Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
1.
Chem Sci ; 6(1): 804-815, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25642316

ABSTRACT

Aiming towards the development of novel nootropic therapeutics to address the cognitive impairment common to a range of brain disorders, we set out to develop highly selective small molecule inhibitors of HDAC2, a chromatin modifying histone deacetylase implicated in memory formation and synaptic plasticity. Novel ortho-aminoanilide inhibitors were designed and evaluated for their ability to selectively inhibit HDAC2 versus the other Class I HDACs. Kinetic and thermodynamic binding properties were essential elements of our design strategy and two novel classes of ortho-aminoanilides, that exhibit kinetic selectivity (biased residence time) for HDAC2 versus the highly homologous isoform HDAC1, were identified. These kinetically selective HDAC2 inhibitors (BRD6688 and BRD4884) increased H4K12 and H3K9 histone acetylation in primary mouse neuronal cell culture assays, in the hippocampus of CK-p25 mice, a model of neurodegenerative disease, and rescued the associated memory deficits of these mice in a cognition behavioural model. These studies demonstrate for the first time that selective pharmacological inhibition of HDAC2 is feasible and that inhibition of the catalytic activity of this enzyme may serve as a therapeutic approach towards enhancing the learning and memory processes that are affected in many neurological and psychiatric disorders.

2.
Oncogene ; 34(12): 1499-509, 2015 Mar 19.
Article in English | MEDLINE | ID: mdl-24704829

ABSTRACT

The cyclic nucleotide phosphodiesterase 10A (PDE10) has been mostly studied as a therapeutic target for certain psychiatric and neurological conditions, although a potential role in tumorigenesis has not been reported. Here we show that PDE10 is elevated in human colon tumor cell lines compared with normal colonocytes, as well as in colon tumors from human clinical specimens and intestinal tumors from Apc(Min/+) mice compared with normal intestinal mucosa, respectively. An isozyme and tumor-selective role of PDE10 were evident by the ability of small-molecule inhibitors and small interfering RNA knockdown to suppress colon tumor cell growth with reduced sensitivity of normal colonocytes. Stable knockdown of PDE10 by short hairpin RNA also inhibits colony formation and increases doubling time of colon tumor cells. PDE10 inhibition selectively activates cGMP/cGMP-dependent protein kinase signaling to suppress ß-catenin levels and T-cell factor (TCF) transcriptional activity in colon tumor cells. Conversely, ectopic expression of PDE10 in normal and precancerous colonocytes increases proliferation and activates TCF transcriptional activity. These observations suggest a novel role of PDE10 in colon tumorigenesis and that inhibitors may be useful for the treatment or prevention of colorectal cancer.


Subject(s)
Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/metabolism , TCF Transcription Factors/genetics , beta Catenin/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HCT116 Cells , HEK293 Cells , HT29 Cells , Humans , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , TCF Transcription Factors/metabolism , Transcription, Genetic , beta Catenin/metabolism
3.
Cell Death Dis ; 4: e933, 2013 Nov 28.
Article in English | MEDLINE | ID: mdl-24287694

ABSTRACT

Endoplasmic reticulum (ER) is a central organelle in eukaryotic cells that regulates protein synthesis and maturation. Perturbation of ER functions leads to ER stress, which has been previously associated with a broad variety of diseases. ER stress is generally regarded as compensatory, but prolonged ER stress has been involved in apoptosis induced by several cytotoxic agents. Choline kinase α (ChoKα), the first enzyme in the Kennedy pathway, is responsible for the generation of phosphorylcholine (PCho) that ultimately renders phosphatidylcholine. ChoKα overexpression and high PCho levels have been detected in several cancer types. Inhibition of ChoKα has demonstrated antiproliferative and antitumor properties; however, the mechanisms underlying these activities remain poorly understood. Here, we demonstrate that ChoKα inhibitors (ChoKIs), MN58b and RSM932A, induce cell death in cancer cells (T47D, MCF7, MDA-MB231, SW620 and H460), through the prolonged activation of ER stress response. Evidence of ChoKIs-induced ER stress includes enhanced production of glucose-regulated protein, 78 kDa (GRP78), protein disulfide isomerase, IRE1α, CHOP, CCAAT/enhancer-binding protein beta (C/EBPß) and TRB3. Although partial reduction of ChoKα levels by small interfering RNA was not sufficient to increase the production of ER stress proteins, silencing of ChoKα levels also show a decrease in CHOP overproduction induced by ChoKIs, which suggests that ER stress induction is due to a change in ChoKα protein folding after binding to ChoKIs. Silencing of CHOP expression leads to a reduction in C/EBPß, ATF3 and GRP78 protein levels and abrogates apoptosis in tumor cells after treatment with ChoKIs, suggesting that CHOP maintains ER stress responses and triggers the pro-apoptotic signal. Consistent with the differential effect of ChoKIs in cancer and primary cells previously described, ChoKIs only promoted a transient and moderated ER stress response in the non-tumorogenic cells MCF10A. In conclusion, pharmacological inhibition of ChoKα induces cancer cell death through a mechanism that involves the activation of exaggerated and persistent ER stress supported by CHOP overproduction.


Subject(s)
Choline Kinase/metabolism , Endoplasmic Reticulum Stress/physiology , Transcription Factor CHOP/metabolism , Apoptosis/genetics , Apoptosis/physiology , Cell Line, Tumor , Cell Survival/genetics , Cell Survival/physiology , Choline Kinase/genetics , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/genetics , Flow Cytometry , Fluorescent Antibody Technique , Humans , Phosphorylcholine/metabolism , Transcription Factor CHOP/genetics
4.
Toxicol In Vitro ; 27(6): 1626-33, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23603476

ABSTRACT

Piperlongumine (PPLGM) is a bioactive compound isolated from long peppers that shows selective toxicity towards a variety of cancer cell types including colon cancer. The signaling pathways that lead to cancer cell death in response to PPLGM exposure have not been previously identified. Our objective was to identify the intracellular signaling mechanisms by which PPLGM leads to enhanced colon cancer cell death. We found that PPLGM inhibited the growth of colon cancer cells in time- and concentration-dependent manners, but was not toxic toward normal colon mucosal cells at concentrations below 10 µM. Acute (0-60 min) and prolonged (24h) exposure of HT-29 cells to PPLGM resulted in phosphorylation of ERK. To investigate whether ERK signaling was involved in PPLGM-mediated cell death, we treated HT-29 cells with the MEK inhibitor U0126, prior to treating with PPLGM. We found that U0126 attenuated PPLGM-induced activation of ERK and partially protected against PPLGM-induced cell death. These results suggest that PPLGM works, at least in part, through the MEK/ERK pathway to result in colon cancer cell death. A more thorough understanding of the molecular mechanisms by which PPLGM induces colon cancer cell death will be useful in developing therapeutic strategies to treat colon cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Colonic Neoplasms/metabolism , Dioxolanes/pharmacology , MAP Kinase Signaling System/drug effects , Apoptosis/drug effects , Cell Line , Colon/cytology , Extracellular Signal-Regulated MAP Kinases/metabolism , HCT116 Cells , HT29 Cells , Humans , Intestinal Mucosa/cytology , Signal Transduction/drug effects
5.
Oncogene ; 32(8): 939-46, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23160372

ABSTRACT

The human protein methyltransferases (PMTs) constitute a large enzyme class composed of two families, the protein lysine methyltransferases (PKMTs) and the protein arginine methyltransferases (PRMTs). Examples have been reported of both PKMTs and PRMTs that are genetically altered in specific human cancers, and in several cases these alterations have been demonstrated to confer a unique dependence of the cancer cells on PMT enzymatic activity for the tumorigenic phenotype. Examples of such driver alterations in PMTs will be presented together with a review of current efforts towards the discovery and development of small-molecule inhibitors of these enzymes as personalized cancer therapeutics.


Subject(s)
Neoplasms/drug therapy , Neoplasms/enzymology , Precision Medicine/methods , Protein Methyltransferases/antagonists & inhibitors , Protein Methyltransferases/genetics , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Drug Discovery , Enzyme Inhibitors/pharmacology , Epigenomics , Humans , Molecular Targeted Therapy/methods , Neoplasms/genetics , Protein Methyltransferases/metabolism
6.
Br J Cancer ; 103(7): 975-86, 2010 Sep 28.
Article in English | MEDLINE | ID: mdl-20717114

ABSTRACT

BACKGROUND: Cyclooxygenase-2 (COX-2) overexpression is strongly associated with colorectal tumourigenesis. It has been demonstrated that the chronic use of non-steroidal anti-inflammatory drugs (COX inhibitors) partially protects patients from colorectal cancer (CRC) development and progression but induces severe cardiovascular side effects. New strategies for selective COX-2 blockade are required. METHODS: We developed an improved technique, based on RNA interference (RNAi), to gain a selective COX-2 silencing in CRC cells by a tumour-dependent expression of anti-COX-2 short-hairpin RNA (shCOX-2). Anti-COX-2 shRNA-expressing vectors were delivered in CRC cells (in vitro) and in colon tissues (ex vivo) using engineered Escherichia coli strains, capable of invading tumour cells (InvColi). RESULTS: A highly tumour-dependent shCOX-2 expression and a significant COX-2 silencing were observed in CRC cells following InvColi strain infection. Cyclooxygenase-2 silencing was associated with a strong reduction in both proliferative and invasive behaviour of tumour cells. We also demonstrated a pivotal role of COX-2 overexpression for the survival of CRC cells after bacterial infection. Moreover, COX-2 silencing was achieved ex vivo by infecting colon tissue samples with InvColi strains, leading to anti-inflammatory and anti-tumour effects. CONCLUSION: Our RNAi/InvColi-mediated approach offers a promising tool for a highly selective COX-2 blockade in vitro and in vivo.


Subject(s)
Colonic Neoplasms/genetics , Cyclooxygenase 2/genetics , Escherichia coli/genetics , RNA Interference , Cell Line, Tumor , Cell Proliferation , Cell Survival , Colonic Neoplasms/enzymology , Dinoprostone/biosynthesis , Humans , Transfection , Up-Regulation
7.
Oncogene ; 28(45): 3983-96, 2009 Nov 12.
Article in English | MEDLINE | ID: mdl-19734940

ABSTRACT

An elevated proteasome activity contributes to tumorigenesis, particularly by providing cancer cells with antiapoptotic protection and efficient clearance from irregular proteins. Still, the underlying mechanisms are poorly known. In this study, we report that in colon cancer patients, higher proteasome activity was detected in tumoral tissue compared with surrounding normal tissue, and also that increased levels of proteasomal subunit proteins, such as S5a/PSMD4 and alpha-5/PSMA5, could be detected. Colon tumors showed higher nuclear levels of nuclear factor E2-related factor 2 (Nrf2), a transcription factor supposed to be involved in the control of proteasomal subunit protein expression. The induction or overexpression of Nrf2 led to stronger S5a and alpha-5 expression in the human colon cancer cell lines, Colo320 and Lovo, as well as in NCM460 colonocytes along with higher proteasome activity. The small interfering RNA (siRNA)-mediated Nrf2 knockdown decreased S5a and alpha-5 expression and reduced proteasome activity. Additionally, Nrf2-dependent S5a and alpha-5 expression conferred protection from tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis, an effect preceded by an increased nuclear factor (NF)-kappaB activation and higher expression of antiapoptotic NF-kappaB target genes. These findings point to an important role of Nrf2 in the gain of proteasome activity, thereby contributing to colorectal carcinogenesis. Nrf2 may therefore serve as a potential target in anticancer therapy.


Subject(s)
Colorectal Neoplasms/metabolism , NF-E2-Related Factor 2/metabolism , Proteasome Endopeptidase Complex/metabolism , Blotting, Western , Cell Line, Tumor , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Epithelial Cells/cytology , Humans , Immunohistochemistry , NF-E2-Related Factor 2/genetics , Oxidative Stress , Proteasome Endopeptidase Complex/biosynthesis , Proteasome Endopeptidase Complex/genetics , Signal Transduction , Ubiquitination
8.
Water Sci Technol ; 56(12): 51-5, 2007.
Article in English | MEDLINE | ID: mdl-18075178

ABSTRACT

Research was initiated to develop an in vitro system to identify disinfection by-products with a potential to transform normal human colonocytes into malignant cells. Tribromomethane and bromochloroacetic acid, rodent colon carcinogens, dibromonitromethane and tribromonitromethane, recently identified in drinking water, and azoxymethane, a classic colon carcinogen, were tested for the ability to transform NCM460 cells. The chronic toxicity was determined for the series of trihalomethanes, haloacetic acids and halonitromethanes as well as NCM460 cell enzymatic capabilities. The order of cytotoxicity was halonitromethanes > haloacetic acids > trihalomethanes. Cytotoxicity within a series increased with the degree of bromination and decreased with the molecular weight. The genotoxicity profile was similar to that for cytotoxicity. Enzymatic analysis demonstrated that NCM460 cells possess glutathione-S transerase-1-1 and CYP450 activity similar to that measured in the large intestine. NCM460 cells were exposed to 10(-6) M of the test chemicals for three days. While NCM460 cells from all treatments had the ability to grow in soft agar to some extent, only cells exposed to azoxymethane or tribromomethane were able to grow in media lacking serum and growth factors. When sub cultured, NCM460 cells exposed to 10(-9) M azoxymethane for three weeks formed colonies with morphology distinct from untreated cells.


Subject(s)
Carcinogens/pharmacology , Cell Transformation, Neoplastic/drug effects , Colon/cytology , Acetates/pharmacology , Azoxymethane/pharmacology , Cell Survival/drug effects , Cells, Cultured , Colon/enzymology , Cytochrome P-450 Enzyme System/metabolism , Glutathione Transferase/metabolism , Humans , Hydrocarbons, Brominated/pharmacology , Inhibitory Concentration 50 , Time Factors , Trihalomethanes/pharmacology
9.
J Pathol ; 212(2): 124-33, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17427169

ABSTRACT

There is increasing evidence for an alternative pathway of sporadic colorectal tumourigenesis that is associated with DNA microsatellite instability (MSI), due to methylation and loss of expression of the mismatch repair gene MLH1. Recent studies have highlighted a serrated pathway of colorectal cancer (CRC) in which serrated polyps with activating mutations in BRAF progress to CRCs with MSI following methylation and silencing of MLH1. The present study provides a novel mechanistic experimental model for these clinical observations. We investigated the role of BRAF activating mutation (BRAF-V600E) in colorectal tumourigenesis by studying the effects of forced expression of BRAF-V600E in the 'normal' colon epithelial NCM460 cell line and by targeting endogenous BRAF-V600E in MSI-High (MSI-H) colon cancer cell lines. The findings indicate that BRAF mutation in colon epithelial cells contributes to a gain in resistance towards apoptotic stimuli, which is likely to be an important characteristic of pre-malignant serrated lesions. BRAF-V600E also plays a role in the development and maintenance of transformed and invasive phenotypes in colon epithelial cells. Our findings also suggest that BRAF mutation potentiates promoter hypermethylation of the MLH1 gene promoter. Together, these results highlight BRAF as a potential target for therapeutic intervention in sporadic MSI-H colorectal cancers.


Subject(s)
Colorectal Neoplasms/genetics , Neoplasm Proteins/genetics , Proto-Oncogene Proteins B-raf/genetics , Adaptor Proteins, Signal Transducing/genetics , Apoptosis/genetics , Cell Line , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Colon/pathology , Colorectal Neoplasms/pathology , CpG Islands/genetics , Epithelial Cells/pathology , Humans , Intestinal Mucosa/pathology , Methylation , Microsatellite Instability , Models, Biological , MutL Protein Homolog 1 , Mutation , Neoplasm Invasiveness , Nuclear Proteins/genetics , Phenotype , Promoter Regions, Genetic/genetics
10.
Br J Cancer ; 86(6): 983-8, 2002 Mar 18.
Article in English | MEDLINE | ID: mdl-11953833

ABSTRACT

INCELL AAFA, an omega-3 polyunsaturated fatty acid product containing a high concentration of long chain fatty acids, was tested for its ability to ameliorate the harmful side effects of CPT-11 chemotherapy including: leukopenia, anaemia, asthenia, weight loss and liver involvement. Four groups of mice were fed an AIN-76 diet modified to contain: 10% w/w corn oil (CO), 0% AAFA; 9% CO, 1% AAFA; 8% CO, 2% AAFA; or 7% CO, 3% AAFA. After 2 weeks on the diets, half of the mice received CPT-11 chemotherapy (60 mg kg(-1) q 4 days, i.v.) the rest of the mice received vehicle for 2 weeks. It was found that 2% AAFA in the diet of the CPT-11 treated mice: decreased apoptotic figures in the duodenal crypts; markedly suppressed the inflammatory eicosanoid, prostaglandin E(2) in the liver; prevented liver hypertrophy; improved white blood cell counts; significantly increased red blood cell counts; decreased numbers of CPT-11 induced immature red blood cell and micronuclei in red blood cells of the peripheral blood; increased eicosapentaenoic acid and docosahexaenoic acid in liver cell membranes and maintained normal grooming behaviour. Thus 2% AAFA in the diet reduced the side effects of CPT-11 treatment in mice.


Subject(s)
Antineoplastic Agents, Phytogenic/toxicity , Camptothecin/analogs & derivatives , Camptothecin/toxicity , Fatty Acids, Omega-3/pharmacology , Animals , Apoptosis/drug effects , Dinoprostone/analysis , Duodenum/drug effects , Duodenum/pathology , Irinotecan , Liver/drug effects , Liver/pathology , Male , Membrane Lipids/analysis , Mice , Organ Size/drug effects
11.
J Biol Chem ; 276(48): 44464-71, 2001 Nov 30.
Article in English | MEDLINE | ID: mdl-11574537

ABSTRACT

Neurotensin (NT), a neuropeptide released in the gastrointestinal tract in response to several stimuli, is involved in the pathophysiology of colonic inflammation. However, the molecular mechanism(s) mediating this proinflammatory response remains unclear. We found that NCM460, non-transformed human colonocytes, express a functional high affinity NT receptor that mediates NT-induced Erk activation. By using NCM460 cells stably transfected with NTR1, we show that NTR1 activation leads to interleukin (IL)-8 secretion that is mediated via both NF-kappaB- and Erk-dependent pathways. In addition, NT-stimulated NF-kappaB activation is dependent on intracellular calcium release. NT-stimulated Erk activity requires Ras activation because overexpression of the dominant negative Ras mutant Ras-17N almost completely inhibits the Erk activation. Furthermore, NT directly stimulates Ras-GTP formation as shown by a Ras-GTP pull-down assay. By using reporter gene constructs containing targeted substitutions in the IL-8 promoter, we show that the NF-kappaB, AP-1, and to a lesser degree the C/EBP sites in the IL-8 promoter region are required for IL-8 gene expression induced by NT. In summary, our results demonstrate that NT stimulates calcium-dependent NF-kappaB and Ras-dependent Erk pathways that mediate the release of IL-8 from non-transformed human colonocytes. We speculate that these NT-related proinflammatory pathways are important in the pathophysiology of colonic inflammation.


Subject(s)
Colonic Neoplasms/metabolism , Interleukin-8/biosynthesis , Neurotensin/metabolism , Signal Transduction , Calcium/metabolism , Cell Line , Cell Line, Transformed , Cloning, Molecular , DNA/metabolism , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Inhibitors/pharmacology , Genes, Dominant/genetics , Genes, Reporter , Humans , Immunoglobulin G/metabolism , Inflammation , Interleukin-8/metabolism , Lac Operon , Ligands , Luciferases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Mutation , NF-kappa B/metabolism , Phosphorylation , Plasmids/metabolism , Promoter Regions, Genetic , Retroviridae/genetics , Time Factors , Transfection , Tumor Cells, Cultured , ras Proteins/metabolism
12.
Pancreas ; 23(2): 157-71, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11484918

ABSTRACT

It has been established that ductal cells or precursor cells within the ductal tree of the pancreas can differentiate into islet cells. Although islet cells can also form exocrine cells, it is unclear whether they arise from precursor (stem) cells or from mature endocrine cells by transdifferentiation. Using a defined culture medium and technique for islet purification, for the first time we were able to maintain human islets in culture for more than a year. Multilabeling immunohistochemical and immunoelectron microscopic examination of the islets at different days of culture using islet cell markers (antibodies to hormones, neuron-specific enolase, chromogranin A) and ductal cell markers (cytokeratins 7 and 19, carbonic anhydrase II, DU-PAN2, CA 19-9, and MUC1) revealed that endocrine cells gradually transdifferentiate to ductal, acinar, and intermediary cells. Although islet hormone secretion ceased after day 28 in culture, endocrine cells were still detectable at day 60. However, later, all endocrine and exocrine cells were replaced by undifferentiated cells that expressed neuron-specific enolase, chromogranin A, laminin, vimentin, cytokeratin 7 and 19, alpha-1-antitrypsin, transforming growth factor-alpha, and epidermal growth factor receptor. Our data thus show that, under proper conditions, human islets can be maintained in vitro over a long period and that, in the culture condition, islet cells seem to transdifferentiate to exocrine cells and undifferentiated cells, which may be considered pancreatic precursor (stem) cells.


Subject(s)
Islets of Langerhans/cytology , Biomarkers , Cell Differentiation , Cell Division , Cells, Cultured , Glucagon/genetics , Glucagon/metabolism , Humans , Immunohistochemistry , Insulin/genetics , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/physiology , Microscopy, Immunoelectron , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stem Cells/cytology , Stem Cells/physiology , Time Factors
13.
Am J Physiol Gastrointest Liver Physiol ; 281(1): G144-50, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11408266

ABSTRACT

Thiamine (vitamin B(1)) is essential for normal cellular functions and growth. Mammals cannot synthesize thiamine and thus must obtain the vitamin via intestinal absorption. The intestine is exposed to a dietary thiamine source and a bacterial source in which the vitamin is synthesized by the normal microflora of the large intestine. Very little is known about thiamine uptake in the large intestine. The aim of this study was, therefore, to address this issue. Our results with human-derived colonic epithelial NCM460 cells as a model system showed thiamine uptake to be 1) temperature- and energy dependent, 2) Na(+) independent, 3) increased with increasing buffer pH from 5 to 8 and after cell acidification but inhibited by amiloride, 4) saturable as a function of concentration, 5) inhibited by thiamine structural analogs but not by unrelated organic cations, and 6) inhibited by modulators of a Ca(2+)/calmodulin-mediated pathway. NCM460 cells and native human colonic mucosa expressed the recently cloned human thiamine transporter THTR-1 (product of the SLC19A2 gene) at both mRNA and protein levels. These results demonstrate for the first time that human NCM460 colonocytes possess a specific carrier-mediated system for thiamine uptake that appears to be under the regulation of an intracellular Ca(2+)/calmodulin-mediated pathway. It is suggested that bacterially synthesized thiamine in the large intestine may contribute to thiamine nutrition of the host, especially toward cellular nutrition of the local colonocytes.


Subject(s)
Colon/cytology , Intestinal Absorption/physiology , Intestinal Mucosa/metabolism , Membrane Transport Proteins , Thiamine/pharmacokinetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Gene Expression/physiology , Humans , Hydrogen-Ion Concentration , Intestinal Mucosa/cytology , Kinetics , RNA, Messenger/analysis , Sodium/pharmacology , Temperature , Tritium
14.
Cancer Lett ; 160(1): 37-43, 2000 Nov 10.
Article in English | MEDLINE | ID: mdl-11098082

ABSTRACT

To identify genes which are differentially transcribed in colorectal tumor cells, we compared the two human tumor cell lines, SW480 and HCT116, with the cell line, NCM460, from normal colon epithelium as a control. Using the methods of differential display reverse transcription PCR and Northern blot hybridization, we detected the differential transcription of seven genes: cholecystokinin, reticulocalbin, Rab5 guanine nucleotide exchange factor Rabex5, caldesmon, differentiation related gene 1 (drg1), taxol resistant associated gene 3 (Trag-3) and the gene for the placental protein, diff33. The yet unidentified cDNA of the human Rabex5 gene and the 3' untranslated region of the human caldesmon gene were cloned.


Subject(s)
Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , Amino Acid Sequence , Calcium-Binding Proteins/genetics , Calmodulin-Binding Proteins/genetics , Cell Cycle Proteins/genetics , Cholecystokinin/genetics , Colorectal Neoplasms/pathology , Guanine Nucleotide Exchange Factors/genetics , Humans , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins , Molecular Sequence Data , Neoplasm Proteins/genetics , Proteins/genetics , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Receptors, Cell Surface/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Transcription, Genetic , Tumor Cells, Cultured
15.
Pancreas ; 21(4): 358-68, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11075990

ABSTRACT

Cultivation and preservation of human pancreatic ductal cells have remained a challenge. With a defined culture medium and refinement of culturing techniques, we have been able to maintain human pancreatic ductal cells without any genetic manipulation in culture for more than 16 months. Freshly isolated ductal fragments were placed on a rocker in M3:5 medium free of collagen for 14 days to remove fibroblasts and endocrine cells before allowing them to attach. The cells produced an excessive amount of mucin and expressed the duct specific cytokeratins (CK) 7 and 19, DU-PAN2, CA19-9, carbonic anhydrase II (CA II), and secretin receptors. During the course of the culture, however, the cells gradually lost the expression of CA II, secretin receptors, DU-PAN2, and CA 19-9 and assumed an undifferentiated phenotype, which showed an upregulation of transforming growth factor alpha (TGFalpha) and epidermal growth factor receptor (EGFR), an increase in the expression of Ki-67, and an increased binding to Phaseolus vulgaris leucoagglutinin (PHA-L) and tomato lectin. These ductal cells present a useful source with which to study physiologic aspects of ductal cells including differentiation.


Subject(s)
Pancreatic Ducts/cytology , Adult , Cell Adhesion , Cell Cycle , Cell Division , Cells, Cultured , Epidermal Growth Factor/metabolism , Humans , Keratins/analysis , Male , Pancreatic Ducts/chemistry , Secretin/metabolism , Vimentin/analysis
16.
Mol Ther ; 2(5): 515-23, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11082325

ABSTRACT

A novel gap junction-independent mechanism for ganciclovir-mediated bystander effect killing by a herpes simplex virus thymidine kinase (HSV-TK)-expressing SW620 human colon tumor cell line has been characterized. The mechanism of the HSV-TK/GCV bystander effect for many tumor cell lines has been demonstrated to be due to connexin gap junction transfer of phosphorylated ganciclovir (GCV) metabolites; however, there may be as yet uncharacterized connexin-independent mechanisms for the effect. To address this, the bystander effect was further evaluated in a panel of cell lines mixed with homologous HSV-TK-expressing cell lines, a SW620.TK cell line, or a high connexin43-expressing PA-317.TK cell line. Of the 10 cell lines tested, 4 were found to be resistant to bystander effect killing by their homologous HSV-TK-expressing cell lines and the PA-317.TK cells, but all of the cell lines were sensitive to GCV killing when mixed with the SW620.TK cells. The SW620.TK cells were then further evaluated for any indication of extracellular GCV metabolite efflux. Culture medium from SW620.TK cells labeled with [(3)H]GCV was evaluated for the presence of GCV nucleotides by ion-exchange column separation and HPLC analysis. The presence of GCV mono-, di-, and triphosphate metabolites in the medium was detected. Inclusion in the medium of inhibitors of extracellular phosphatases and ecto-ATPases increased the proportion of GCV metabolites recovered. These results indicate that phosphorylated GCV metabolites can be effluxed from SW620.TK cells and that some type of cellular uptake mechanism independent of gap junctions exists for nucleotide entry into neighboring cells.


Subject(s)
Colonic Neoplasms/pathology , Ganciclovir/pharmacology , Simplexvirus/genetics , Thymidine Kinase/genetics , Cell Count , Cell Death , Colonic Neoplasms/metabolism , Colonic Neoplasms/virology , Connexin 43/metabolism , Ganciclovir/metabolism , Humans , Phosphorylation , Simplexvirus/enzymology , Tumor Cells, Cultured
17.
Article in English | MEDLINE | ID: mdl-10982956

ABSTRACT

OBJECTIVE: To develop a human oral epithelial cell line to constitute a continuous source of cells readily available for human oral epithelial cell research. STUDY DESIGN: Oral epithelial cells from a 30-week gestational, stillborn male fetus were grown in serum-free medium and transfected by lipid-mediation with the shuttle vector plasmid, pZ189, containing the T-antigen coding region and replication origin from the SV40 virus. RESULTS: Resulting cultures produced foci of rapidly multiplying cells that failed to senesce, in contrast to controls. The transformed culture, designated GMSM-K, was polyclonal. The original culture possessed a normal human male karyotype, and the transformed line was largely hypotetraploid. Multiple clones, isolated from soft agar studies and low density plating, showed decreased doubling times. Electron microscopy and immunohistochemistry confirmed an epithelial phenotype. Cells did not generate tumors in nude mice. CONCLUSION: Few human epithelial cell lines are available to investigators and most are tumor-derived. The nontumor-derived GMSM-K line has value as a resource for human oral epithelial cell research.


Subject(s)
Cell Line, Transformed , Keratinocytes , Mouth Mucosa/cytology , Animals , Cell Culture Techniques/methods , Clone Cells , Humans , Karyotyping , Male , Mice , Mice, Nude , Simian virus 40 , Transfection
18.
Pancreas ; 20(4): 337-47, 2000 May.
Article in English | MEDLINE | ID: mdl-10824687

ABSTRACT

Our previous studies in the hamster pancreatic cancer model have shown that exocrine pancreatic cancer arises from ductal/ductular cells, as well as from within the islets, most probably from islet precursor (stem) cells. To identify and characterize these cells, we established a long-term culture from isolated hamster islets and investigated their growth, differentiation, and expression of biomarkers. Islets maintained their original form and structure within the first 14 days in culture. However, beginning at day 7, ductular structures began to form within the islets. At day 21 in culture, acinar cells, intermediary cells, oncocytes, and cells comparable to pancreatic hepatocytes also appeared between ductular and endocrine cells. The number of duct-like cells gradually increased, whereas the number of hormone-producing cells decreased. After 35 days in culture, the exocrine cells disappeared, and undifferentiated cells formed a monolayer. These cells expressed cytokeratins, alpha1-antitrypsin, transforming growth factor-alpha, epidermal growth factor receptor, carbonic anhydrase II, vimentin, laminin, and showed binding to tomato lectin and Phaseolus vulgaris leukoagglutinin. They did not express the regulatory transcriptional factors, insulin-promoting factor 1, NKx6.1, Pax6, and NeuroD. The results thus indicate that islet cells have potential to form exocrine cells. At present, it is not clear whether these cells originate from preexisting stem cells or from transdifferentiated islet cells.


Subject(s)
Cell Differentiation , Islets of Langerhans/ultrastructure , Animals , Cells, Cultured , Cricetinae , Exocrine Glands/chemistry , Exocrine Glands/ultrastructure , Female , Gene Expression , Insulin/genetics , Islets of Langerhans/chemistry , Keratins/analysis , Mesocricetus , Microscopy, Electron , Phenotype , Time Factors , Transcription Factors/genetics
19.
Cancer Gene Ther ; 7(4): 563-73, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10811474

ABSTRACT

Herpes simplex virus thymidine kinase (HSV-TK) and ganciclovir (GCV) gene therapy can induce apoptosis in tumor cells that are normally resistant to this type of cell death, although the cellular mechanisms by which this occurs remain to be elucidated. Human colon tumor cell lines expressing HSV-TK were treated with GCV or four other inducers of apoptosis: butyrate, camptothecin (CPT), Taxol (paclitaxel), or 7-hydroxystaurosporine (UCN-01). Over a 2-4 day treatment period with GCV or the other four drugs, protein levels of the apoptosis agonist Bak increased 1.5- to 3-fold, whereas a corresponding decrease in the levels of the apoptosis antagonist, Bcl-X(L), was observed in butyrate-, CPT-, and 7-hydroxystaurosporine (UCN-01)-treated cells. GCV and paclitaxel treatments resulted in increased levels of Bcl-X(L). In two-drug combinations with GCV plus one of the four other drugs, increased tumor cell killing was found with GCV plus UCN-01 or with some GCV/butyrate combinations; the other two tested combinations were largely antagonistic. The GCV/UCN-01 and GCV/butyrate combinations resulted in increased Bak and decreased Bcl-X(L) protein levels, while the GCV/CPT and GCV/paclitaxel combinations resulted in increased levels of both proteins. The results highlight the potential for new combination therapies of HSV-TK/GCV and chemotherapeutic drugs that result in increased tumor cell apoptosis for future treatments of colon cancer.


Subject(s)
Antineoplastic Agents/toxicity , Apoptosis/drug effects , Ganciclovir/toxicity , Membrane Proteins/biosynthesis , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Thymidine Kinase/genetics , Alkaloids/toxicity , Butyrates/toxicity , Camptothecin/toxicity , Caspase 3 , Caspases/metabolism , Cell Survival/drug effects , Colonic Neoplasms , Drug Interactions , Humans , Membrane Proteins/analysis , Paclitaxel/toxicity , Peptide Hydrolases/metabolism , Proto-Oncogene Proteins c-bcl-2/analysis , Recombinant Proteins/metabolism , Simplexvirus/enzymology , Simplexvirus/genetics , Staurosporine/analogs & derivatives , Thymidine Kinase/metabolism , Transfection , Tumor Cells, Cultured , bcl-2 Homologous Antagonist-Killer Protein , bcl-X Protein
20.
Cancer Lett ; 151(2): 145-51, 2000 Apr 14.
Article in English | MEDLINE | ID: mdl-10738108

ABSTRACT

A549 xenografts were allowed to grow in nude mice to about 5 mm in diameter, then diets were changed to modified AIN-76 diets containing 19% wt./wt. fish oil (FO) or 20% wt./wt. corn oil (CO). Ten days later dietary ferric citrate (0.3% wt./dry wt.) was added and doxoribicin (DOX) treatment (3.6 mg/kg i.v. each of the 5 days for 18 days) commenced. Treatment with DOX halted the growth of tumors in the CO fed mice. However, in those mice, which consumed FO or FO with ferric citrate, treatment with DOX caused significant tumor regression.


Subject(s)
Antineoplastic Agents/therapeutic use , Dietary Fats, Unsaturated/administration & dosage , Doxorubicin/therapeutic use , Fish Oils/administration & dosage , Lung Neoplasms/drug therapy , Analysis of Variance , Animals , Body Weight/drug effects , Cell Division/drug effects , Corn Oil/administration & dosage , Corn Oil/pharmacology , Dietary Fats, Unsaturated/pharmacology , Drug Synergism , Ferric Compounds/pharmacology , Fish Oils/pharmacology , Humans , Lung Neoplasms/diet therapy , Lung Neoplasms/pathology , Male , Mice , Mice, Nude , Neoplasm Transplantation , Remission Induction , Transplantation, Heterologous , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...