Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Genomics Proteomics ; 6(3): 161-75, 2009.
Article in English | MEDLINE | ID: mdl-19487545

ABSTRACT

BACKGROUND: The conformationally restricted polyamine analog PG-11047 has significant growth inhibitory activity against prostate and lung cancer cell lines and is currently under evaluation in several clinical trials, both alone and in combination with other drugs, for the treatment of relapsed or refractory cancer. The objective of this study was to identify the molecular signature of genes responsive to PG-11047 treatment and the biochemical effects of this drug in the HCT116 colon cancer cell line. MATERIALS AND METHODS: Gene expression analysis was performed using Affymetrix GeneChip human genome U133 Plus 2.0 arrays. Changes in protein expression were evaluated using 2D polyacrylamide gels followed by LCMS/MS. RESULTS: Treatment of cells with PG-11047 at concentrations ranging from 0.1 to 10 microM caused inhibition of cell growth. The activity of PG-11047 was found to correlate with its transcriptional effects on cell cycle control, focal adhesion, adherent and gap junction genes, MAPK-, Wnt- and, TGF-beta signaling pathways, transport and DNA/RNA transcription factor genes. PG-11047 caused depletion of polyamine pools. Proteomics analysis showed that PG-11047 restricts the modification of eukaryotic translation initiation factor 5A (eIF5A), resulting in suppression of general protein synthesis in PG-11047-treated cells. CONCLUSION: These data show that PG-11047 has a broad spectrum of anticancer activity in colon cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression/drug effects , Spermine/analogs & derivatives , Antineoplastic Agents/chemistry , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Drug Screening Assays, Antitumor , Genes, Neoplasm/drug effects , HCT116 Cells , Humans , Protein Synthesis Inhibitors/pharmacology , Proteins/genetics , Proteins/metabolism , Spermine/pharmacology
2.
J Biol Chem ; 283(39): 26428-35, 2008 Sep 26.
Article in English | MEDLINE | ID: mdl-18660501

ABSTRACT

SLC3A2, a member of the solute carrier family, was identified by proteomics methods as a component of a transporter capable of exporting the diamine putrescine in the Chinese hamster ovary (CHO) cells selected for resistance to growth inhibition by high exogenous concentrations of putrescine. Putrescine transport was increased in inverted plasma membrane vesicles prepared from cells resistant to growth inhibition by putrescine compared with transport in inverted vesicles prepared from non-selected cells. Knockdown of SLC3A2 in human cells, using short hairpin RNA, caused an increase in putrescine uptake and a decrease in arginine uptake activity. SLC3A2 knockdown cells accumulated higher polyamine levels and grew faster than control cells. The growth of SLC3A2 knockdown cells was inhibited by high concentrations of putrescine. Knockdown of SLC3A2 reduced export of polyamines from cells. Expression of SLC3A2 was suppressed in human HCT116 colon cancer cells, which have an activated K-RAS, compared with their isogenic clone, Hkh2 cells, which lack an activated K-RAS allele. Spermidine/spermine N(1)-acetyltransferase (SAT1) was co-immunoprecipitated by an anti-SLC3A2 antibody as was SLC3A2 with an anti-SAT1 antibody. SLC3A2 and SAT1 colocalized on the plasma membrane. These data provide the first molecular characterization of a polyamine exporter in animal cells and indicate that the diamine putrescine is exported by an arginine transporter containing SLC3A2, whose expression is negatively regulated by K-RAS. The interaction between SLC3A2 and SAT1 suggests that these proteins may facilitate excretion of acetylated polyamines.


Subject(s)
Acetyltransferases/metabolism , Cell Membrane/metabolism , Epithelial Cells/metabolism , Fusion Regulatory Protein 1, Heavy Chain/metabolism , Intestinal Mucosa/metabolism , Putrescine/metabolism , Animals , Arginine/metabolism , Biological Transport/physiology , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Epithelial Cells/cytology , Gene Expression Regulation/physiology , Humans , Intestinal Mucosa/cytology , Proteomics/methods , Proto-Oncogene Proteins p21(ras)/metabolism , Putrescine/pharmacology
3.
Int J Cancer ; 120(3): 459-68, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17096347

ABSTRACT

Elevated polyamine and nitric oxide levels (both derived from arginine) promote tumorigenesis, whereas non-steroidal anti-inflammatory drugs (NSAIDs) inhibit colorectal cancer (CRC) incidence in experimental and epidemiologic studies. We investigated dietary arginine-induced intestinal tumorigenesis and NSAID-inhibitory effects in Apc(Min/+) mice differentially expressing nitric oxide synthase-2 (Nos2). We also studied effects of estimated arginine exposures through meat consumption on tumor characteristics and survival in human CRC cases. Dietary arginine increased high-grade colon adenoma incidence in Apc(Min/+)Nos2(+/+) mice, but not in Nos2 knockout mice. Additionally, celecoxib suppressed intestinal steady state ornithine decarboxylase RNA levels (p < 0.001), induced steady state spermidine/spermine N(1)-acetyltransferase RNA levels (p = 0.002), decreased putrescine levels (p = 0.04) and decreased tumor number in the small intestines of Apc(Min/+)Nos2(+/+) mice (p = 0.0003). Five hundred and eleven cases from our NCI-supported CRC gene-environment study were analyzed based on self-reported meat (as a surrogate for arginine) consumption. Familial CRC cases (n = 144) in the highest meat consumption quartile (Q4) had no statistically significant differences in tumor grade compared to cases in Q1-Q3 (p = 0.32); however, they were observed to have decreased overall survival (OS) (10-year OS = 42% vs. 65%; p = 0.017), and increased risk of death in an adjusted analysis (hazards ratio [HR] = 2.24; p = 0.007). No differences in tumor grade, OS or adjusted HR were observed for sporadic CRC cases (n = 367) based on meat consumption. Our results suggest important roles for arginine and meat consumption in CRC pathogenesis, and have implications for CRC prevention.


Subject(s)
Arginine/administration & dosage , Colorectal Neoplasms/etiology , Eating/physiology , Meat/adverse effects , Acetyltransferases/genetics , Adult , Aged , Aged, 80 and over , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arginine/metabolism , Celecoxib , Colonic Neoplasms/etiology , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colorectal Neoplasms/physiopathology , Female , Gene Expression/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Ornithine Decarboxylase/genetics , Polyamines/metabolism , Pyrazoles/pharmacology , Risk Factors , Sulfonamides/pharmacology , Survival Analysis
4.
Technol Cancer Res Treat ; 5(6): 553-64, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17121431

ABSTRACT

Polyamine analogs are known to inhibit tumorigenesis at least in part by mimicking some of the regulatory roles of natural polyamines. To begin the identification of those signaling pathways that are involved in differential cellular responses to the synthetic conformationally restricted polyamine analog CGC-11093, we conducted gene expression profiling, proteomic, and genome-wide DNA methylation and histone acetylation analyses of the HCT116 colon adenocarcinoma cell line after treatment with this analog. Gene expression analysis was performed using Affymetrix GeneChip human genome U133 Plus 2.0 arrays. Changes in protein expression were evaluated using 2D polyacrylamide gels followed by LCMS/MS. DNA methylation was measured using 6,800 element CpG island microarrays. Treatment of cells with CGC-11093 at concentrations ranging from 0.1 to 10 microM caused inhibition of cell growth and metabolic activity, but only minimally affected cell viability. Gene expression analysis showed concentration-dependent effects of CGC-11093 on the DNA/RNA binding transcription factor, cell cycle, signaling, transport, cytoskeletal/structural, and serine protease genes. Functional gene analysis revealed distinct expression patterns related to inhibition of cell cycle control, TGF beta signaling, proteasome and RNA polymerase pathways, upregulation of the aminoacyl-tRNA synthesis pathway, and perturbations in the MAPK and Wnt signaling pathways. Microarray results were validated for selected genes with real time RT PCR. Proteomics analysis showed correlative changes in the expression of proteins involved in the regulation of proteasome function (proteasome subunit Y) and tRNA synthesis. CGC-11093 treatment did not produce any detectable changes in DNA methylation or histone acetylation in cells. This study validates specific target pathways for a specific conformationally restricted polyamine analog and suggests the utility of combined gene and DNA methylation microarrays along with proteomic analyses as a useful approach to the evaluation of the mechanisms of action of anticancer drugs.


Subject(s)
Adenocarcinoma/drug therapy , Biomarkers, Tumor/metabolism , Colonic Neoplasms/drug therapy , Polyamines/therapeutic use , Acetylation , Adenocarcinoma/genetics , Biomarkers, Tumor/genetics , Chromatin Immunoprecipitation , Colonic Neoplasms/genetics , CpG Islands , DNA Methylation , Gene Expression Profiling , HCT116 Cells/drug effects , HCT116 Cells/metabolism , Histones/metabolism , Humans , Neoplasm Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Pharmacogenetics , Proteomics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
5.
Mol Carcinog ; 45(10): 764-73, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16705737

ABSTRACT

We evaluated the role of polyamines in arginine-dependent intestinal tumorigenesis in Apc(Min) (/+) mice. Arginine is a substrate for ornithine synthesis and thus can influence polyamine production. Supplementing the diet with arginine increased intestinal and colonic polyamine levels and colonic carcinogenesis. Inhibiting polyamine synthesis with D,L-alpha-diflouromethylornithine (DFMO) decreased small intestinal and colonic polyamine pools. In mice provided basal diet, but not when supplemented with arginine, DFMO decreased small intestinal tumor number and burden, and increased intestinal apoptosis. In mice provided supplemental arginine in the diet, DFMO induced late apoptosis and decreased tumorigenesis in the colon. DFMO slightly reduced tumor incidence, number, and size while significantly decreasing tumor burden and grade. These changes in colon tumorigenesis did not occur in mice not provided supplemental arginine. Our study indicates that polyamines play unique roles in intestinal and colonic carcinogenesis in Apc(Min) (/+) mice. Inhibition of polyamine synthesis suppresses the arginine-dependent risk of colon tumorigenesis, resulting in apoptosis induction and decreased tumorigenesis, in this murine model.


Subject(s)
Arginine/pharmacology , Carcinogens/pharmacology , Colonic Neoplasms/chemically induced , Polyamines/pharmacology , Animal Feed , Animals , Apoptosis/drug effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/pathology , Colonic Neoplasms/pathology , Intestinal Absorption/drug effects , Mice , Mice, Inbred C57BL , Polyamines/pharmacokinetics
6.
Mol Carcinog ; 45(2): 93-105, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16329147

ABSTRACT

Arginine is catabolized by NOS2 and other nitric oxide synthases to form nitric oxide. We evaluated the roles of dietary arginine and Nos2 in Apc-dependent intestinal tumorigenesis in Min mice with and without a functional Nos2 gene. NOS2 protein was expressed only in intestinal tissues of Apc(Min/+) Nos2+/+ mice. NOS3 expression was higher in intestinal tissues of mice lacking Nos2, mainly in the small intestine. When diet was supplemented with arginine (0.2% and 2% in drinking water), lack of Nos2 results in decreased tumorigenesis in both small intestine and colon. In Nos2 knockout mice, supplemental arginine (up to 2%) caused a decrease in small intestinal tumor number and size. The arginine-dependent decrease was associated with an increase in nitrotyrosine formation and apoptosis in the region of intestinal stem cells. Mice expressing Nos2 did not show these changes. These mice did, however, show an arginine-dependent increase in colon tumor number and incidence, while no effect on apoptosis was seen. These changes were associated with increased nitrotyrosine formation in epithelial cells. Mice lacking Nos2 did not show changes in tumorigenesis or nitrotyrosine formation, while demonstrating an arginine-dependent increase in apoptosis. These data suggest that Nos2 and dietary arginine have significant effects on intestinal and colonic tumorigenesis in Min mice. In both tissues, loss of Nos2 is associated with decreased tumorigenesis when mice are supplemented with dietary arginine. In the small intestine, Nos2 prevents the arginine-induced decrease in tumor number and size, which is associated with NOS3 expression and increased apoptosis. In the colon, Nos2 is required for the arginine-induced increase in tumor number and incidence.


Subject(s)
Arginine/pharmacology , Colonic Neoplasms/enzymology , Intestinal Neoplasms/enzymology , Intestine, Small/pathology , Nitric Oxide Synthase Type II/metabolism , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Arginine/metabolism , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Dietary Supplements , Female , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Intestine, Small/drug effects , Male , Mice , Mice, Knockout , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type III/drug effects , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Tyrosine/analogs & derivatives , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...