Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters








Publication year range
1.
Exp Hematol Oncol ; 2(1): 25, 2013 Sep 08.
Article in English | MEDLINE | ID: mdl-24010672

ABSTRACT

BACKGROUND: Expression of the Epidermal Growth Factor Receptor ligand, Amphiregulin, has been associated with estrogen receptor positive breast cancer. As Amphiregulin is proteolytically released from the surface of breast cancer cells, we investigated the levels of Amphiregulin in the serum of breast cancer patients and cancer-free women to evaluate its potential utility as a breast cancer biomarker. FINDINGS: Serum Amphiregulin levels were quantified by ELISA from 125 cancer-free women and 114 breast cancer patients. No significant association between serum Amphiregulin levels and breast cancer status was detected at two cut-points evaluated. CONCLUSIONS: Measurement of serum Amphiregulin levels lacks the necessary sensitivity and specificity for breast cancer screening in the general population.

2.
J Biomed Biotechnol ; 2012: 180363, 2012.
Article in English | MEDLINE | ID: mdl-23093837

ABSTRACT

UNLABELLED: Valproic Acid (VPA), a histone deacetylase inhibitor, has been demonstrated to cause a marked decrease in proliferation of prostate cancer (PCa) cells in vitro and a significant reduction in tumor volume in vivo. The goal of this study is to better understand the VPA-induced growth inhibition in vivo, by studying expression of various markers in PCa xenografts. METHODS: For in vitro experiments, PCa cells were treated with 0, 0.6, and 1.2 mM VPA for 14 days. For in vivo models, experimental animals received 0.4% VPA in drinking water for 35 days. Tissue microarray was generated using cell pellets and excised xenografts. RESULTS: VPA treatment causes cell cycle arrest in PCa cells in vivo, as determined by increase in p21 and p27 and decrease in cyclin D1 expression. Increased expression of cytokeratin18 was also seen in xenografts. LNCaP xenografts in treated animals had reduced androgen receptor (AR) expression. While decreased proliferation was found in vitro, increase in apoptosis was found to be the reason for decreased tumor growth in vivo. Also, an anti-angiogenic effect was observed after VPA treatment. CONCLUSION: VPA inhibits tumor growth by multiple mechanisms including cell cycle arrest, induction of differentiation, and inhibition of growth of tumor vasculature.


Subject(s)
Cell Cycle Checkpoints/drug effects , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/physiopathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/physiopathology , Valproic Acid/administration & dosage , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Histone Deacetylase Inhibitors/administration & dosage , Humans , Male , Mice , Mice, Nude
3.
Int J Biochem Cell Biol ; 44(11): 1761-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22743332

ABSTRACT

Chk1 phosphorylation by the PI3-like kinases ATR and ATM is critical for its activation and its role in prevention of premature mitotic entry in response to DNA damage or stalled replication. The breast and ovarian tumor suppressor, BRCA1, is among several checkpoint mediators that are required for Chk1 activation by ATM and ATR. Previously we showed that BRCA1 is necessary for Chk1 phosphorylation and activation following ionizing radiation. BRCA1 has been implicated in S-phase checkpoint control yet its mechanism of action is not well characterized. Here we report that BRCA1 is critical for Chk1 phosphorylation in response to inhibition of replication by either cisplatin or hydroxyurea. While Chk1 phosphorylation of S317 is fully dependent on BRCA1, additional proteins may mediate S345 phosphorylation at later time points. In addition, we show that a subset of phosphorylated Chk1 is released from the chromatin in a BRCA1-dependent manner which may lead to the phosphorylation of Chk1 substrate, Cdc25C, on S216 and to S-phase checkpoint activation. Inhibition of Chk1 kinase by UCN-01 or expression of Chk1 phosphorylation mutants in which the serine residues were substituted with alanine residues abrogates BRCA1-dependent cell cycle arrest in response replication inhibition. These data reveal that BRCA1 facilitates Chk1 phosphorylation and its partial chromatin dissociation following replication inhibition that is likely to be required for S-phase checkpoint signaling.


Subject(s)
BRCA1 Protein/metabolism , Cell Cycle Checkpoints , Chromatin/metabolism , Protein Kinases/metabolism , S Phase , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/radiation effects , Cell Line, Tumor , Checkpoint Kinase 1 , Chromatin/drug effects , DNA Damage , DNA Replication/drug effects , DNA Replication/radiation effects , Enzyme Activation/drug effects , Enzyme Activation/radiation effects , Humans , Hydroxyurea/pharmacology , Mutant Proteins/metabolism , Phosphorylation/drug effects , Phosphorylation/radiation effects , Phosphoserine/metabolism , Radiation, Ionizing , S Phase/drug effects , S Phase/radiation effects , Signal Transduction/drug effects , Signal Transduction/radiation effects , Stress, Physiological/drug effects , Stress, Physiological/radiation effects
4.
Clin Biochem ; 45(6): 460-3, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22306169

ABSTRACT

OBJECTIVES: Prior to large studies in breast cancer patients, we have sought to establish the normal range of a potential serum biomarker, Amphiregulin, in healthy women and to determine whether sampling during the menstrual cycle influences the detected Amphiregulin levels. DESIGN AND METHODS: Serum Amphiregulin levels were quantified using a commercially available ELISA in 85 normal female donors. RESULTS: The range of circulating Amphiregulin was 0-4467 pg/mL. The majority of women had no detectable circulating Amphiregulin (n=54), and only five women had levels exceeding 500 pg/mL. Serum Amphiregulin levels did not vary significantly during the menstrual cycle (n=7 women). CONCLUSIONS: Detection of circulating Amphiregulin in a significant minority of healthy women suggests that it may not have the specificity necessary for a population screening tool; however its potential utility for monitoring response to treatment or disease progression should be examined in breast cancer cases.


Subject(s)
Glycoproteins/blood , Intercellular Signaling Peptides and Proteins/blood , Adult , Amphiregulin , EGF Family of Proteins , Female , Health , Humans , Menstrual Cycle/blood , Middle Aged , Protein Stability , Reference Values , Young Adult
5.
Cancer Prev Res (Phila) ; 5(2): 229-39, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22139053

ABSTRACT

Insufficient dose of dietary methyl groups are associated with a host of conditions ranging from neural tube defects to cancer. On the other hand, it is not certain what effect excess dietary methyl groups could have on cancer. This is especially true for prostate cancer, a disease that is characterized by increasing DNA methylation changes with increasing grade of the cancer. In this three-part study in animals, we look at (i) the effect of excess methyl donors on the growth rate of prostate cancer in vivo, (ii) the ability of 5-aza-2'-deoxycytidine (AdC), a demethylating agent, to demethylate in the presence of excess dietary methyl donors, and (iii) the effect of in utero feeding of excess methyl donors to the later onset of prostate cancer. The results show that when mice are fed a dietary excess of methyl donors, we do not see (i) an increase in the growth rate of DU-145 and PC-3 xenografts in vivo, or (ii) interference in the ability of AdC to demethylate the promoters of androgen receptor or Reprimo of prostate cancer xenografts but (iii) a protective effect on the development of higher grades of prostate cancer in the "Hi-myc" mouse model of prostate cancer which were fed the increased methyl donors in utero. We conclude that the impact of dietary methyl donors on prostate cancer progression depends upon the timing of exposure to the dietary agents. When fed before the onset of cancer, that is, in utero, excess methyl donors can have a protective effect on the progression of cancer.


Subject(s)
Azacitidine/analogs & derivatives , Cell Transformation, Neoplastic , DNA Methylation , Diet , Prostatic Neoplasms/pathology , Animals , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/pharmacology , Choline/administration & dosage , Decitabine , Disease Progression , Folic Acid/administration & dosage , Homocysteine/administration & dosage , Humans , Male , Methionine/administration & dosage , Mice , Mice, Nude , Prostatic Neoplasms/etiology , Tumor Cells, Cultured , Vitamin B 6/administration & dosage
6.
J Biomed Biotechnol ; 2011: 607480, 2011.
Article in English | MEDLINE | ID: mdl-20981253

ABSTRACT

Valproic Acid (VPA) is a histone deacetylase inhibitor that holds promise for cancer therapy. Here, we investigate whether VPA treatment induces neuroendocrine differentiation of Prostate Cancer (PCa). A tissue microarray of VPA-treated and untreated tumor xenografts and cell lines of human PCa (LNCaP, C4-2, DU145, and PC-3) were generated and were analyzed by immunohistochemical analysis (IHC) for NE markers chromogranin A (CgA), synaptophysin, and NCAM (neural cell adhesion molecule). Western blot analysis for CgA was performed to confirm the results of the TMA. IHC analysis did not reveal any induction of CgA, synaptophysin, or NCAM in any xenograft after VPA treatment in vivo. In vitro, VPA treatment induced little synaptophysin expression in C4-2 and PC-3 cells and NCAM expression in LNCaP and PC-3 cells. In the case of CgA, VPA treatment decreased its expression in vitro in a dose-dependent manner, as determined by western blot analysis. Thus our data demonstrates that VPA does not induce NE differentiation of PCa cells in the physiologically relevant in vivo setting.


Subject(s)
Cell Differentiation/drug effects , Neuroendocrine Cells/pathology , Prostatic Neoplasms/pathology , Valproic Acid/pharmacology , Animals , Cell Line, Tumor , Chromogranin A/metabolism , Humans , Male , Mice , Mice, Nude , Neural Cell Adhesion Molecules/metabolism , Prostatic Neoplasms/metabolism , Staining and Labeling , Synaptophysin/metabolism , Xenograft Model Antitumor Assays
7.
PLoS One ; 5(6): e11208, 2010 Jun 18.
Article in English | MEDLINE | ID: mdl-20585447

ABSTRACT

BACKGROUND: Histone deacetylase inhibitors (HDACis) re-express silenced tumor suppressor genes and are currently undergoing clinical trials. Although HDACis have been known to induce gene expression, an equal number of genes are downregulated upon HDAC inhibition. The mechanism behind this downregulation remains unclear. Here we provide evidence that several DNA repair genes are downregulated by HDAC inhibition and provide a mechanism involving the E2F1 transcription factor in the process. METHODOLOGY/PRINCIPAL FINDINGS: Applying Analysis of Functional Annotation (AFA) on microarray data of prostate cancer cells treated with HDACis, we found a number of genes of the DNA damage response and repair pathways are downregulated by HDACis. AFA revealed enrichment of homologous recombination (HR) DNA repair genes of the BRCA1 pathway, as well as genes regulated by the E2F1 transcription factor. Prostate cancer cells demonstrated a decreased DNA repair capacity and an increased sensitization to chemical- and radio-DNA damaging agents upon HDAC inhibition. Recruitment of key HR repair proteins to the site of DNA damage, as well as HR repair capacity was compromised upon HDACi treatment. Based on our AFA data, we hypothesized that the E2F transcription factors may play a role in the downregulation of key repair genes upon HDAC inhibition in prostate cancer cells. ChIP analysis and luciferase assays reveal that the downregulation of key repair genes is mediated through decreased recruitment of the E2F1 transcription factor and not through active repression by repressive E2Fs. CONCLUSIONS/SIGNIFICANCE: Our study indicates that several genes in the DNA repair pathway are affected upon HDAC inhibition. Downregulation of the repair genes is on account of a decrease in amount and promoter recruitment of the E2F1 transcription factor. Since HDAC inhibition affects several pathways that could potentially have an impact on DNA repair, compromised DNA repair upon HDAC inhibition could also be attributed to several other pathways besides the ones investigated in this study. However, our study does provide insights into the mechanism that governs downregulation of HR DNA repair genes upon HDAC inhibition, which can lead to rationale usage of HDACis in the clinics.


Subject(s)
DNA Repair/genetics , Down-Regulation/physiology , E2F1 Transcription Factor/physiology , Histone Deacetylases/metabolism , Prostatic Neoplasms/genetics , Recombination, Genetic , Cell Line, Tumor , Chromatin Immunoprecipitation , Comet Assay , DNA Damage , Fluorescent Antibody Technique , Humans , Male , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Prostatic Neoplasms/enzymology
8.
Cancer Biol Ther ; 8(3): 272-8, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19197146

ABSTRACT

In recent years, various dietary components that can potentially be used for the prevention and treatment of cancer have been identified. In this study, we demonstrate that extract (FE) from the seeds of the plant Trigonella foenum graecum, commonly called fenugreek, are cytotoxic in vitro to a panel of cancer but not normal cells. Treatment with 10-15 ug/mL of FE for 72 h was growth inhibitory to breast, pancreatic and prostate cancer cell lines (PCa). When tested at higher doses (15-20 ug/mL), FE continued to be growth inhibitory to PCa cell lines but not to either primary prostate or hTert-immortalized prostate cells. At least part of the growth inhibition is due to induction of cell death, as seen by incorporation of Ethidium Bromide III into cancer cells exposed to FE. Molecular changes induced in PCa cells are: in DU-145 cells: downregulation of mutant p53, and in PC-3 cells upregulation of p21 and inhibition of TGFbeta induced phosphorylation of Akt. The surprising finding of our studies is that death of cancer cells occurs despite growth stimulatory pathways being simultaneously upregulated (phosphorylated) by FE. Thus, these studies add another biologically active agent to our armamentarium of naturally occurring agents with therapeutic potential.


Subject(s)
Neoplasms/drug therapy , Phytotherapy , Plant Extracts/administration & dosage , Spices , Trigonella , Anticarcinogenic Agents/administration & dosage , Breast Neoplasms/drug therapy , Cell Death/drug effects , Cell Line, Tumor , Diosgenin/administration & dosage , Dose-Response Relationship, Drug , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Isothiocyanates , Male , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/drug therapy , Prostatic Neoplasms/drug therapy , Signal Transduction/drug effects , Sulfoxides , Thiocyanates/administration & dosage
9.
Clin Cancer Res ; 14(21): 6886-94, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18980983

ABSTRACT

PURPOSE: Although microarray technology has been widely adopted by the scientific community, analysis of the ensuing data remains challenging. In this article, we present our experience with a complex design microarray experiment on resistance mechanisms of histone deacetylase inhibitors (HDACI). EXPERIMENTAL DESIGN: To improve our understanding of the underlying mechanism of HDACI resistance in prostate cancer cells, we designed a novel "multiple-loop, double-cube" cDNA microarray experiment. In the experiment of 22 arrays, DU145 and PC3 cells were treated with two different HDACIs (vorinostat and valproic acid) and incubation periods (48 and 96 h). Preprocessing included exploratory analyses of the quality of the arrays and intensity-dependent within-array Loess normalization. An ANOVA model was used for inference. The results were validated by Western blot analysis of known treatment targets. RESULTS: Treatment of PC3 and DU145 cells with HDACIs caused 2.8% to 10% (P<0.001) differential expression across conditions; 51% to 73% of these genes were up-regulated and 28% to 49% were down-regulated. The extent of differential expression was associated with cell line (DU145>PC3), HDACI (valproic acid >or= vorinostat), and duration of treatment (96>48 h). We identified known and new treatment targets involved in cell cycle and apoptosis. CONCLUSION: A multiple-loop, double-cube microarray design can be used to identify HDACI-induced changes in gene expression possibly related to drug resistance.


Subject(s)
Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Histone Deacetylase Inhibitors , Microarray Analysis/methods , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm , Gene Expression Profiling , Humans , Hydroxamic Acids/therapeutic use , Male , Valproic Acid/therapeutic use , Vorinostat
10.
PLoS One ; 2(9): e844, 2007 Sep 05.
Article in English | MEDLINE | ID: mdl-17786215

ABSTRACT

Cell to cell adhesion is mediated by adhesion molecules present on the cell surface. Downregulation of molecules that form the adhesion complex is a characteristic of metastatic cancer cells. Downregulation of the N-myc down regulated gene1 (NDRG1) increases prostate and breast metastasis. The exact function of NDRG1 is not known. Here by using live cell confocal microscopy and in vitro reconstitution, we report that NDRG1 is involved in recycling the adhesion molecule E-cadherin thereby stabilizing it. Evidence is provided that NDRG1 recruits on recycling endosomes in the Trans Golgi network by binding to phosphotidylinositol 4-phosphate and interacts with membrane bound Rab4aGTPase. NDRG1 specifically interacts with constitutively active Rab4aQ67L mutant protein and not with GDP-bound Rab4aS22N mutant proving NDRG1 as a novel Rab4a effector. Transferrin recycling experiments reveals NDRG1 colocalizes with transferrin during the recycling phase. NDRG1 alters the kinetics of transferrin recycling in cells. NDRG1 knockdown cells show a delay in recycling transferrin, conversely NDRG1 overexpressing cells reveal an increase in rate of transferrin recycling. This novel finding of NDRG1 as a recycling protein involved with recycling of E-cadherin will aid in understanding NDRG1 role as a metastasis suppressor protein.


Subject(s)
Cadherins/metabolism , Cell Cycle Proteins/physiology , Intracellular Signaling Peptides and Proteins/physiology , rab4 GTP-Binding Proteins/physiology , Cell Cycle Proteins/genetics , Cell Line , Gene Knockdown Techniques , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/genetics , Microscopy, Confocal , Tissue Array Analysis
11.
BMC Bioinformatics ; 8: 142, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17472750

ABSTRACT

BACKGROUND: Pre-processing methods for two-sample long oligonucleotide arrays, specifically the Agilent technology, have not been extensively studied. The goal of this study is to quantify some of the sources of error that affect measurement of expression using Agilent arrays and to compare Agilent's Feature Extraction software with pre-processing methods that have become the standard for normalization of cDNA arrays. These include log transformation followed by loess normalization with or without background subtraction and often a between array scale normalization procedure. The larger goal is to define best study design and pre-processing practices for Agilent arrays, and we offer some suggestions. RESULTS: Simple loess normalization without background subtraction produced the lowest variability. However, without background subtraction, fold changes were biased towards zero, particularly at low intensities. ROC analysis of a spike-in experiment showed that differentially expressed genes are most reliably detected when background is not subtracted. Loess normalization and no background subtraction yielded an AUC of 99.7% compared with 88.8% for Agilent processed fold changes. All methods performed well when error was taken into account by t- or z-statistics, AUCs > or = 99.8%. A substantial proportion of genes showed dye effects, 43% (99% CI: 39%, 47%). However, these effects were generally small regardless of the pre-processing method. CONCLUSION: Simple loess normalization without background subtraction resulted in low variance fold changes that more reliably ranked gene expression than the other methods. While t-statistics and other measures that take variation into account, including Agilent's z-statistic, can also be used to reliably select differentially expressed genes, fold changes are a standard measure of differential expression for exploratory work, cross platform comparison, and biological interpretation and can not be entirely replaced. Although dye effects are small for most genes, many array features are affected. Therefore, an experimental design that incorporates dye swaps or a common reference could be valuable.


Subject(s)
Oligonucleotide Array Sequence Analysis/methods , Animals , Cell Line, Tumor , Databases, Genetic/classification , Databases, Genetic/statistics & numerical data , Dogs , Gene Expression Profiling/methods , Gene Expression Profiling/statistics & numerical data , Humans , Mice , Oligonucleotide Array Sequence Analysis/statistics & numerical data
12.
Prostate ; 67(10): 1099-110, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17477369

ABSTRACT

BACKGROUND: Valproic acid (VPA), is a drug approved by the FDA for epilepsy and bipolar disorders. It is a known Histone Deacetylase Inhibitor (HDACI). We tested VPA, for its anti-proliferative activity in prostate cancer (PCa) cell lines in vitro and in vivo. METHODS: DU-145 and PC-3 PCa cell lines were cultured with different doses of VPA. Cells were examined for their viability, cell cycle status and expression of cell cycle arrest, and proliferation markers. Nude mice bearing xenografts of human PCa cell lines, DU-145, and PC-3, were administered VPA in their drinking water. RESULTS: VPA displayed a dose- and time-dependent anti-proliferative effect on DU-145 and PC-3 PCa cell lines in vitro. A sustained effect of the drug was seen on cell cycle arrest even at 24 hr after removal of the drug, after which the effects returned to the basal state. Administration of 0.4% w/v VPA in drinking water (resulting in 0.4 mM VPA, in plasma) was effective in inducing growth arrest, cell death, and senescence in vivo and was also anti-angiogenic. The activation of all or some of these anti-proliferative pathways may be contingent on acetylation status of histones, confirmed by detection of increased acetyl-H3K9 in VPA-treated samples when compared with untreated controls. Pharmacodynamic studies showed an increase in expression of p21 and decrease in PCNA in xenografts of VPA-treated mice compared with protein expression in untreated controls. CONCLUSIONS: VPA may be functioning as an HDACI to inhibit growth of PCa cells in vitro and in vivo by modulating multiple pathways including cell cycle arrest, apoptosis, angiogenesis, and senescence.


Subject(s)
Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , Prostatic Neoplasms/pathology , Valproic Acid/pharmacology , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cellular Senescence/drug effects , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic , Histone Deacetylase Inhibitors , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Male , Mice , Mice, Nude , Neovascularization, Pathologic , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Xenograft Model Antitumor Assays
14.
Cancer Res ; 66(14): 7237-44, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16849572

ABSTRACT

Valproic acid (VPA) is an established drug in the long-term therapy of seizure disorders. Recently, VPA has been associated with anticancer activity, an effect thought to be mediated through the inhibition of cellular histone deacetylase 1. We investigated the effect of various doses of VPA (0, 1.2, and 5.0 mmol/L) administered either acutely or chronically on histone acetylation, p21 gene expression, androgen receptor expression, prostate-specific antigen (PSA) expression, and cell survival and proliferation in prostate cancer cell lines. We also studied the effect of chronic VPA on tumor xenograft growth in vivo. Our results show that acute treatment (3 days) VPA can increase net histone H3 acetylation and up-regulate p21, AR, and cytosolic PSA expression. Interestingly, the effects on AR and PSA are reversed with chronic treatment. In addition, acute VPA reduces cell survival but has no effect on the subsequent proliferation of surviving cells following drug withdrawal. However, when VPA is chronically administered (10-14 days) to prostate cancer cells, even lower doses of VPA result in marked decreases in the net proliferation rate, correlating with increased caspase-2 and caspase-3 activation. These effects are evident in both androgen receptor-positive (LNCaP and C4-2) and androgen receptor-negative (DU145 and PC3) prostate cancer cells. Moreover, chronic VPA treatment results in statistically significant reduction of tumor xenograft growth in vivo. We conclude that acute treatment has nominal effects on prostate cancer cell survival and proliferation, but chronic VPA results in profound decreases in proliferation, independently of androgen regulation.


Subject(s)
Prostatic Neoplasms/drug therapy , Valproic Acid/administration & dosage , Acetylation/drug effects , Animals , Cell Growth Processes/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Administration Schedule , Histones/metabolism , Humans , Male , Metribolone/pharmacology , Mice , Mice, Nude , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/pathology , Xenograft Model Antitumor Assays
15.
Clin Cancer Res ; 12(2): 634-42, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16428510

ABSTRACT

PURPOSE: Angiogenesis is required for tumor progression and represents a rational target for therapeutic intervention. Histone deacetylase (HDAC) inhibitors have been shown to have activity against various tumor cell types by inhibiting proliferation and inducing apoptosis both in vitro and in vivo. HDAC inhibitors have also been reported to inhibit angiogenesis. The goal of this study was to characterize the antiangiogenic and antitumor activity of a recently developed HDAC inhibitor, the hydroxamic derivative LBH589. MATERIALS AND METHODS: To evaluate the antiangiogenesis activity of LBH589, we did cell cycle analysis, cell proliferation, tube formation, invasion assays in vitro, and Matrigel plug assay in vivo. To determine the antitumor activity of LBH589, we established human prostate carcinoma cell PC-3 xenografts in vivo. To evaluate the effect of LBH589 on endothelial signaling pathways, gene expression, and protein acetylation, we did Western blots and reverse transcription-PCR in human umbilical vein endothelial cells (HUVEC). Immunohistochemical analysis was done to evaluate new blood vessel formation in vivo. RESULTS: LBH589 induced acetylation of histone H3 and alpha-tubulin protein in HUVECs. Histone and nonhistone protein acetylation correlated with induction of G(2)-M cell cycle arrest, inhibition of HUVEC proliferation, and viability. Noncytotoxic concentrations of LBH589 inhibited endothelial tube formation, Matrigel invasion, AKT, extracellular signal-regulated kinase 1/2 phosphorylation, and chemokine receptor CXCR4 expression. In vivo dosing of mice with LBH589 (10 mg/kg/d) reduced angiogenesis and PC-3 tumor growth. CONCLUSION: This study provides evidence that LBH589 induces a wide range of effects on endothelial cells that lead to inhibition of tumor angiogenesis. These results support the role of HDAC inhibitors as a therapeutic strategy to target both the tumor and endothelial compartment and warrant the clinical development of these agents in combination with angiogenesis inhibitors.


Subject(s)
Cell Division/drug effects , Cell Proliferation/drug effects , G2 Phase/drug effects , Neovascularization, Pathologic/drug therapy , Prostatic Neoplasms/drug therapy , Acetylation/drug effects , Angiogenesis Inhibitors/pharmacology , Animals , Blotting, Western , Collagen/metabolism , Drug Combinations , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Histones/metabolism , Humans , Hydroxamic Acids/therapeutic use , Indoles , Laminin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Invasiveness , Panobinostat , Phosphorylation/drug effects , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proteoglycans/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, CXCR4/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Transplantation, Heterologous , Tubulin/metabolism , Umbilical Veins/cytology , Umbilical Veins/drug effects , Umbilical Veins/metabolism , Vascular Endothelial Growth Factor A/metabolism
16.
IDrugs ; 8(2): 144-54, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15696416

ABSTRACT

Preclinical studies have demonstrated that several genes silenced in cancer cells can be re-expressed in their fully functional state by epigenetic modifiers such as DNA methyltransferase inhibitors and histone deacetylase inhibitors (HDACIs). While gene re-expression may be a reason for the success of HDACIs in preclinical and clinical studies, it is not the only factor. HDACIs display pleiotropic effects, including inhibition of cell cycle progression, differentiation, apoptosis and anti-proliferative effects. In addition, many studies have indicated that combining HDACIs with other agents results in an enhanced anti-proliferative effect. Structure-activity relationship studies of HDACIs with their substrates, histone deacetylases, have enabled design and synthesis of improved HDACIs. Due to their activity and perceived low toxicity, HDACIs have gained popularity as agents for clinical investigation. This review focuses on the cellular and biological effects of HDACIs, either alone or in combination with other agents. A brief summary on completed and ongoing cancer clinical trials is provided.


Subject(s)
Enzyme Inhibitors/therapeutic use , Histone Deacetylase Inhibitors , Histone Deacetylases/metabolism , Neoplasms/drug therapy , Acetylation , Animals , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Neoplasms/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL