Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
PLoS One ; 14(1): e0211046, 2019.
Article in English | MEDLINE | ID: mdl-30682194

ABSTRACT

BACKGROUND: Vascular endothelial growth factor (VEGF) is commonly overexpressed in a variety of tumor types including lung cancer. As a key regulator of angiogenesis, it promotes tumor survival, growth, and metastasis through the activation of the downstream protein kinase B (AKT) and extracellular signal-regulated kinase (ERK 1/2) activation. The VEGF promoter contains a 36 bp guanine-rich sequence (VEGFq) which is capable of forming quadruplex (four-stranded) DNA. This sequence has been implicated in the down-regulation of both basal and inducible VEGF expression and represents an ideal target for inhibition of VEGF expression. RESULTS: Our experiments demonstrate sequence-specific interaction between a G-rich quadruplex-forming oligonucleotide encoding a portion of the VEGFq sequence and its double stranded target sequence, suggesting that this G-rich oligonucleotide binds specifically to its complementary C-rich sequence in the genomic VEGF promoter by strand invasion. We show that treatment of A549 non-small lung cancer cells (NSCLC) with this oligonucleotide results in decreased VEGF expression and growth inhibition. The VEGFq oligonucleotide inhibits proliferation and invasion by decreasing VEGF mRNA/protein expression and subsequent ERK 1/2 and AKT activation. Furthermore, the VEGFq oligonucleotide is abundantly taken into cells, localized in the cytoplasm/nucleus, inherently stable in serum and intracellularly, and has no effect on non-transformed cells. Suppression of VEGF expression induces cytoplasmic accumulation of autophagic vacuoles and increased expression of LC3B, suggesting that VEGFq may induce autophagic cell death. CONCLUSION: Our data strongly suggest that the G-rich VEGFq oligonucleotide binds specifically to the C-rich strand of the genomic VEGF promoter, via strand invasion, stabilizing the quadruplex structure formed by the genomic G-rich sequence, resulting in transcriptional inhibition. Strand invading oligonucleotides represent a new approach to specifically inhibit VEGF expression that avoids many of the problems which have plagued the therapeutic use of oligonucleotides. This is a novel approach to specific inhibition of gene expression.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , G-Quadruplexes , Lung Neoplasms/drug therapy , Neoplasm Proteins/biosynthesis , Oligonucleotides/pharmacology , Promoter Regions, Genetic , Vascular Endothelial Growth Factor A/biosynthesis , A549 Cells , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Drug Delivery Systems , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Neoplasm Proteins/genetics , Oligonucleotides/genetics , Vascular Endothelial Growth Factor A/genetics
2.
Cancer Invest ; 36(9-10): 475-491, 2018.
Article in English | MEDLINE | ID: mdl-30396283

ABSTRACT

AS1411 is an antiproliferative DNA aptamer, which binds the ubiquitous protein, nucleolin. In this study, we show that constitutive overexpression of nucleolin confers increased sensitivity to the growth inhibitory effects of AS1411. HeLa cells overexpressing nucleolin have an increased growth rate and invasiveness relative to control cells. Nucleolin overexpressing cells demonstrate increased growth inhibition in response to the AS1411 treatment, which correlates with increased apoptosis and cell cycle arrest, when compared to non-transfected cells. AS1411 induces nucleolin expression at the RNA and protein level in HeLa cells, suggesting a feedback loop with important implications for the clinical use of AS1411.


Subject(s)
Oligodeoxyribonucleotides/pharmacology , Phosphoproteins/genetics , RNA-Binding Proteins/genetics , Up-Regulation , Uterine Cervical Neoplasms/genetics , Aptamers, Nucleotide , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Feedback, Physiological , Female , Gene Expression Regulation, Neoplastic/drug effects , HeLa Cells , Humans , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , Transfection , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/metabolism , Nucleolin
3.
PLoS One ; 11(8): e0161588, 2016.
Article in English | MEDLINE | ID: mdl-27551915

ABSTRACT

G-quadruplex forming sequences are particularly enriched in the promoter regions of eukaryotic genes, especially of oncogenes. One of the most well studied G-quadruplex forming sequences is located in the nuclease hypersensitive element (NHE) III1 of the c-MYC promoter region. The oncoprotein c-MYC regulates a large array of genes which play important roles in growth regulation and metabolism. It is dysregulated in >70% of human cancers. The silencer NHEIII1 located upstream of the P1 promoter regulates up-to 80% of c-MYC transcription and includes a G-quadruplex structure (Pu27) that is required for promoter inhibition. We have identified, for the first time, a family of seventeen G-quadruplex-forming motifs with >90% identity with Pu27, located on different chromosomes throughout the human genome, some found near or within genes involved in stem cell maintenance or neural cell development. Notably, all members of the Pu27 family interact specifically with NHEIII1 sequence, in vitro. Crosslinking studies demonstrate that Pu27 oligonucleotide binds specifically to the C-rich strand of the NHEIII1 resulting in the G-quadruplex structure stabilization. Pu27 homologous sequences (Pu27-HS) significantly inhibit leukemic cell lines proliferation in culture. Exposure of U937 cells to the Pu27-HS induces cell growth inhibition associated with cell cycle arrest that is most likely due to downregulation of c-MYC expression at the RNA and/or protein levels. Expression of SOX2, another gene containing a Pu27-HS, was affected by Pu27-HS treatment as well. Our data suggest that the oligonucleotides encoding the Pu27 family target complementary DNA sequences in the genome, including those of the c-MYC and SOX2 promoters. This effect is most likely cell type and cell growth condition dependent. The presence of genomic G-quadruplex-forming sequences homologous to Pu27 of c-MYC silencer and the fact that they interact specifically with the parent sequence suggest a common regulatory mechanism for genes whose promoters contain these sequences.


Subject(s)
Epistasis, Genetic , Gene Expression Regulation , Genome, Human , Genomics , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/genetics , Binding Sites , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Proliferation , Databases, Genetic , G-Quadruplexes , Genomics/methods , Humans , Models, Biological , Multigene Family , Protein Binding , Proto-Oncogene Proteins c-myc/chemistry , Transcription Factors/metabolism
4.
Oncotarget ; 6(26): 22270-81, 2015 Sep 08.
Article in English | MEDLINE | ID: mdl-26045302

ABSTRACT

AS1411 is a quadruplex-forming DNA oligonucleotide that functions as an aptamer to target nucleolin, a protein present on the surface of cancer cells. Clinical trials of AS1411 have indicated it is well tolerated with evidence of therapeutic activity, but improved pharmacology and potency may be required for optimal efficacy. In this report, we describe how conjugating AS1411 to 5 nm gold nanospheres influences its activities in vitro and in vivo. We find that the AS1411-linked gold nanospheres (AS1411-GNS) are stable in aqueous and serum-containing solutions. Compared to unconjugated AS1411 or GNS linked to control oligonucleotides, AS1411-GNS have superior cellular uptake and markedly increased antiproliferative/cytotoxic effects. Similar to AS1411, AS1411-GNS show selectivity for cancer cells compared to non-malignant cells. In a mouse model of breast cancer, systemic administration of AS1411-GNS could completely inhibit tumor growth with no signs of toxicity. These results suggest AS1411-GNS are promising candidates for clinical translation.


Subject(s)
Breast Neoplasms/therapy , Gold/administration & dosage , Metal Nanoparticles/administration & dosage , Nanospheres/administration & dosage , Oligodeoxyribonucleotides/administration & dosage , Animals , Aptamers, Nucleotide , Breast Neoplasms/genetics , Cell Line, Tumor , Female , Humans , MCF-7 Cells , Mice , Mice, Nude , Random Allocation , Xenograft Model Antitumor Assays
5.
PLoS One ; 9(12): e115580, 2014.
Article in English | MEDLINE | ID: mdl-25526084

ABSTRACT

Mutations occur at four specific sites in the hTERT promoter in >75% of glioblastomas and melanomas, but the mechanism by which the mutations affect gene expression remains unexplained. We report biophysical computational studies that show that the hTERT promoter sequence forms a novel G-quadruplex structure consisting of three contiguous, stacked parallel quadruplexes. The reported hTERT mutations map to the central quadruplex within this structure, and lead to an alteration of its hydrodynamic properties and stability.


Subject(s)
G-Quadruplexes , Models, Molecular , Promoter Regions, Genetic , Telomerase/chemistry , Base Sequence , Circular Dichroism , Computational Biology/methods , Humans , Hydrodynamics , Molecular Dynamics Simulation , Mutation , Telomerase/genetics
6.
J Biol Chem ; 289(12): 8521-31, 2014 Mar 21.
Article in English | MEDLINE | ID: mdl-24464582

ABSTRACT

Quadruplex-forming DNA sequences are present throughout the eukaryotic genome, including in telomeric DNA. We have shown that the c-Myc promoter quadruplex-forming sequence Pu-27 selectively kills transformed cells (Sedoris, K. C., Thomas, S. D., Clarkson, C. R., Muench, D., Islam, A., Singh, R., and Miller, D. M. (2012) Genomic c-Myc quadruplex DNA selectively kills leukemia. Mol. Cancer Ther. 11, 66-76). In this study, we show that Pu-27 induces profound DNA damage, resulting in striking chromosomal abnormalities in the form of chromatid or chromosomal breaks, radial formation, and telomeric DNA loss, which induces γ-H2AX in U937 cells. Pu-27 down-regulates telomeric shelterin proteins, DNA damage response mediators (RAD17 and RAD50), double-stranded break repair molecule 53BP1, G2 checkpoint regulators (CHK1 and CHK2), and anti-apoptosis gene survivin. Interestingly, there are no changes of DNA repair molecules H2AX, BRCA1, and the telomere maintenance gene, hTERT. ΔB-U937, where U937 cells stably transfected with deleted basic domain of TRF2 is partially sensitive to Pu-27 but exhibits no changes in expression of shelterin proteins. However, there is an up-regulation of CHK1, CHK2, H2AX, BRCA1, and survivin. Telomere dysfunction-induced foci assay revealed co-association of TRF1with γ-H2AX in ATM deficient cells, which are differentially sensitive to Pu-27 than ATM proficient cells. Alt (alternating lengthening of telomere) cells are relatively resistant to Pu-27, but there are no significant changes of telomerase activity in both Alt and non-Alt cells. Lastly, we show that this Pu-27-mediated sensitivity is p53-independent. The data therefore support two conclusions. First, Pu-27 induces DNA damage within both telomeric and nontelomeric regions of the genome. Second, Pu-27-mediated telomeric damage is due, at least in part, to compromise of the telomeric shelterin protein complex.


Subject(s)
DNA Damage , DNA/genetics , G-Quadruplexes , Genes, myc , Neoplasms/genetics , Telomere/genetics , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Base Sequence , Cell Death , Cell Line, Tumor , DNA/chemistry , DNA/metabolism , Histones/genetics , Histones/metabolism , Humans , Neoplasms/metabolism , Neoplasms/pathology , Phosphorylation , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Shelterin Complex , Telomere/chemistry , Telomere/metabolism , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
7.
Exp Mol Pathol ; 94(1): 84-90, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23000426

ABSTRACT

The primo vascular system (PVS), which is composed of very small primo-vessels (PV) and primo-nodes (PN), has recently emerged as a third component of circulatory system. Here, we report the presence of a tumor derived PVS in murine xenografts of human histiocytic lymphoma (U937) in close proximity to the tumor. Within this system, PNs are small (~500-600 µM diameter) membranous sac-like structures which contain numerous small cells which can be demonstrated by DAPI staining. Hematoxylin and Eosin (H&E) staining of the peri-tumoral PVS shows the presence of loose structures lined by fibroblasts but filled with dense fibers, cells, lacunae and nerve-like structures. The origin and type of cells within the PVS was characterized by immunostaining with antibodies for CD68, CD45 and lysozyme. The results of these studies reveal that the PVS of the xenograft originates from the human U937 tumor cells. qRT-PCR analysis of mRNA isolated from PVS cells reveals a striking predominance of human, rather than mouse, sequences. Of particular interest, human stem cell specific transcription factors were overexpressed, most notably KLF4, an upstream regulator of NANOG which maintains the pluripotent and undifferentiated state of stem cells. These results suggest that the cells present within the PVS are derived from the human xenograft and suggests that the primo-vessels associated with the xenografted tumor may provide a safe haven for a select population of cancer stem cells. Further understanding of the biological properties of these cells may allow the development of new anti-cancer interventions.


Subject(s)
Lymphoma, Large B-Cell, Diffuse/pathology , Neoplastic Stem Cells , Animals , Antigens, CD/analysis , Antigens, Differentiation, Myelomonocytic/analysis , Cell Line, Tumor , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Leukocyte Common Antigens/analysis , Leukocyte Common Antigens/immunology , Meridians , Mice , Mice, Inbred BALB C , Mice, Nude , Muramidase/metabolism , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , RNA, Messenger/analysis , Stem Cell Niche , Transplantation, Heterologous , U937 Cells
8.
Clin Cancer Res ; 18(20): 5546-53, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-23071356

ABSTRACT

The processes of cellular growth regulation and cellular metabolism are closely interrelated. The c-Myc oncogene is a "master regulator" which controls many aspects of both of these processes. The metabolic changes which occur in transformed cells, many of which are driven by c-Myc overexpression, are necessary to support the increased need for nucleic acids, proteins, and lipids necessary for rapid cellular proliferation. At the same time, c-Myc overexpression results in coordinated changes in level of expression of gene families which result in increased cellular proliferation. This interesting duality of c-Myc effects places it in the mainstream of transformational changes and gives it a very important role in regulating the "transformed phenotype." The effects induced by c-Myc can occur either as a "primary oncogene" which is activated by amplification or translocation or as a downstream effect of other activated oncogenes. In either case, it appears that c-Myc plays a central role in sustaining the changes which occur with transformation. Although efforts to use c-Myc as a therapeutic target have been quite frustrating, it appears that this may change in the next few years.


Subject(s)
Cell Transformation, Neoplastic , Glycolysis , Neoplasms , Proto-Oncogene Proteins c-myc , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Neoplastic , Glutamine/metabolism , Humans , Mitochondria/metabolism , Molecular Targeted Therapy , Neoplasms/genetics , Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism
9.
Mol Cancer Ther ; 11(1): 66-76, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22084162

ABSTRACT

c-Myc, a key regulator of cell cycle and proliferation, is commonly overexpressed in leukemia and associated with poor prognosis. Conventional antisense oligonucleotides targeting c-myc may attenuate leukemic cell growth, however, are poorly taken into cells, rapidly degraded, and have unwanted effects on normal cells. The c-myc promoter contains a guanine-rich sequence (PU27) capable of forming quadruplex (four-stranded) DNA, which may negatively regulate c-myc transcription. However, its biological significance is unknown. We show that treatment of leukemia with an oligonucleotide encoding the genomic PU27 sequence induces cell-cycle arrest and death by oncotic necrosis due to PU27-mediated suppression of c-myc mRNA/protein expression. Furthermore, PU27 is abundantly taken into cells, localized in the cytoplasm/nucleus, inherently stable in serum and intracellularly, and has no effect on normal cells. Suppression of c-myc expression by PU27 caused significant DNA damage, cell and mitochondrial swelling, and membrane permeability characteristic of oncotic necrosis. Induction of oncosis caused mitochondrial dysfunction, depletion of cellular ATP levels, and enhanced oxidative stress. This novel antileukemic strategy addresses current concerns of oligonucleotide therapeutics including problems with uptake, stability, and unintentional effects on normal cells and is the first report of selective cancer cell killing by a genomic DNA sequence.


Subject(s)
G-Quadruplexes , Leukemia/metabolism , Oligonucleotides, Antisense/pharmacology , Proto-Oncogene Proteins c-myc/genetics , Adenosine Triphosphate/metabolism , Cell Cycle/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Humans , Leukemia/pathology , Mitochondria/drug effects , Mitochondria/pathology , Oligonucleotides, Antisense/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Transcription, Genetic
10.
BMC Cancer ; 10: 157, 2010 Apr 22.
Article in English | MEDLINE | ID: mdl-20412594

ABSTRACT

BACKGROUND: Hypoxic microenvironments in tumors contribute to transformation, which may alter metabolism, growth, and therapeutic responsiveness. The alpha-enolase gene encodes both a glycolytic enzyme (alpha-enolase) and a DNA-binding tumor suppressor protein, c-myc binding protein (MBP-1). These divergent alpha-enolase gene products play central roles in glucose metabolism and growth regulation and their differential regulation may be critical for tumor adaptation to hypoxia. We have previously shown that MBP-1 and its binding to the c-myc P2 promoter regulates the metabolic and cellular growth changes that occur in response to altered exogenous glucose concentrations. RESULTS: To examine the regulation of alpha-enolase and MBP-1 by a hypoxic microenvironment in breast cancer, MCF-7 cells were grown in low, physiologic, or high glucose under 1% oxygen. Our results demonstrate that adaptation to hypoxia involves attenuation of MBP-1 translation and loss of MBP-1-mediated regulation of c-myc transcription, evidenced by decreased MBP-1 binding to the c-myc P2 promoter. This allows for a robust increase in c-myc expression, "early c-myc response", which stimulates aerobic glycolysis resulting in tumor acclimation to oxidative stress. Increased alpha-enolase mRNA and preferential translation/post-translational modification may also allow for acclimatization to low oxygen, particularly under low glucose concentrations. CONCLUSIONS: These results demonstrate that malignant cells adapt to hypoxia by modulating alpha-enolase/MBP-1 levels and suggest a mechanism for tumor cell induction of the hyperglycolytic state. This important "feedback" mechanism may help transformed cells to escape the apoptotic cascade, allowing for survival during limited glucose and oxygen availability.


Subject(s)
Breast Neoplasms/genetics , Cell Hypoxia/genetics , DNA-Binding Proteins/genetics , Phosphopyruvate Hydratase/genetics , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle/genetics , Cell Growth Processes/genetics , Cell Line, Tumor , DNA-Binding Proteins/biosynthesis , Excitatory Amino Acid Transporter 2/metabolism , Gene Expression Regulation, Neoplastic , Glucose/metabolism , Humans , Lactic Acid/biosynthesis , Phosphopyruvate Hydratase/biosynthesis , Phosphorylation , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Translocation, Genetic , Up-Regulation
11.
Exp Mol Pathol ; 86(3): 151-64, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19454272

ABSTRACT

Certain guanine-rich (G-rich) DNA and RNA molecules can associate intermolecularly or intramolecularly to form four stranded or "quadruplex" structures, which have unusual biophysical and biological properties. Several synthetic G-rich quadruplex-forming oligodeoxynucleotides have recently been investigated as therapeutic agents for various human diseases. We refer to these biologically active G-rich oligonucleotides as aptamers because their activities arise from binding to protein targets via shape-specific recognition (analogous to antibody-antigen binding). As therapeutic agents, the G-rich aptamers may have some advantages over monoclonal antibodies and other oligonucleotide-based approaches. For example, quadruplex oligonucleotides are non-immunogenic, heat stable and they have increased resistance to serum nucleases and enhanced cellular uptake compared to unstructured sequences. In this review, we describe the characteristics and activities of G-rich oligonucleotides. We also give a personal perspective on the discovery and development of AS1411, an antiproliferative G-rich phosphodiester oligonucleotide that is currently being tested as an anticancer agent in Phase II clinical trials. This molecule functions as an aptamer to nucleolin, a multifunctional protein that is highly expressed by cancer cells, both intracellularly and on the cell surface. Thus, the serendipitous discovery of the G-rich oligonucleotides also led to the identification of nucleolin as a new molecular target for cancer therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Oligodeoxyribonucleotides/therapeutic use , Animals , Antineoplastic Agents/chemistry , Aptamers, Nucleotide , Cell Proliferation/drug effects , Clinical Trials as Topic , Drug Discovery , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Oligodeoxyribonucleotides/chemistry , Phosphoproteins/drug effects , Phosphoproteins/metabolism , RNA-Binding Proteins/drug effects , RNA-Binding Proteins/metabolism , Nucleolin
12.
Cancer Res ; 67(21): 10491-500, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17974993

ABSTRACT

AS1411 is a quadruplex-forming oligonucleotide aptamer that targets nucleolin. It is currently in clinical trials as a treatment for various cancers. We have proposed that AS1411 inhibits cancer cell proliferation by affecting the activities of certain nucleolin-containing complexes. Here, we report that protein arginine methyltransferase 5 (PRMT5), an enzyme that catalyzes the formation of symmetrical dimethylarginine (sDMA), is a nucleolin-associated protein whose localization and activity are altered by AS1411. Levels of PRMT5 were found to be decreased in the nucleus of AS1411-treated DU145 human prostate cancer cells, but increased in the cytoplasm. These changes were dependent on nucleolin and were not observed in cells pretreated with nucleolin-specific small interfering RNA. Treatment with AS1411 altered levels of PRMT5 activity (assessed by sDMA levels) in accord with changes in its localization. In addition, our data indicate that nucleolin itself is a substrate for PRMT5 and that distribution of sDMA-modified nucleolin is altered by AS1411. Because histone arginine methylation by PRMT5 causes transcriptional repression, we also examined expression of selected PRMT5 target genes in AS1411-treated cells. For some genes, including cyclin E2 and tumor suppressor ST7, a significant up-regulation was noted, which corresponded with decreased PRMT5 association with the gene promoter. We conclude that nucleolin is a novel binding partner and substrate for PRMT5, and that AS1411 causes relocalization of the nucleolin-PRMT5 complex from the nucleus to the cytoplasm. Consequently, the nuclear activity of PRMT5 is decreased, leading to derepression of some PRMT5 target genes, which may contribute to the biological effects of AS1411.


Subject(s)
Oligodeoxyribonucleotides/pharmacology , Phosphoproteins/metabolism , Protein Methyltransferases/metabolism , RNA-Binding Proteins/metabolism , Aptamers, Nucleotide , Arginine/analogs & derivatives , Arginine/metabolism , Cell Cycle , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoplasm/metabolism , Gene Expression Regulation/drug effects , Humans , Male , Phosphoproteins/chemistry , Protein Transport , Protein-Arginine N-Methyltransferases , RNA-Binding Proteins/chemistry , Nucleolin
13.
Biochemistry ; 46(29): 8659-68, 2007 Jul 24.
Article in English | MEDLINE | ID: mdl-17595061

ABSTRACT

Alpha-enolase is a bifunctional gene encoding both a glycolytic enzyme and a DNA binding protein, c-myc binding protein (MBP-1). MBP-1 binds the c-myc promoter and downregulates c-myc transcription. Since these alpha-enolase gene products have important functions in glucose metabolism and growth regulation, this gene may play a central role in regulating the abnormal proliferative characteristics of transformed cells. To determine the role of alpha-enolase and MBP-1 in the cellular response to altered exogenous glucose concentration, MCF-7 cells were cultured in low (1 nM), physiological (5 mM), or high (25 mM) levels of glucose. Levels of alpha-enolase, MBP-1, and c-myc expression were compared to levels of cell proliferation and lactate production. At all glucose concentrations, MCF-7 cells demonstrated an initial increase in MBP-1 expression and a parallel decrease in c-myc transcript levels, which were accompanied by decreased proliferation. Cells grown in low glucose maintained the increased MBP-1 expression through 48 h, resulting in persistently lower rates of proliferation. However, physiologic or high glucose levels resulted in decreased MBP-1 expression, which was associated with increased cellular proliferation and lactate production. In these cells, c-myc mRNA returned to control levels as MBP-1 expression decreased. Cells grown in low glucose demonstrated a dramatic increase in c-myc mRNA at 48 h, which was associated with a loss in c-myc P2 promoter binding by MBP-1. This suggests that post-translational modifications of MBP-1 likely alter its DNA binding activity. These results demonstrate an important role for MBP-1 in the altered cell proliferation and energy utilization that occur in response to an altered glucose concentration.


Subject(s)
DNA-Binding Proteins/metabolism , Glucose/pharmacology , Transcription Factors/metabolism , Binding Sites , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , Cell Survival , DNA-Binding Proteins/genetics , Electrophoretic Mobility Shift Assay , Female , Genes, myc , Humans , Phosphopyruvate Hydratase/genetics , Phosphopyruvate Hydratase/metabolism , Promoter Regions, Genetic , RNA, Messenger/metabolism , Time Factors , Transcription Factors/genetics , Tumor Cells, Cultured , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
14.
Mol Cancer Ther ; 5(7): 1790-9, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16891465

ABSTRACT

AGRO100, also known as AS1411, is an experimental anticancer drug that recently entered human clinical trials. It is a member of a novel class of antiproliferative agents known as G-rich oligonucleotides (GRO), which are non-antisense, guanosine-rich phosphodiester oligodeoxynucleotides that form stable G-quadruplex structures. The biological activity of GROs results from their binding to specific cellular proteins as aptamers. One important target protein of GROs has been previously identified as nucleolin, a multifunctional protein expressed at high levels by cancer cells. Here, we report that AGRO100 also associates with nuclear factor-kappaB (NF-kappaB) essential modulator (NEMO), which is a regulatory subunit of the inhibitor of kappaB (IkappaB) kinase (IKK) complex, and also called IKKgamma. In the classic NF-kappaB pathway, the IKK complex is required for phosphorylation of IkappaBalpha and subsequent activation of the transcription factor NF-kappaB. We found that treatment of cancer cells with AGRO100 inhibits IKK activity and reduces phosphorylation of IkappaBalpha in response to tumor necrosis factor-alpha stimulation. Using a reporter gene assay, we showed that AGRO100 blocks both tumor necrosis factor-alpha-induced and constitutive NF-kappaB activity in human cancer cell lines derived from cervical, prostate, breast, and lung carcinomas. In addition, we showed that, in AGRO100-treated cancer cells, NEMO is coprecipitated by nucleolin, indicating that both proteins are present in the same complex. Our studies suggest that abrogation of NF-kappaB activity may contribute to the anticancer effects of AGRO100 and that nucleolin may play a previously unknown role in regulating the NF-kappaB pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Aptamers, Nucleotide/pharmacology , I-kappa B Kinase/metabolism , NF-kappa B/antagonists & inhibitors , Oligodeoxyribonucleotides/pharmacology , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Aptamers, Nucleotide/metabolism , Aptamers, Nucleotide/therapeutic use , Cell Line, Tumor , Female , Genes, Reporter/drug effects , Humans , I-kappa B Kinase/antagonists & inhibitors , Immunoprecipitation , Male , NF-kappa B/agonists , NF-kappa B/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Oligodeoxyribonucleotides/metabolism , Oligodeoxyribonucleotides/therapeutic use , Phosphorylation/drug effects , Nucleolin
15.
J Biol Chem ; 278(10): 8572-9, 2003 Mar 07.
Article in English | MEDLINE | ID: mdl-12506112

ABSTRACT

Molecular defects in apoptotic pathways are thought to often contribute to the abnormal expansion of malignant cells and their resistance to chemotherapy. Therefore, a comprehensive knowledge of the mechanisms controlling induction of apoptosis and subsequent cellular disintegration could result in improved methods for prognosis and treatment of cancer. In this study, we have examined apoptosis-induced alterations in two proteins, nucleolin and poly(ADP-ribose) polymerase-1 (PARP-1), in U937 leukemia cells. Nucleolin is expressed at high levels in malignant cells, and it is a multifunctional and mobile protein that can shuttle among the nucleolus, nucleoplasm, cytoplasm, and plasma membrane. Here, we report our findings that UV irradiation or camptothecin treatment of U937 cells induced apoptosis and caused a significant change in the levels and localization of nucleolin within the nucleus. Additionally, nucleolin levels were dramatically decreased in extracts containing the cytoplasm and plasma membrane. These alterations could be abrogated by pre-incubation with an inhibitor of PARP-1 (3-aminobenzamide), and our data support a potential role for nucleolin in removing cleaved PARP-1 from dying cells. Furthermore, both nucleolin and cleaved PARP-1 were detected in the culture medium of cells undergoing apoptosis, associated with particles of a size consistent with apoptotic bodies. These results indicate that nucleolin plays an important role in apoptosis, and could be a useful marker for assessing apoptosis or detecting apoptotic bodies. In addition, the data provide a possible explanation for the appearance of nucleolin and PARP-1 autoantibodies in some autoimmune diseases.


Subject(s)
Apoptosis , Leukemia, Myeloid/pathology , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , Apoptosis/radiation effects , Benzamides/pharmacology , Camptothecin/pharmacology , Enzyme Inhibitors/pharmacology , Humans , In Situ Nick-End Labeling , Leukemia, Myeloid/enzymology , Leukemia, Myeloid/metabolism , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/metabolism , Precipitin Tests , U937 Cells , Ultraviolet Rays , Nucleolin
16.
Nucleic Acids Res ; 30(11): 2565-74, 2002 Jun 01.
Article in English | MEDLINE | ID: mdl-12034846

ABSTRACT

The c-myc protooncogene plays a key role in the abnormal growth regulation of melanoma cells. We have targeted three polypurine sequences within the mouse myc mRNA with acridine-modified, clamp-forming antisense oligonucleotides (AS ODNs) in an effort to inhibit growth of murine melanoma cells. These ODNs are unique in that they hybridize to the target mRNA by both Watson-Crick and Hoogsteen hydrogen bond interactions, forming a triple-stranded structure. At a concentration of 3 microM E1C, E2C and E3C inhibit B16-F0 proliferation by 76, 66 and 78%, respectively. Both immunofluorescent staining and western blot analysis corroborate a proportional reduction in c-Myc expression by all three ODNs. There were clear distinctions in the ability of these ODNs to inhibit tumor progression in C57BL/6 mice as a function of Myc expression. There was no synergy demonstrated between ODN E1C with cisplatin (DDP), which inhibited tumor growth by 77% alone and 82% in combination. Although E2C inhibited growth by 54%, its effect was decreased to 32% with DDP, when compared with controls. E3C, on the other hand, demonstrated a synergistic effect with DDP, inhibiting growth by 72% in combination, but only by 1% as a single agent. Immunofluorescence analysis of tumors for each group revealed a concomitant reduction in c-Myc expression in tumors from mice treated with the most active clamp ODN alone (E1C) or clamp ODN + DDP (E1C/E3C + DDP). Western blot analysis confirmed this decrease in target protein expression. Our results document the growth-inhibitory activity of two myc-targeting antisense clamp ODNs; E1C, which has activity as a single agent, and E3C, which has in vivo synergy with DDP pretreatment. These data confirm the antiproliferative effects of these novel ODNs and document an interesting synergy with the chemotherapeutic agent DDP.


Subject(s)
Acridines/metabolism , Cisplatin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Genes, myc/genetics , Melanoma/genetics , Melanoma/pathology , Oligonucleotides, Antisense/pharmacology , Animals , Apoptosis/drug effects , Base Sequence , Blotting, Western , Cell Division/drug effects , Cisplatin/therapeutic use , Disease Progression , Drug Synergism , Fluorescent Antibody Technique , In Situ Nick-End Labeling , Melanoma/drug therapy , Mice , Mice, Inbred C57BL , Oligonucleotides, Antisense/metabolism , Oligonucleotides, Antisense/therapeutic use , Proliferating Cell Nuclear Antigen/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Tumor Cells, Cultured
17.
Biochemistry ; 41(11): 3676-85, 2002 Mar 19.
Article in English | MEDLINE | ID: mdl-11888284

ABSTRACT

Oligonucleotide-based therapies have considerable potential in cancer, viral, and cardiovascular disease therapies. However, it is becoming clear that the biological effects of oligonucleotides are not solely due to the intended sequence-specific interactions with nucleic acids. Oligonucleotides are also capable of interacting with numerous cellular proteins owing to their polyanionic character or specific secondary structure. We have examined the antiproliferative activity, protein binding, and G-quartet formation of a series of guanosine-rich oligonucleotides, which are analogues of GRO29A, a G-quartet forming, growth-inhibitory oligonucleotide, whose effects we have previously described [Bates P. J., Kahlon, J. B., Thomas, S. D., Trent, J. O., and Miller, D. M. (1999) J. Biol. Chem. 274, 26369-26377]. The GRO29A analogues include phosphorothioate (PS29A), 2'-O-methyl RNA (MR29A), and mixed DNA/2'-O-methyl RNA (MRdG29A) oligonucleotides. We demonstrate by UV spectroscopy that all of the modified analogues form stable structures, which are consistent with G-quartet formation. We find that the phosphorothioate and mixed DNA/2'-O-methyl analogues are able to significantly inhibit proliferation in a number of tumor cell lines, while the 2'-O-methyl RNA has no significant effects. Similar to the original oligonucleotide, GRO29A, the growth inhibitory oligonucleotides were able to compete with the human telomere sequence oligonucleotide for binding to a specific cellular protein. The less active MR29A does not compete significantly for this protein. On the basis of molecular modeling of the oligonucleotide structures, it is likely that the inactivity of MR29A is due to the differences in the groove structure of the quadruplex formed by this oligonucleotide. Interestingly, all GRO29A analogues, including an unmodified DNA phosphodiester oligonucleotide, are remarkably resistant to nuclease degradation in the presence of serum-containing medium, indicating that secondary structure plays an important role in biological stability. The remarkable stability and strong antiproliferative activity of these oligonucleotides confirm their potential as therapeutic agents.


Subject(s)
Cell Division/drug effects , Guanosine/chemistry , Oligonucleotides/pharmacology , Base Sequence , Circular Dichroism , Cytoplasm/metabolism , HeLa Cells , Humans , Models, Molecular , Oligonucleotides/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...