Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 14 de 14
1.
Carcinogenesis ; 39(2): 146-157, 2018 02 09.
Article En | MEDLINE | ID: mdl-29106440

Microsatellite instability (MSI) is present in ulcerative colitis (UC) and colitis-associated colorectal cancers (CAC). Certain factors released by polymorphonuclear cells (PMNs) may drive mucosal frameshift mutations resulting in MSI and cancer. Here, we applied a co-culture system with PMNs and colon epithelial cells to identify such culprit factors. Subjecting HCT116 + chr3 and human colonic epithelial cells (HCEC)-1CT MSI-reporter cell lines harboring mono-, di- or tetranucleotide DNA repeats linked to enhanced green fluorescent protein (EGFP) to activated PMNs induced frameshift mutations within all repeats, as quantified by flow cytometry. Activated PMNs released superoxide and hydrogen peroxide (H2O2), as measured by lucigenin-amplified chemiluminescence and fluorometry, respectively. Catalase, which scavenges H2O2, reduced such PMN-induced MSI. The NADPH-oxidase inhibitor apocynin, which blocks the oxidative burst in PMNs, similarly inhibited PMN-induced MSI. A bead-based multiplex assay revealed that PMNs release a wide range of cytokines such as interleukin (IL)-8, IL-6 and tumor necrosis factor-α (TNF-α). In vitro, these cytokines increased MSI in colon epithelial cells, and the Janus kinase (JAK) inhibitor tofacitinib abolished IL-6-induced or PMN-induced MSI. Intracellular reactive oxygen species (ROS) formation, as measured by 2',7'-dichlorofluorescein diacetate (DCFDA) assay, was induced upon cytokine treatment. DNA oxidation upon IL-6 was present, as detected by formamidopyrimidine glycosylase (FPG)-modified comet assay. In conclusion, activated PMNs induce frameshift mutations in colon epithelial cells resulting in MSI. Both oxidative burst with release of ROS and PMN-secreted cytokines, such as IL-8, IL-6 or TNF-α, contribute to MSI. ROS scavengers and/or specific inhibitors of cytokine signaling may delay or prevent cancer development in the setting of colitis.


Colitis/complications , Colorectal Neoplasms/etiology , Microsatellite Instability , Mutagenesis/physiology , Neutrophils/metabolism , Cell Line, Tumor , Coculture Techniques , Colitis/metabolism , Cytokines/metabolism , Frameshift Mutation , Humans , Oxidative Stress/physiology , Reactive Oxygen Species/adverse effects , Reactive Oxygen Species/metabolism
2.
Gut ; 64(12): 1905-12, 2015 Dec.
Article En | MEDLINE | ID: mdl-25429050

OBJECTIVE: Lynch syndrome is caused by germline mutations in DNA mismatch repair genes leading to microsatellite instability (MSI) and colorectal cancer. Mesalazine, commonly used for the treatment of UC, reduces MSI in vitro. Here, we tested natural compounds for such activity and applied mesalazine and thymoquinone in a Msh2(loxP/loxP) Villin-Cre mouse model for Lynch syndrome. DESIGN: Flow cytometry was used for quantitation of mutation rates at a CA13 microsatellite in human colon cancer (HCT116) cells that had been stably transfected with pIREShyg2-enhanced green fluorescent protein/CA13, a reporter for frameshift mutations. Mice were treated for 43 weeks with mesalazine, thymoquinone or control chow. Intestines were analysed for tumour incidence, tumour multiplicity and size. MSI testing was performed from microdissected normal intestinal or tumour tissue, compared with mouse tails and quantified by the number of mutations per marker (NMPM). RESULTS: Besides mesalazine, thymoquinone significantly improved replication fidelity at 1.25 and 2.5 µM in HCT116 cells. In Msh2(loxP/loxP) Villin-Cre mice, tumour incidence was reduced by mesalazine from 94% to 69% (p=0.04) and to 56% (p=0.003) by thymoquinone. The mean number of tumours was reduced from 3.1 to 1.4 by mesalazine (p=0.004) and to 1.1 by thymoquinone (p<0.001). Interestingly, MSI was reduced in normal intestinal tissue from 1.5 to 1.2 NMPM (p=0.006) and to 1.1 NMPM (p=0.01) by mesalazine and thymoquinone, respectively. Thymoquinone, but not mesalazine, reduced MSI in tumours. CONCLUSIONS: Mesalazine and thymoquinone reduce tumour incidence and multiplicity in Msh2(loxP/loxP) Villin-Cre mice by reduction of MSI independent of a functional mismatch repair system. Both substances are candidate compounds for chemoprevention in Lynch syndrome mutation carriers.


Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Benzoquinones/therapeutic use , Colorectal Neoplasms, Hereditary Nonpolyposis/prevention & control , Mesalamine/therapeutic use , MutS Homolog 2 Protein/genetics , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Benzoquinones/pharmacology , Cell Proliferation/drug effects , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , Disease Models, Animal , Female , Frameshift Mutation , HCT116 Cells , Humans , Intestinal Mucosa/metabolism , Male , Mesalamine/pharmacology , Mice , Microsatellite Instability/drug effects , MutS Homolog 2 Protein/metabolism , Mutation Rate , Tumor Burden/drug effects
3.
Biochem Pharmacol ; 87(2): 312-20, 2014 Jan 15.
Article En | MEDLINE | ID: mdl-24184502

Genome wide association studies have implicated intestinal barrier function genes in the pathogenesis of ulcerative colitis. One of such loci CDH1, encoding E-cadherin, a transmembrane glycoprotein with known tumor suppressor functions, is also linked to the susceptibility to colorectal cancer. Loss of membranous E-cadherin expression is common in both colitis and cancer. We have recently demonstrated that mesalamine (5-ASA); the anti-inflammatory drug used to treat ulcerative colitis, induces membranous expression of E-cadherin and increases intercellular adhesion. Using colorectal cancer epithelial cells with aberrant E-cadherin expression, we investigated the mechanism underlying such an effect of 5-ASA. Post-translational modification of E-cadherin glycosylation was analyzed by biotin/streptavidin detection of sialylated glycoproteins. GnT-III (N-acetylglucosaminyltransferase III) expression was assessed by qRT-PCR, Western blot and immunofluorescence. GnT-III activity was analyzed by reactivity with E-4/L-4-PHA. Expression, localization and interaction of E-cadherin and ß-catenin were analyzed by Western blot, immunocytochemistry and RNA interference. 5-ASA activity modulated E-cadherin glycosylation and increased both mRNA and protein levels of GnT-III and its activity as detected by increased E4-lectin reactivity. Intestinal APC(Min) polyps in mice showed low expression of GnT-III and 5-ASA was effective in increasing its expression. The data demonstrated that remodeling of glycans by GnT-III mediated bisect glycosylation, contributes to the membranous retention of E-cadherin by 5-ASA; facilitating intercellular adhesion. Induction of membranous expression of E-cadherin by 5-ASA is a novel mechanism for mucosal healing in colitis that might impede tumor progression by modulation of GnT-III expression.


Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cadherins/biosynthesis , Colon/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Intestinal Mucosa/metabolism , Mesalamine/pharmacology , Animals , Colon/drug effects , Colon/pathology , Female , Glycosylation/drug effects , HCT116 Cells , HT29 Cells , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
4.
PLoS One ; 8(8): e73399, 2013.
Article En | MEDLINE | ID: mdl-24015303

BACKGROUND: Pulmonary arterial hypertension is characterized by increased thickness of pulmonary vessel walls due to both increased proliferation of pulmonary arterial smooth muscle cell (PASMC) and deposition of extracellular matrix. In patients suffering from pulmonary arterial hypertension, endothelin-1 (ET-1) synthesis is up-regulated and may increase PASMC activity and vessel wall remodeling through transforming growth factor beta-1 (TGF-ß1) and connective tissue growth factor. OBJECTIVE: To assess the signaling pathway leading to ET-1 induced proliferation and extracellular matrix deposition by human PASMC. METHODS: PASMC were serum starved for 24 hours before stimulation with either ET-1 and/or TGF-ß1. ET-1 was inhibited by Bosentan, ERK1/2 mitogen activated protein kinase (MAPK) was inhibited by U0126 and p38 MAPK was inhibited by SB203580. RESULTS: ET-1 increased PASMC proliferation when combined with serum. This effect involved the mitogen activated protein kinases (MAPK) ERK1/2 MAPK and was abrogated by Bosentan which caused a G1- arrest through activation of p27((Kip)). Regarding the contribution of extracellular matrix deposition in vessel wall remodeling, TGF-ß1 increased the deposition of collagen type-I and fibronectin, which was further increased when ET-1 was added mainly through ERK1/2 MAPK. In contrast, collagen type-IV was not affected by ET-1. Bosentan dose-dependently reduced the stimulatory effect of ET-1 on collagen type-I and fibronectin, but had no effect on TGF-ß1. CONCLUSION AND CLINICAL RELEVANCE: ET-1 alone does not induce PASMC proliferation and extracellular matrix deposition. However, ET-1 significantly up-regulates serum induced proliferation and TGF-ß1 induced extracellular matrix deposition, specifically of collagen type-I and fibronectin. The synergistic effects of ET-1 on serum and TGF-ß1 involve ERK1/2 MAPK and may thus present a novel mode of action in the pathogenesis of pulmonary arterial hypertension.


Cell Proliferation , Endothelial Cells/metabolism , Endothelin-1/metabolism , Hypertension, Pulmonary/metabolism , Transforming Growth Factor beta1/metabolism , Cell Line , Collagen Type I/biosynthesis , Endothelial Cells/pathology , Endothelin-1/agonists , Enzyme Inhibitors/pharmacology , Extracellular Matrix/metabolism , Fibronectins/biosynthesis , Humans , Hypertension, Pulmonary/pathology , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Transforming Growth Factor beta1/agonists
5.
Mol Cancer ; 12(1): 41, 2013 May 13.
Article En | MEDLINE | ID: mdl-23668310

BACKGROUND: Patients with familial adenomatous polyposis (FAP) are at increased risk for the development of colorectal cancer. Surgery and chemoprevention are the most effective means to prevent cancer development. Thymoquinone (TQ) is considered the main compound of the volatile Nigella sativa seed oil and has been reported to possess anticarcinogenic properties. In this study we evaluated the chemopreventive properties of TQ in a mouse model of FAP. METHODS: APCMin mice were fed with chow containing 37.5 mg/kg or 375 mg/kg TQ for 12 weeks. H&E stained intestine tissue sections were assessed for tumor number, localization, size, and grade. Immunohistochemistry for ß-catenin, c-myc, Ki-67 and TUNEL-staining was performed to investigate TQ's effect on major colorectal cancer pathways. TQ's impact on GSK-3ß and ß-catenin were studied in RKO cells. RESULTS: 375 mg/kg but not 37.5 mg/kg TQ decreased the number of large polyps in the small intestine of APCMin mice. TQ induced apoptosis in the neoplastic tissue but not in the normal mucosa. Furthermore, upon TQ treatment, ß-catenin was retained at the membrane and c-myc decreased in the nucleus, which was associated with a reduced cell proliferation in the villi. In vitro, TQ activated GSK-3ß, which induced membranous localization of ß-catenin and reduced nuclear c-myc expression. CONCLUSIONS: In summary, TQ interferes with polyp progression in ApcMin mice through induction of tumor-cell specific apoptosis and by modulating Wnt signaling through activation of GSK-3ß. Nigella sativa oil (or TQ) might be useful as nutritional supplement to complement surgery and chemoprevention in FAP.


Adenomatous Polyposis Coli/drug therapy , Anticarcinogenic Agents/pharmacology , Benzoquinones/pharmacology , Colorectal Neoplasms/prevention & control , Wnt Signaling Pathway , Adenomatous Polyposis Coli/metabolism , Adenomatous Polyposis Coli/pathology , Animals , Anticarcinogenic Agents/therapeutic use , Apoptosis , Benzoquinones/therapeutic use , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Male , Mice , Mice, Mutant Strains , Plant Oils/chemistry , beta Catenin/metabolism
6.
Biochem Pharmacol ; 85(2): 234-44, 2013 Jan 15.
Article En | MEDLINE | ID: mdl-23146664

Mesalamine (5-ASA) is widely used for the treatment of ulcerative colitis, a remitting condition characterized by chronic inflammation of the colon. Knowledge about the molecular and cellular targets of 5-ASA is limited and a clear understanding of its activity in intestinal homeostasis and interference with neoplastic progression is lacking. We sought to identify molecular pathways interfered by 5-ASA, using CRC cell lines with different genetic background. Microarray was performed for gene expression profile of 5-ASA-treated and untreated cells (HCT116 and HT29). Filtering and analysis of data identified three oncogenic pathways interfered by 5-ASA: MAPK/ERK pathway, cell adhesion and ß-catenin/Wnt signaling. PAK1 emerged as a consensus target of 5-ASA, orchestrating these pathways. We further investigated the effect of 5-ASA on cell adhesion. 5-ASA increased cell adhesion which was measured by cell adhesion assay and transcellular-resistance measurement. Moreover, 5-ASA treatment restored membranous expression of adhesion molecules E-cadherin and ß-catenin. Role of PAK1 as a mediator of mesalamine activity was validated in vitro and in vivo. Inhibition of PAK1 by RNA interference also increased cell adhesion. PAK1 expression was elevated in APC(min) polyps and 5-ASA treatment reduced its expression. Our data demonstrates novel pharmacological mechanism of mesalamine in modulation of cell adhesion and role of PAK1 in APC(min) polyposis. We propose that inhibition of PAK1 expression by 5-ASA can impede with neoplastic progression in colorectal carcinogenesis. The mechanism of PAK1 inhibition and induction of membranous translocation of adhesion proteins by 5-ASA might be independent of its known anti-inflammatory action.


Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anticarcinogenic Agents/pharmacology , Down-Regulation/drug effects , Mesalamine/pharmacology , p21-Activated Kinases/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Anticarcinogenic Agents/therapeutic use , Cadherins/genetics , Cadherins/metabolism , Cell Adhesion/drug effects , Cell Line, Tumor , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Colonic Neoplasms/prevention & control , Female , Gene Expression Profiling , Humans , Intestinal Polyps/metabolism , Intestinal Polyps/pathology , Intestinal Polyps/prevention & control , MAP Kinase Signaling System/drug effects , Male , Mesalamine/therapeutic use , Mice , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , RNA Interference , Wnt Signaling Pathway/drug effects , beta Catenin/genetics , beta Catenin/metabolism , p21-Activated Kinases/antagonists & inhibitors , p21-Activated Kinases/genetics
7.
PLoS One ; 7(11): e50541, 2012.
Article En | MEDLINE | ID: mdl-23209772

BACKGROUND/AIM: Elevated microsatellite instability at selected tetranucleotide repeats (EMAST) is a genetic signature in certain cases of sporadic colorectal cancer and has been linked to MSH3-deficiency. It is currently controversial whether EMAST is associated with oncogenic properties in humans, specifically as cancer development in Msh3-deficient mice is not enhanced. However, a mutator phenotype is different between species as the genetic positions of repetitive sequences are not conserved. Here we studied the molecular effects of human MSH3-deficiency. METHODS: HCT116 and HCT116+chr3 (both MSH3-deficient) and primary human colon epithelial cells (HCEC, MSH3-wildtype) were stably transfected with an EGFP-based reporter plasmid for the detection of frameshift mutations within an [AAAG]17 repeat. MSH3 was silenced by shRNA and changes in protein expression were analyzed by shotgun proteomics. Colony forming assay was used to determine oncogenic transformation and double strand breaks (DSBs) were assessed by Comet assay. RESULTS: Despite differential MLH1 expression, both HCT116 and HCT116+chr3 cells displayed comparable high mutation rates (about 4×10(-4)) at [AAAG]17 repeats. Silencing of MSH3 in HCECs leads to a remarkable increased frameshift mutations in [AAAG]17 repeats whereas [CA]13 repeats were less affected. Upon MSH3-silencing, significant changes in the expression of 202 proteins were detected. Pathway analysis revealed overexpression of proteins involved in double strand break repair (MRE11 and RAD50), apoptosis, L1 recycling, and repression of proteins involved in metabolism, tRNA aminoacylation, and gene expression. MSH3-silencing did not induce oncogenic transformation and DSBs increased 2-fold. CONCLUSIONS: MSH3-deficiency in human colon epithelial cells results in EMAST, formation of DSBs and significant changes of the proteome but lacks oncogenic transformation. Thus, MSH3-deficiency alone is unlikely to drive human colon carcinogenesis.


Colon/metabolism , Colon/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA-Binding Proteins/deficiency , Epithelial Cells/metabolism , Microsatellite Instability , Microsatellite Repeats/genetics , Cell Line , Colon/cytology , DNA Breaks, Double-Stranded , DNA-Binding Proteins/metabolism , Epithelial Cells/pathology , Humans , Male , MutS Homolog 3 Protein
8.
Int J Oncol ; 40(5): 1331-8, 2012 May.
Article En | MEDLINE | ID: mdl-22366868

Epidemiological evidence on the chemopreventive activity of mesalazine against colitis-associated cancer has accumulated in recent years. Together with the variety of mesalazine molecular antitumor effects this has prompted the development of novel mesalazine derivatives. The objective of this study was to test five novel derivatives (compounds 2-14, 2-17, 2-28, 2-34L, 2-39) for their effect on cell proliferation, their capability to scavenge superoxide anions, to induce a cell cycle arrest and to improve replication fidelity in cultured colorectal cells. Compound 2-14 was identified as the strongest inhibitor of cell proliferation and functioned as a potent superoxide scavenger, as did 2-17 and 2-34L. 2-14 induced a G2/M-arrest in HCT116 and a G0/G1-arrest in HT29 cells. 2-17 caused a G0/G1-arrest and 2-34L a G2/M-arrest in HT29 cells. 2-17 and 2-34L reduced mutation rates at a (CA)13 repeat in a dose-dependent fashion. These data suggest that certain mesalazine derivatives share important antitumor effects. From this experimental profile compounds 2-17 and 2-34L both improve replication fidelity, which is biologically relevant not only for colitis-associated cancer but also potentially for individuals with hereditary non-polyposis colorectal cancer.


Anticarcinogenic Agents/pharmacology , Colorectal Neoplasms/prevention & control , DNA Replication/drug effects , Mesalamine/pharmacology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Checkpoints/drug effects , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Checkpoint Kinase 1 , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Dose-Response Relationship, Drug , Free Radical Scavengers/pharmacology , HCT116 Cells , HT29 Cells , Humans , MutL Protein Homolog 1 , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oxidative Stress/drug effects , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Superoxides/metabolism , Transfection , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
9.
Best Pract Res Clin Gastroenterol ; 25(4-5): 535-46, 2011 Aug.
Article En | MEDLINE | ID: mdl-22122769

Aminosalicylates are the most common drugs for the primary treatment of inflammatory bowel disease. Various pro-drugs and formulations were developed in order to improve pharmacological profiles, optimize bioavailability and to gain highest efficacy in the treatment of ulcerative colitis (UC) and Crohn's disease. In vitro studies have greatly contributed to the understanding of the molecular actions in vivo and clinical studies have proven aminosalicylates to be effective and safe. This review summarizes the current knowledge on the molecular, pharmacological and clinical properties of aminosalicylates with respect to chemoprevention for UC-associated colorectal cancer.


Aminosalicylic Acids/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Anticarcinogenic Agents/therapeutic use , Colitis, Ulcerative/drug therapy , Colorectal Neoplasms/prevention & control , Crohn Disease/drug therapy , Gastrointestinal Agents/therapeutic use , Precancerous Conditions/drug therapy , Aminosalicylic Acids/pharmacokinetics , Animals , Anti-Inflammatory Agents/pharmacokinetics , Anticarcinogenic Agents/pharmacokinetics , Chemoprevention , Colitis, Ulcerative/pathology , Colorectal Neoplasms/pathology , Gastrointestinal Agents/pharmacokinetics , Humans , Precancerous Conditions/pathology , Treatment Outcome
10.
Hum Mol Genet ; 19(13): 2648-57, 2010 Jul 01.
Article En | MEDLINE | ID: mdl-20421367

Microsatellite instability is a key mechanism of colon carcinogenesis. We have previously studied mutations within a (CA)13 microsatellite using an enhanced green fluorescent protein (EGFP)-based reporter assay that allows the distinction of replication errors and mismatch repair (MMR) activity. Here we utilize this assay to compare mutations of mono- and dinucleotide repeats in human colorectal cells. HCT116 and HCT116+chr3 cells were stably transfected with EGFP-based plasmids harboring A10, G10, G16, (CA)13 and (CA)26 repeats. EGFP-positive mutant fractions were quantitated by flow cytometry, mutation rates were calculated and the mutant spectrum was analyzed by cycle sequencing. EGFP fluorescence pattern changed with the microsatellite's nucleotide sequence and cell type and clonal variations were observed in mononucleotide repeats. Replication errors (as calculated in HCT116) at A10 repeats were 5-10-fold higher than in G10, G16 were 30-fold higher than G10 and (CA)26 were 10-fold higher than (CA)13. The mutation rates in hMLH1-proficient HCT116+chr3 were 30-230-fold lower than in HCT116. MMR was more efficient in G16 than in A10 clones leading to a higher stability of poly-G tracts. Mutation spectra revealed predominantly 1-unit deletions in A10, (CA)13 and G10 and 2-unit deletions or 1-unit insertion in (CA)26. These findings indicate that both replication fidelity and MMR are affected by the microsatellite's nucleotide composition.


Colorectal Neoplasms/genetics , DNA Mismatch Repair , Dinucleotide Repeats , Microsatellite Repeats/genetics , Mutation , Base Sequence , Chromosomes, Human, Pair 3 , Colorectal Neoplasms/metabolism , DNA Replication/genetics , Flow Cytometry , Green Fluorescent Proteins , HCT116 Cells , Humans , Sequence Deletion , Tumor Cells, Cultured
11.
Clin Cancer Res ; 16(6): 1950-6, 2010 Mar 15.
Article En | MEDLINE | ID: mdl-20197483

PURPOSE: Mesalazine (5-aminosalicylic acid, 5-ASA) has chemopreventive properties in colitis-associated cancer. In vitro, it improves replication fidelity at (CA)13 microsatellites independent of mismatch repair proficiency. Therefore, 5-ASA might be advantageous in patients with hereditary nonpolyposis colorectal cancer. At this point, however, it is uncertain whether this improvement of replication fidelity is specific for (CA)13 repetitive sequences. Here, we tested the effect of 5-ASA on replication fidelity in mononucleotide, dinucleotide, and tetranucleotide repeats. EXPERIMENTAL DESIGN: HCT116 and HCT116+chr3 cells were transfected with pIREShyg2-EGFP reporter plasmids harboring the following microsatellites: A10, G10, (CA)13, (CA)26, (AAAG)17, poly-A tracts, and their flanking sequences of transforming growth factor beta receptor II (TGFBR2; A10) and activin type II receptor (ACVR2; A8). Stably transfected single-cell clones were selected, characterized by Southern blotting, sorted into six-well plates, and cultured with or without 5-ASA. Frameshift mutations that shift the enhanced green fluorescence protein into its proper reading frame were quantified by flow cytometry. RESULTS: In HCT116, 5-ASA reduced the mutant fraction at (CA)13 by 48.3%, at A10 by 35.6-43.6%, at G10 by 74.9-83.6%, and at (AAAG)17 by 37.6-44.4%. Similar results were observed in hMLH1-proficient HCT116+chr3 cells. Moreover, the presence of 5-ASA significantly reduced mutations in TGFBR2 (A10) and ACVR2 (A8) by 39.9% and 46.2%, respectively. CONCLUSIONS: 5-ASA increases replication fidelity in mononucleotide, dinucleotide, and tetranucleotide repeats and reduces mutations in tumor suppressor genes TGFBR2 and ACVR2, a finding that may provoke in vivo studies for the prevention of colorectal cancer in hereditary nonpolyposis colorectal cancer.


Activin Receptors, Type II/genetics , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Colorectal Neoplasms/genetics , DNA Replication/drug effects , Frameshift Mutation/genetics , Mesalamine/pharmacology , Microsatellite Repeats/genetics , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , Blotting, Southern , Colorectal Neoplasms/pathology , Colorectal Neoplasms/prevention & control , Flow Cytometry , Humans , Receptor, Transforming Growth Factor-beta Type II , Tumor Cells, Cultured
12.
Inflamm Bowel Dis ; 16(4): 576-82, 2010 Apr.
Article En | MEDLINE | ID: mdl-19821510

BACKGROUND: Individuals with ulcerative colitis are at high risk of developing colitis-associated cancer. 5-Aminosalicylate (5-ASA) protects from cancer by its antiinflammatory activity as well as by altering cell growth, inducing apoptosis, and reducing replication errors. So far neither 5-ASA's structural specificity nor its pharmacophore group have been identified. Here we compared 5-ASA with its analogs (4-ASA and 3-ASA) and its metabolite N-acetyl-5-ASA (NAc-5-ASA). METHODS: Superoxide scavenging was analyzed by lucigenin-amplified chemiluminescence. Cell growth, cell cycle distribution, and replication fidelity at a (CA)13 microsatellite were measured in HCT116 and HT29 colon epithelial cells by MTT and flow cytometry. Nuclear protein extracts were blotted for replication protein A (RPA), claspin, p53, and p53(Ser15). RESULTS: All compounds inhibited the growth of colon epithelial cells at a similar level and displayed potent scavenging properties, with 3-ASA being the most active, followed by 5-ASA, 4-ASA, and NAc-5-ASA. Besides 5-ASA, only 4-ASA caused an increase in the S-phase population (56%-69% and 49%-62% in HCT116 and HT29 cells, respectively). This was accompanied by nuclear recruitment of replication proteins RPA and claspin as well as phosphorylation of p53(Ser15), both of which were weaker or absent with 3-ASA or NAc-5-ASA. 5-ASA was the only compound that lowered mutations at a (CA)13 microsatellite. CONCLUSIONS: 5-ASA shares its growth inhibitory and superoxide scavenging properties with its structural analogs and metabolite, but the position of the amino group is critical for reducing replication errors.


Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Benzene/chemistry , DNA Replication/drug effects , Mesalamine/pharmacology , Replication Protein A/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Blotting, Western , Cell Cycle/drug effects , Cell Proliferation/drug effects , Flow Cytometry , HT29 Cells/drug effects , Humans , Microsatellite Repeats , Neutrophils/metabolism , Superoxides/metabolism , Tumor Suppressor Protein p53/metabolism
13.
Carcinogenesis ; 28(10): 2207-17, 2007 Oct.
Article En | MEDLINE | ID: mdl-17510082

Colorectal cancer (CRC) is the most common gastrointestinal malignancy. Most of the clinical data on CRC prevention have come from the use of aspirin. Besides inhibition of cyclooxygenases, aspirin has a diversity of molecular effects that counteract colon carcinogenesis. Aspirin restrains cell proliferation by inducing a G1 arrest in colorectal cells. To determine which cell cycle checkpoint pathways are involved in this response, colorectal cell lines wild-type or defective for p53 and p21Waf1/Cip1 were treated with aspirin or the anti-proliferative drug sulindac sulfide, then assayed for proliferative activity, for cell cycle progression and apoptosis, for the activation and phosphorylation of checkpoint components and for the transcriptional up-regulation of p21Waf1/Cip1 and Bax. Aspirin and sulindac sulfide induced a G1 arrest within 48 h. While all cell lines responded in a comparable way to sulindac sulfide, the aspirin-induced G1 arrest was dependent on p21Waf1/Cip1--as cells lacking the cyclin-dependent kinase inhibitor failed to show this arrest--and on ataxia-telangiectasia-mutated kinase (ATM)--as the inhibitor caffeine abrogated the checkpoint. Moreover, aspirin induced cell death mainly in cells expressing p53. Aspirin induced the phosphorylation of p53 at residue Ser15 within 8 h in a caffeine-dependent manner, and also caused the activation of checkpoint kinase 2 and the cleavage of caspase 7. Our results suggest that aspirin induces a G1 arrest and apoptosis by activating p53 and p21Waf1/Cip1 in an ATM-dependent way. By activating these checkpoint pathways, aspirin may restrain uncontrolled proliferation of colorectal cells, enhance their response to stresses such as DNA damage and promote entry of abnormal cells into apoptosis.


Apoptosis/drug effects , Aspirin/pharmacology , Cell Cycle Proteins/metabolism , Cell Division/drug effects , Colonic Neoplasms/pathology , DNA-Binding Proteins/metabolism , Enzyme Activation/drug effects , G1 Phase/drug effects , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/genetics , Cell Line, Tumor , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/genetics , Flow Cytometry , Humans , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/drug effects , Tumor Suppressor Proteins/genetics
14.
Gastroenterology ; 132(1): 221-35, 2007 Jan.
Article En | MEDLINE | ID: mdl-17241873

BACKGROUND & AIMS: Individuals with inflammatory bowel disease are at risk of developing colorectal cancer (CRC). Epidemiologic, animal, and laboratory studies suggest that 5-amino-salicylic acid (5-ASA) protects from the development of CRC by altering cell cycle progression and by inducing apoptosis. Our previous results indicate that 5-ASA improves replication fidelity in colorectal cells, an effect that is active in reducing mutations. In this study, we hypothesized that 5-ASA restrains cell cycle progression by activating checkpoint pathways in colorectal cell lines, which would prevent tumor development and improve genomic stability. METHODS: CRC cells with different genetic backgrounds such as HT29, HCT116, HCT116(p53-/-), HCT116+chr3, and LoVo were treated with 5-ASA for 2-96 hours. Cell cycle progression, phosphorylation, and DNA binding of cell cycle checkpoint proteins were analyzed. RESULTS: We found that 5-ASA at concentrations between 10 and 40 mmol/L affects cell cycle progression by inducing cells to accumulate in the S phase. This effect was independent of the hMLH1, hMSH2, and p53 status because it was observed to a similar extent in all cell lines under investigation. Moreover, wash-out experiments demonstrated reversibility within 48 hours. Although p53 did not have a causative role, p53 Ser15 was strongly phosphorylated. Proteins involved in the ATM-and-Rad3-related kinase (ATR)-dependent S-phase checkpoint response (Chk1 and Rad17) were also phosphorylated but not ataxia telengectasia mutated kinase. CONCLUSIONS: Our data demonstrate that 5-ASA causes cells to reversibly accumulate in S phase and activate an ATR-dependent checkpoint. The activation of replication checkpoint may slow down DNA replication and improve DNA replication fidelity, which increases the maintenance of genomic stability and counteracts carcinogenesis.


Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Colorectal Neoplasms/prevention & control , DNA Replication/drug effects , Mesalamine/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis/drug effects , Ataxia Telangiectasia Mutated Proteins , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Cell Division/drug effects , Checkpoint Kinase 1 , Colorectal Neoplasms/pathology , DNA Mismatch Repair/drug effects , DNA Polymerase III/metabolism , DNA Replication/physiology , DNA-Binding Proteins/metabolism , Flow Cytometry , HT29 Cells , Humans , Minichromosome Maintenance Complex Component 2 , Minichromosome Maintenance Complex Component 7 , Mitosis/drug effects , MutL Protein Homolog 1 , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , S Phase/drug effects , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
...