-cycle-promoting, and metabolic genes. Tumor cells undergo "metabolic rewiring" through overexpression of genes involved in such pathways, many of which are under NF-Y control. In addition, NF-YA appears to be overexpressed in many tumor types. Thus, limiting NF-Y activity may represent a desirable anti-cancer strategy, which is an ongoing field of research. With virtual-screening docking simulations on a library of pharmacologically active compounds, we identified suramin as a potential NF-Y inhibitor. We focused on suramin given its high water-solubility that is an important factor for in vitro testing, since NF-Y is sensitive to DMSO. By electrophoretic mobility shift assays (EMSA), isothermal titration calorimetry (ITC), STD NMR, X-ray crystallography, and molecular dynamics (MD) simulations, we showed that suramin binds to the histone fold domains (HFDs) of NF-Y, preventing DNA-binding. Our analyses, provide atomic-level detail on the interaction between suramin and NF-Y and reveal a region of the protein, nearby the suramin-binding site and poorly conserved in other HFD-containing TFs, that may represent a promising starting point for rational design of more specific and potent inhibitors with potential therapeutic applications.


Subject(s)
CCAAT-Binding Factor/antagonists & inhibitors , CCAAT-Binding Factor/chemistry , Suramin/chemistry , Suramin/pharmacology , Transcription Factors/antagonists & inhibitors , Transcription Factors/chemistry , Amino Acid Sequence , Biophysical Phenomena , DNA/metabolism , Humans , Magnetic Resonance Spectroscopy , Molecular Dynamics Simulation , Protein Multimerization , Structure-Activity Relationship
2.
Oncogene ; 35(11): 1468-74, 2016 Mar 17.
Article in English | MEDLINE | ID: mdl-26073082

ABSTRACT

Neurofibromatosis type 1 (NF1) patients are predisposed to neurofibromas but the driver(s) that contribute to neurofibroma formation are not fully understood. By cross comparison of microarray gene lists on human neurofibroma-initiating cells and developed neurofibroma Schwann cells (SCs) we identified RUNX1 overexpression in human neurofibroma initiation cells, suggesting RUNX1 might relate to neurofibroma formation. Immunostaining confirmed RUNX1 protein overexpression in human plexiform neurofibromas. Runx1 overexpression was confirmed in mouse Schwann cell progenitors (SCPs) and mouse neurofibromas at the messenger RNA and protein levels. Genetic inhibition of Runx1 expression by small hairpin RNA or pharmacological inhibition of Runx1 function by a Runx1/Cbfß interaction inhibitor, Ro5-3335, decreased mouse neurofibroma sphere number in vitro. Targeted genetic deletion of Runx1 in SCs and SCPs delayed mouse neurofibroma formation in vivo. Mechanistically, loss of Nf1 increased embryonic day 12.5 Runx1(+)/Blbp(+) progenitors that enable tumor formation. These results suggest that Runx1 has an important role in Nf1 neurofibroma initiation, and inhibition of RUNX1 function might provide a novel potential therapeutic treatment strategy for neurofibroma patients.


Subject(s)
Cell Transformation, Neoplastic/pathology , Core Binding Factor Alpha 2 Subunit/genetics , Neurofibroma/pathology , Neurofibromatosis 1/pathology , Neurofibromin 1/genetics , Animals , CCAAT-Binding Factor/antagonists & inhibitors , Carrier Proteins/metabolism , Cell Line, Tumor , Core Binding Factor Alpha 2 Subunit/biosynthesis , Core Binding Factor Alpha 2 Subunit/metabolism , Fatty Acid-Binding Protein 7 , Humans , Mice , Neurofibroma, Plexiform/pathology , RNA Interference , RNA, Small Interfering/genetics , Schwann Cells/cytology , Stem Cells/cytology , Tumor Suppressor Proteins/metabolism
3.
Exp Dermatol ; 23 Suppl 1: 2-6, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25234828

ABSTRACT

Transforming growth factor-ß (TGF-ß) is a major regulator of collagen gene expression in human skin fibroblasts. Cellular responses to TGF-ß are mediated primarily through its cell surface type I (TßRI) and type II (TßRII) receptors. Ultraviolet (UV) irradiation impairs TGF-ß signalling largely due to reduced TßRII gene expression, thereby decreasing type I procollagen synthesis, in human skin fibroblasts. UV irradiation does not alter either TßRII mRNA or protein stability, indicating that UV reduction in TßRII expression likely results from transcriptional or translational repression. To understand how UV irradiation regulates TßRII transcription, we used a series of TßRII promoter-luciferase 5'-deletion constructs (covering 2 kb of the TßRII proximal promoter) to determine transcriptional rate in response to UV irradiation. We identified a 137-bp region upstream of the transcriptional start site that exhibited high promoter activity and was repressed 60% by UV irradiation, whereas all other TßRII promoter reporter constructs exhibited either low promoter activities or no regulation by UV irradiation. Mutation of potential transcription factor binding sites within the promoter region revealed that an inverted CCAAT box (-81 bp from transcription start site) is required for promoter activity. Mutation of the CCAAT box completely abolished UV irradiation regulation of the TßRII promoter. Protein-binding assay, as determined by electrophoretic mobility-shift assays (EMSAs) using the inverted CCAAT box as probe (-100/-62), demonstrated significantly enhanced protein binding in response to UV irradiation. Super shift experiments indicated that nuclear factor Y (NFY) is able to binding to this sequence, but NFY binding was not altered in response to UV irradiation, indicating additional protein(s) are capable of binding this sequence in response to UV irradiation. Taken together, these data indicate that UV irradiation reduces TßRII expression, at least partially, through transcriptional repression. This repression is mediated by a 38-bp sequence in TßRII promoter, in human skin fibroblasts.


Subject(s)
Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , Skin/metabolism , Skin/radiation effects , Ultraviolet Rays/adverse effects , Base Sequence , Binding Sites/genetics , CCAAT-Binding Factor/antagonists & inhibitors , CCAAT-Binding Factor/genetics , CCAAT-Binding Factor/metabolism , Cells, Cultured , Down-Regulation/radiation effects , Fibroblasts/metabolism , Fibroblasts/radiation effects , Humans , Models, Genetic , Mutagenesis, Site-Directed , Promoter Regions, Genetic/radiation effects , Protein Binding/radiation effects , RNA, Small Interfering/genetics , Receptor, Transforming Growth Factor-beta Type II
4.
Arterioscler Thromb Vasc Biol ; 33(5): 1036-45, 2013 May.
Article in English | MEDLINE | ID: mdl-23430616

ABSTRACT

OBJECTIVE: Atherosclerosis and restenosis are multifactorial diseases associated with abnormal vascular smooth muscle cell (VSMC) proliferation. Nuclear factor-Y (NF-Y) plays a major role in transcriptional activation of the CYCLIN B1 gene (CCNB1), a key positive regulator of cell proliferation and neointimal thickening. Here, we investigated the role of NF-Y in occlusive vascular disease. APPROACH AND RESULTS: We performed molecular and expression studies in cultured cells, animal models, and human tissues. We find upregulation of NF-Y and cyclin B1 expression in proliferative regions of murine atherosclerotic plaques and mechanically induced lesions, which correlates with higher binding of NF-Y to target sequences in the CCNB1 promoter. NF-YA expression in neointimal lesions is detected in VSMCs, macrophages, and endothelial cells. Platelet-derived growth factor-BB, a main inductor of VSMC growth and neointima development, induces the recruitment of NF-Y to the CCNB1 promoter and augments both CCNB1 mRNA expression and cell proliferation through extracellular signal-regulated kinase 1/2 and Akt activation in rat and human VSMCs. Moreover, adenovirus-mediated overexpression of a NF-YA-dominant negative mutant inhibits platelet-derived growth factor-BB-induced CCNB1 expression and VSMC proliferation in vitro and neointimal lesion formation in a mouse model of femoral artery injury. We also detect NF-Y expression and DNA-binding activity in human neointimal lesions. CONCLUSIONS: Our results identify NF-Y as a key downstream effector of the platelet-derived growth factor-BB-dependent mitogenic pathway that is activated in experimental and human vasculoproliferative diseases. They also identify NF-Y inhibition as a novel and attractive strategy for the local treatment of neointimal formation induced by vessel denudation.


Subject(s)
CCAAT-Binding Factor/physiology , Muscle, Smooth, Vascular/cytology , Neointima/etiology , Animals , Apolipoproteins E/physiology , Atherosclerosis/etiology , Becaplermin , CCAAT-Binding Factor/antagonists & inhibitors , Cell Proliferation , Cells, Cultured , Cyclin B1/genetics , Endothelial Cells/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Neointima/therapy , Proto-Oncogene Proteins c-sis/pharmacology , Rats , Rats, Wistar
5.
Eukaryot Cell ; 11(6): 806-19, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22523368

ABSTRACT

When iron is scarce, Schizosaccharomyces pombe cells repress transcription of several genes that encode iron-using proteins. Php4 mediates this transcriptional control by specifically interacting with the CCAAT-binding core complex that is composed of Php2, Php3, and Php5. In contrast, when there is sufficient iron, Php4 is inactivated, thus allowing the transcription of many genes that encode iron-requiring proteins. Analysis by bimolecular fluorescence complementation and two-hybrid assays showed that Php4 and the monothiol glutaredoxin Grx4 physically interact with each other. Deletion mapping analysis revealed that the glutaredoxin (GRX) domain of Grx4 associates with Php4 in an iron-dependent manner. Site-directed mutagenesis identified the Cys172 of Grx4 as being required for this iron-dependent association. Subsequent analysis showed that, although the thioredoxin (TRX) domain of Grx4 interacts strongly with Php4, this interaction is insensitive to iron. Fine mapping analysis revealed that the Cys35 of Grx4 is necessary for the association between the TRX domain and Php4. Taken together, the results revealed that whereas the TRX domain interacts constitutively with Php4, the GRX domain-Php4 association is both modulated by iron and required for the inhibition of Php4 activity in response to iron repletion.


Subject(s)
CCAAT-Binding Factor/antagonists & inhibitors , Glutaredoxins/metabolism , Iron/pharmacology , Schizosaccharomyces pombe Proteins/antagonists & inhibitors , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/drug effects , Schizosaccharomyces/metabolism , Sulfhydryl Compounds/metabolism , CCAAT-Binding Factor/metabolism , Cysteine/metabolism , Glutaredoxins/chemistry , Protein Binding/drug effects , Protein Interaction Mapping , Protein Structure, Tertiary , Schizosaccharomyces/cytology , Schizosaccharomyces pombe Proteins/chemistry , Structure-Activity Relationship
6.
Nucleic Acids Res ; 39(13): 5356-68, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21415014

ABSTRACT

Regulated gene expression is essential for a proper progression through the cell cycle. The transcription factor NF-Y has a fundamental function in transcriptional regulation of cell cycle genes, particularly of G2/M genes. In order to investigate common and distinct functions of NF-Y subunits in cell cycle regulation, NF-YA, NF-YB and NF-YC have been silenced by shRNAs in HCT116 cells. NF-YA loss led to a delay in S-phase progression, DNA damage and apoptosis: we showed the activation of the replication checkpoint, through the recruitment of Δp53 and of the replication proteins PCNA and Mcm7 to chromatin. Differently, NF-YB depletion impaired cells from exiting G2/M, but did not interfere with S-phase progression. Gene expression analysis of NF-YA and NF-YB inactivated cells highlighted a common set of hit genes, as well as a plethora of uncommon genes, unveiling a different effect of NF-Y subunits loss on NF-Y binding to its target genes. Chromatin extracts and ChIP analysis showed that NF-YA depletion was more effective than NF-YB in hitting NF-Y recruitment to CCAAT-promoters. Our data suggest a critical role of NF-Y expression, highlighting that the lack of the single subunits are differently perceived by the cells, which activate diverse cell cycle blocks and signaling pathways.


Subject(s)
CCAAT-Binding Factor/antagonists & inhibitors , Cell Proliferation , Cell Cycle , Cell Line, Tumor , DNA Damage , DNA Replication , Gene Expression , Gene Silencing , Humans , Protein Subunits/antagonists & inhibitors , S Phase , Tumor Suppressor Protein p53/metabolism
7.
Sci Rep ; 1: 136, 2011.
Article in English | MEDLINE | ID: mdl-22355653

ABSTRACT

We previously demonstrated that CBF activity is needed for cell proliferation and early embryonic development. To examine the in vivo function of CBF in differentiated hepatocytes, we conditionally deleted CBF-B in hepatocytes after birth. Deletion of CBF-B resulted in progressive liver injury and severe hepatocellular degeneration 4 weeks after birth. Electron microscopic examination demonstrated pleiotropic changes of hepatocytes including enlarged cell and nuclear size, intracellular lipid deposition, disorganized endoplasmic reticulum, and mitochondrial abnormalities. Gene expression analyses showed that deletion of CBF-B activated expression of specific endoplasmic reticulum (ER) stress-regulated genes. Inactivation of CBF-B also inhibited expression of C/EBP alpha, an important transcription factor controlling various metabolic processes in adult hepatocytes. Altogether, our study reveals for the first time that CBF is a key transcription factor controlling ER function and metabolic processes in mature hepatocytes.


Subject(s)
CCAAT-Binding Factor/antagonists & inhibitors , Liver/metabolism , Liver/pathology , Animals , Animals, Newborn , CCAAT-Binding Factor/deficiency , CCAAT-Binding Factor/genetics , Endoplasmic Reticulum Stress , Fatty Liver/genetics , Fatty Liver/metabolism , Fatty Liver/pathology , Gene Expression , Hepatocytes/metabolism , Hepatocytes/pathology , Hepatomegaly/genetics , Hepatomegaly/metabolism , Hepatomegaly/pathology , Lipid Metabolism , Liver/injuries , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Electron, Transmission
8.
Biochem J ; 429(2): 347-57, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20450493

ABSTRACT

FDPS (farnesyl diphosphate synthase) catalyses the formation of farnesyl diphosphate, a key intermediate in the synthesis of cholesterol and isoprenylated cellular metabolites. FDPS is also the molecular target of nitrogen-containing bisphosphonates, which are used as bone-antiresorptive drugs in various disorders. In the present study, we characterized the sterol-response element and NF-Y (nuclear factor Y)-binding site in the human FDPS promoter. Using a luciferase assay, electrophoretic mobility-shift assay and chromatin immunoprecipitation assay, we demonstrated that these elements are responsible for the transcription of the FDPS gene, and that its transcriptional activation is mediated by SREBP-2 (sterol-regulatory-element-binding protein 2) and NF-Y. We also investigated whether sterol-mediated FDPS expression is involved in the cell proliferation induced by zoledronic acid, an FDPS inhibitor. We show that the SREBP-2- and NF-Y-mediated regulation of FDPS gene transcription modulates cell proliferation. These results suggest that SREBP-2 and NF-Y are required to trigger cell proliferation through the induction of FDPS expression and that the pharmacological action of zoledronic acid is involved in this pathway.


Subject(s)
CCAAT-Binding Factor/metabolism , Geranyltranstransferase/genetics , Hepatoblastoma/genetics , Hepatoblastoma/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Sterol Regulatory Element Binding Protein 2/metabolism , Base Sequence , Binding Sites/genetics , Bone Density Conservation Agents/pharmacology , CCAAT-Binding Factor/antagonists & inhibitors , CCAAT-Binding Factor/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Conserved Sequence , DNA Primers/genetics , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Diphosphonates/pharmacology , Gene Knockdown Techniques , Geranyltranstransferase/biosynthesis , Hepatoblastoma/pathology , Humans , Imidazoles/pharmacology , Liver Neoplasms/pathology , Molecular Sequence Data , Mutagenesis, Site-Directed , Promoter Regions, Genetic , RNA, Small Interfering/genetics , Sequence Homology, Nucleic Acid , Sterol Regulatory Element Binding Protein 2/antagonists & inhibitors , Sterol Regulatory Element Binding Protein 2/genetics , Sterols/metabolism , Transcriptional Activation , Transfection , Zoledronic Acid
9.
Gene ; 459(1-2): 11-23, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20338228

ABSTRACT

Dihydrodiol dehydrogenases are a family of aldo-keto reductases (AKR1Cs) involved in the metabolism of steroid hormones and xenobiotics. Herein, we have cloned and characterized the proximal promoter region of the human AKR1C1 gene. The 5' flanking proximal promoter region of the AKR1C1 gene consists of a TATA box and an inverted CCAAT binding site. Deletion analysis of the 5' flanking, approximately 3.0 kb region of the human AKR1C1 gene identified the region between -128 and -88 as the minimal proximal promoter essential for basal transcription of AKR1C1 in human ovarian (2008 and 2008/C13*), lung (H23 and A549) and liver carcinoma (HepG2) cells. Site-directed mutagenesis studies indicated that the transcription factor binding sites for NF-Y/CEBP were involved in controlling the basal transcription of AKR1C1 in all the cancer cells studied. Electrophoretic mobility shift (EMSAs) and gel-supershift assays demonstrated that the transcription factor NF-Y preferentially binds to the inverted CCAAT box at (-109)ATTGG(-105) of the AKR1C1 gene. Chromatin immunoprecipitation (ChIP) analysis confirmed the in vivo association between NF-Y and human AKR1C1 gene promoter in human ovarian, lung and liver carcinoma cells. Ectopic expression of NF-Ys increased the AKR1C1 gene transcription, whereas expression of a dominant-negative NF-YA or suppression of NF-YA decreased the AKR1C1 gene transcription. A 2-fold increase in AKR1C1 transcription was observed specifically in cisplatin-treated 2008 cells that were CCAAT box-dependent. These results indicate that the NF-Y regulates the basal transcription of AKR1C1 in human ovarian, lung and liver carcinoma cells and the cisplatin-induced transcription in human ovarian carcinoma cells.


Subject(s)
20-Hydroxysteroid Dehydrogenases/genetics , CCAAT-Binding Factor/metabolism , Gene Expression Regulation, Neoplastic , Promoter Regions, Genetic/genetics , Transcription, Genetic , 20-Hydroxysteroid Dehydrogenases/metabolism , Antineoplastic Agents/pharmacology , Binding Sites , CCAAT-Binding Factor/antagonists & inhibitors , CCAAT-Binding Factor/genetics , Chromatin Immunoprecipitation , Cisplatin/pharmacology , Electrophoretic Mobility Shift Assay , Female , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mutagenesis, Site-Directed , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , RNA, Small Interfering/pharmacology , Regulatory Sequences, Nucleic Acid/genetics , Sequence Deletion , Tumor Cells, Cultured
10.
Mol Cancer Ther ; 7(5): 1319-28, 2008 May.
Article in English | MEDLINE | ID: mdl-18483319

ABSTRACT

Many genes involved in cell cycle control have promoters that bind the heterotrimeric transcription factor NF-Y. Several minor-groove binding drugs have been shown to block interactions of transcription factors with cognate DNA-binding sequences. We showed previously that noncovalent minor-groove binding agents block interactions of NF-Y with the promoter of topoisomerase IIalpha (topo IIalpha). In this study, we investigated the ability of GWL-78, a pyrrolobenzodiazepine-poly(N-methylpyrrole) conjugate, to inhibit the binding of NF-Y to DNA. Electrophoretic mobility shift assays showed that GWL-78 could displace NF-Y bound to several CCAAT motifs within promoters of genes involved in cell cycle progression. DNase I footprinting of the topo IIalpha promoter confirmed binding of GWL-78 to AT-rich sequences corresponding to the preferred binding site of NF-Y. Incubation with GWL-78 resulted in displacement of NF-Y binding to DNA. Chromatin immunoprecipitation assays on the topo IIalpha promoter showed that GWL-78 was able to enter the nucleus and interact with specific DNA sequences. Treatment of NIH3T3 cells with GWL-78 resulted in a block of cell cycle progression, which did not involve activation of p53. Thus, agents such as GWL-78 may be useful in modulating transcription and blocking cellular proliferation.


Subject(s)
Antineoplastic Agents/pharmacology , Benzodiazepines/pharmacology , CCAAT-Binding Factor/antagonists & inhibitors , CCAAT-Binding Factor/metabolism , DNA/metabolism , Dipeptides/pharmacology , Amino Acid Motifs , Animals , Antigens, Neoplasm/drug effects , Antigens, Neoplasm/genetics , Base Sequence , Binding Sites , CCAAT-Binding Factor/genetics , Cell Cycle , DNA Topoisomerases, Type II/drug effects , DNA Topoisomerases, Type II/genetics , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/genetics , Electrophoretic Mobility Shift Assay , Mice , Molecular Sequence Data , NIH 3T3 Cells
11.
Genes Cells ; 13(2): 117-30, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18233955

ABSTRACT

The CCAAT motif-binding factor, nuclear factor Y (NF-Y) consists of three different subunits, NF-YA, NF-YB and NF-YC. Knockdown of Drosophila NF-YA (dNF-YA) in the notum compartment of wing discs by a pannir-GAL4 and UAS-dNF-YAIR mainly resulted in a thorax disclosed phenotype. Reduction of the Drosophila c-Jun N-terminal kinase (JNK) basket (bsk) gene dose enhanced the knockdown of dNF-YA-induced phenotype. Monitoring of JNK activity in the wing disc by LacZ expression in a puckered (puc)-LacZ enhancer trap line revealed reduction in the level of the JNK reporter, puc-LacZ signals, in dNF-YA RNAi clones. In addition, expression of wild-type Bsk effectively suppressed the phenotype induced by knockdown of dNF-YA. The bsk gene promoter contains a CCAAT motif and this motif plays a positive role in the promoter activity. We performed chromatin immunoprecipitation (ChIP) assays in S2 cells with anti-dNF-YA IgG and quantitative real-time PCR. The bsk gene promoter region containing the CCAAT boxes was effectively amplified in the immunoprecipitates by PCR. However, this region was not amplified in the immunoprecipitates from dNF-YA knockdown cells. Furthermore, the level of endogenous bsk mRNA is reduced in the dNF-YA knockdown larvae. These results suggest that dNF-Y is necessary for proper bsk expression and activity of JNK pathway during thorax development.


Subject(s)
CCAAT-Binding Factor/metabolism , Drosophila Proteins/metabolism , Drosophila/growth & development , Drosophila/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Animals , Animals, Genetically Modified , Base Sequence , CCAAT-Binding Factor/antagonists & inhibitors , CCAAT-Binding Factor/genetics , DNA Primers/genetics , Drosophila/genetics , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Genes, Insect , Plasmids/genetics , Promoter Regions, Genetic , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Thorax/growth & development , Wings, Animal/growth & development
12.
EMBO J ; 27(6): 827-39, 2008 Mar 19.
Article in English | MEDLINE | ID: mdl-18288205

ABSTRACT

In Huntington's disease (HD), mutant Huntingtin, which contains expanded polyglutamine stretches, forms nuclear aggregates in neurons. The interactions of several transcriptional factors with mutant Huntingtin, as well as altered expression of many genes in HD models, imply the involvement of transcriptional dysregulation in the HD pathological process. The precise mechanism remains obscure, however. Here, we show that mutant Huntingtin aggregates interact with the components of the NF-Y transcriptional factor in vitro and in HD model mouse brain. An electrophoretic mobility shift assay using HD model mouse brain lysates showed reduction in NF-Y binding to the promoter region of HSP70, one of the NF-Y targets. RT-PCR analysis revealed reduced HSP70 expression in these brains. We further clarified the importance of NF-Y for HSP70 transcription in cultured neurons. These data indicate that mutant Huntingtin sequesters NF-Y, leading to the reduction of HSP70 gene expression in HD model mice brain. Because suppressive roles of HSP70 on the HD pathological process have been shown in several HD models, NF-Y could be an important target of mutant Huntingtin.


Subject(s)
CCAAT-Binding Factor/metabolism , HSP70 Heat-Shock Proteins/antagonists & inhibitors , HSP70 Heat-Shock Proteins/genetics , Mutation , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Animals , CCAAT-Binding Factor/antagonists & inhibitors , Cell Line, Tumor , Disease Models, Animal , HSP70 Heat-Shock Proteins/biosynthesis , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/metabolism , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/physiology , Nuclear Proteins/physiology , Protein Binding/genetics
13.
Nucleic Acids Res ; 36(5): 1415-28, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18187512

ABSTRACT

The transcription factor NF-Y is a trimer with histone-like subunits that binds and activates CCAAT-containing promoters. NF-Y controls the expression of several key regulators of the cell cycle. In this study, we examined the functional and molecular effects of NF-YB knockdown. Cell cycle progression is affected with a G2/M-specific depletion. This is due to the inability of activation of G2/M-specific genes, as evidenced by expression profiling, RT-PCR and ChIP data. Surprisingly, apoptosis is also observed, with Caspase 3/7/8 cleavage. A role of p53 and Bcl-2 family members is important. NF-YB inactivation is sufficient to functionally activate p53, in the absence of DNA damage. Failure to maintain a physiologic level of CCAAT-dependent transcription of anti-apoptotic genes contributes to impairment of Bax/Bcl-2 and Bax/Bcl-X(L) ratios. Our data highlight the importance of fine balancing the NF-Y-p53 duo for cell survival by (i) maintaining transcription of anti-apoptotic genes and (ii) preventing p53 activation that triggers the apoptotic cascade.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Apoptosis/genetics , CCAAT-Binding Factor/metabolism , DNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , CCAAT-Binding Factor/antagonists & inhibitors , CCAAT-Binding Factor/genetics , Caspases/metabolism , Cell Cycle/genetics , Cell Line, Tumor , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Gene Expression Profiling , Humans , RNA Interference , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription, Genetic
14.
Biochim Biophys Acta ; 1780(2): 274-81, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18078822

ABSTRACT

An inverted CCAAT sequence is recognized by both transcription factors NF-Y and DNA/RNA binding protein YB-1. In the process of examining the effect of nuclear RNA on an inverted CCAAT-containing promoter of MDR1 gene, we found that U7 snRNA inhibits NF-Y and suppresses the promoter activity both in vitro and in NG108-15 tumor cells. Analysis using a designed RNA, which was structurally unrelated to U7 snRNA, revealed that RNA binding by YB-1 is not specific and that the protein is not involved in the transcription. Furthermore, we demonstrated that in the nucleus of doxorubicin-treated cells, DNA binding by NF-Y and transcriptional activity of the promoter were inhibited without either a decrease of NF-Y or an increase of the p53 tumor suppressor, which is known to inhibit DNA binding by NF-Y. In these cells, U7 snRNA was specifically increased and associated with NF-Y, and treatment with RNase A eliminated the inhibition of NF-Y activity. These results suggest that U7 snRNA has a novel function as a transcriptional regulator to control NF-Y.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , CCAAT-Binding Factor/antagonists & inhibitors , CCAAT-Binding Factor/metabolism , Gene Expression Regulation , RNA, Small Nuclear/metabolism , Animals , Base Sequence , Cell Line, Tumor , Mice , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Transcription, Genetic
15.
Bioorg Med Chem ; 16(4): 2093-102, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-17977733

ABSTRACT

The synthesis and DNA binding characteristics of a polyamide-intercalator conjugate, designed to inhibit NF-Y binding to the ICB-2 site of the topoisomerase IIalpha promoter and up-regulate the expression of the enzyme in confluent cells, are reported. Thermal denaturation and CD titration studies demonstrated binding to the cognate sequence (5'-AAGCTA-3'). Formation of ligand-induced CD bands at approximately 330 nm provided indication that the molecule interacts selectively in the minor groove of DNA. Intercalation was evidenced by a fivefold increase in emission of the intercalator moiety upon binding to the ICB-2 hairpin oligonucleotide. An increase in viscosity of a solution of calf-thymus DNA on addition of the conjugate provided further evidence. The binding affinity of the conjugate was ascertained using SPR (5.6x10(6) M(-1)), which according to a gel shift assay was capable of inhibiting the binding of NF-Y at a concentration of 50 microM. DNaseI footprinting, using the topoIIalpha promoter sequence, highlighted the specificity of the conjugate for the cognate site (5'-AAGCTA-3'). Finally, through Western blot analysis, confluent murine NIH 3T3 cells treated with conjugate were found to have enhanced expression of topoIIalpha. These results suggest that the conjugate can enter the nucleus, bind to its target site, presumably as a stacked dimer, and up-regulate the expression of topoIIalpha by blocking the binding of NF-Y.


Subject(s)
Antigens, Neoplasm/genetics , CCAAT-Binding Factor/antagonists & inhibitors , DNA Topoisomerases, Type II/genetics , DNA-Binding Proteins/genetics , Intercalating Agents/pharmacology , Transcription Factors/antagonists & inhibitors , 3T3 Cells , Animals , Base Sequence , Binding Sites , DNA , Dimerization , Intercalating Agents/chemistry , Mice , Nylons/chemistry , Nylons/pharmacology , Promoter Regions, Genetic , Protein Binding/drug effects , Up-Regulation/drug effects
16.
Nucleic Acids Res ; 34(21): 6272-85, 2006.
Article in English | MEDLINE | ID: mdl-17098936

ABSTRACT

Previous studies showed that binding of the CBF/NF-Y (CBF) transcription factor to cellular promoters is essential for cell proliferation. This observation prompted us to investigate the function of CBF in relation to cell cycle progression and in cell-cycle-regulated transcription. In this study, we used a tetracycline-inducible adenoviral vector to express a truncated CBF-B subunit, Bdbd, lacking a transcription activation domain in various mammalian cell lines. The Bdbd polypeptide interacts with cellular CBF-A/CBF-C and binds to promoters containing CBF-binding sites. Interestingly, expression of Bdbd in various mammalian cells resulted in the inhibition of cell proliferation and specific cell cycle arrest at G2/M phase. Gene expression analysis demonstrated that the expression of Bdbd strongly suppressed cell cycle-dependent transcription activation of Cyclin B1, Aurora A and CDK1 genes, key regulators for cell cycle progression at G2/M phase. Chromatin immunoprecipitation analysis showed that Bdbd significantly inhibited binding of TATA-binding protein, TBP to both Cyclin B1 and Aurora A promoters, but did not inhibit binding of E2F3 activator to Cyclin B1 promoter. This study suggested that the activation domain of CBF-B plays an essential role in the transcription activation of Cyclin B1 and Aurora A genes at G2/M phase, thus regulating cell cycle progression at G2/M phase.


Subject(s)
CCAAT-Binding Factor/metabolism , Cell Division/genetics , Cyclin B/genetics , G2 Phase/genetics , Protein Serine-Threonine Kinases/genetics , Transcriptional Activation , Adenoviridae/genetics , Aurora Kinases , CCAAT-Binding Factor/antagonists & inhibitors , CCAAT-Binding Factor/genetics , Cyclin B1 , Genetic Vectors , HeLa Cells , Humans , Promoter Regions, Genetic , Protein Structure, Tertiary , Sequence Deletion , TATA-Box Binding Protein/metabolism , Tetracycline/pharmacology
17.
Nucleic Acids Res ; 34(10): 3116-27, 2006.
Article in English | MEDLINE | ID: mdl-16757577

ABSTRACT

Response to stresses that alter the function of the endoplasmic reticulum is an important cellular function, which relies on the activation of specific genes. Several transcription factors (TFs) are known to affect this pathway. Using RT-PCR and ChIP assays, we studied the recruitment of promoter-specific TFs, general TFs and epigenetic marks in activated promoters. H3-K4 di- and tri-methylation and H3-K79 di-methylation are present before induction. H3 acetylation is generally high before induction, and H4 acetylation shows a promoter-specific increase. Interestingly, there is a depletion of histone H3 under maximal induction, explaining an apparent decrease of H3-K4 tri-methylation and H3-K79 di-methylation. Pol II is found enriched on some promoters under basal conditions, unlike TBP and p300, which are recruited selectively. Most genes are bound by XBP-1 after induction, some before induction, presumably by the inactive isoform. ATF6 and CHOP associate to largely different set of genes. C/EBPbeta is selective and binding to the CHOP promoter precedes that of XBP-1, ATF6 and CHOP. Finally, one of the ER-stress inducible genes analyzed, HRD1, is not bound by any of these factors. Among the constitutive TFs, NF-Y, but not Sp1, is found on all genes before induction. Intriguingly, siRNA interference of the NF-YB subunit indicates transcriptional impairment of some, but not all genes. These data highlight a previously unappreciated complexity of TFs binding and epigenetic changes, pointing to different TFs-specific pathways within this broad response.


Subject(s)
Epigenesis, Genetic , Histones/metabolism , Promoter Regions, Genetic , Transcription Factors/metabolism , Transcriptional Activation , Acetylation , CCAAT-Binding Factor/antagonists & inhibitors , CCAAT-Binding Factor/metabolism , Cell Line, Tumor , DNA-Binding Proteins/antagonists & inhibitors , Endoplasmic Reticulum/metabolism , Humans , Kinetics , Methylation , Transcription Factors/antagonists & inhibitors , Transcription Factors, General/metabolism
18.
Nucleic Acids Res ; 32(12): 3632-41, 2004.
Article in English | MEDLINE | ID: mdl-15243141

ABSTRACT

To understand the role of the CCAAT-binding factor, CBF, in transcription, we developed a strategy to purify the heterotrimeric CBF complex from HeLa cell extracts using two successive immunoaffinity chromatography steps. Here we show that the p32 protein, previously identified as the ASF/SF2 splicing factor-associated protein, copurified with the CBF complex. Studies of protein-protein interaction demonstrated that p32 interacts specifically with CBF-B subunit and also associates with CBF-DNA complex. Cellular localization by immunofluorescence staining revealed that p32 is present in the cell throughout the cytosol and nucleus, whereas CBF is present primarily in the nucleus. A portion of the p32 colocalizes with CBF-B in the nucleus. Interestingly, reconstitution of p32 in an in vitro transcription reaction demonstrated that p32 specifically inhibits CBF-mediated transcription activation. Altogether, our study identified p32 as a novel and specific corepressor of CBF-mediated transcription activation in vitro.


Subject(s)
CCAAT-Binding Factor/antagonists & inhibitors , Hyaluronan Receptors , Membrane Glycoproteins , Receptors, Complement/physiology , Repressor Proteins/physiology , Transcriptional Activation , CCAAT-Binding Factor/analysis , CCAAT-Binding Factor/metabolism , Carrier Proteins , Cell Nucleus/chemistry , Chromatography, Affinity , HeLa Cells , Humans , Macromolecular Substances , Mitochondrial Proteins , Protein Subunits/metabolism , Receptors, Complement/analysis , Receptors, Complement/metabolism , Recombinant Proteins/metabolism , Repressor Proteins/analysis , Repressor Proteins/metabolism
19.
J Am Chem Soc ; 125(28): 8420-1, 2003 Jul 16.
Article in English | MEDLINE | ID: mdl-12848532

ABSTRACT

Small molecule microarrays were screened to identify a small molecule ligand for Hap3p, a subunit of the yeast Hap2/3/4/5p transcription factor complex. The compound, named haptamide A, was determined to have a KD of 5.03 muM for binding to Hap3p using surface plasmon resonance analysis. Haptamide A also inhibited activation of a GDH1-lacZ reporter gene in a dose-dependent fashion. To explore structure-activity relationships, 11 derivatives of haptamide A were prepared using the same synthetic route that was developed for the original library synthesis. Analysis of dissociation constants and IC50 values for the reporter gene assay revealed a more potent inhibitor, haptamide B, with a KD of 330 nM. Whole-genome transcriptional profiling was used to compare effects of haptamide B with a hap3Delta yeast strain. Treatment with haptamide B, like the deletion mutant, reduced lactate-induced transcription of several genes from wild-type levels. Profiling the genetic "knockout" and the chemical genetic "knockdown" led to the identification of several genes that are regulated by Hap3p under nonfermentative conditions. These results demonstrate that a small molecule discovered using the small molecule microarray binding assay can permeate yeast cells and reach its target transcription factor protein in cells.


Subject(s)
Amides/pharmacology , CCAAT-Binding Factor/antagonists & inhibitors , Fungal Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Amides/chemical synthesis , Amides/metabolism , CCAAT-Binding Factor/genetics , CCAAT-Binding Factor/metabolism , Combinatorial Chemistry Techniques/methods , Fungal Proteins/genetics , Fungal Proteins/metabolism , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Kinetics , Pyrans/chemical synthesis , Pyrans/pharmacology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Structure-Activity Relationship , Surface Plasmon Resonance , Transcription Factors/genetics , Transcription Factors/metabolism
20.
J Biol Chem ; 278(21): 18868-76, 2003 May 23.
Article in English | MEDLINE | ID: mdl-12637525

ABSTRACT

Here we investigated the mechanisms by which mechanical stretch regulates the production of IL-8 in primary human airway smooth muscle cells (HASMC). Bronchial HASMC were subjected to cyclic mechanical stretch (12%, 1 Hz) using the computer-controlled Flexcell Strain system. Mechanical stretch increased IL-8 mRNA expression and protein production. Cyclic stretch of HASMC also increased the kinase activities of ERK1/2, JNK1, p38, and the DNA binding activities of AP-1 and C/EBP transcription factors with little effect on NF-kappa B. The inhibition of AP-1 and C/EBP transcriptional activities blocked the production of IL-8 in culture supernatants. Furthermore, the inhibition of ERK1/2 and p38 but not JNK1 caused a significant down-regulation in the expression and production of IL-8 in response to cyclic stretch. Although protein tyrosine kinases were required for the activation of both ERK1/2 and p38 kinase, stretch-activated channels, small GTPase proteins, and extracellular Ca2+ influx were required only for the activation of p38 kinase whereas phosphoinositide 3-kinase was needed for ERK1/2 activation. In addition, the phosphorylation of ERK1/2 was essential for the activation of AP-1 whereas p38 MAP kinase was needed for the activation of C/EBP. Our data demonstrate that the cyclic stretch of HASMC causes the increased production of IL-8 by activating the AP-1 and C/EBP transcription factors through the activation of ERK1/2 and p38 kinase signaling pathways.


Subject(s)
CCAAT-Binding Factor/physiology , Gene Expression Regulation , Interleukin-1/genetics , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth/physiology , Transcription Factor AP-1/physiology , Binding Sites , Biomechanical Phenomena , CCAAT-Binding Factor/antagonists & inhibitors , Calcium/pharmacology , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation , Enzyme Inhibitors/pharmacology , Humans , Ion Channels/physiology , MAP Kinase Kinase 1 , MAP Kinase Kinase 2 , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase 8 , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Muscle Spindles/physiology , Phosphatidylinositol 3-Kinases/metabolism , Physical Stimulation , Promoter Regions, Genetic , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , RNA, Messenger/analysis , Respiratory System , Reverse Transcriptase Polymerase Chain Reaction , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction , Transcription Factor AP-1/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...