Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Sci Rep ; 11(1): 18952, 2021 09 23.
Article in English | MEDLINE | ID: mdl-34556693

ABSTRACT

Proper regulation of the cell cycle is necessary for normal growth and development of all organisms. Conversely, altered cell cycle regulation often underlies proliferative diseases such as cancer. Long non-coding RNAs (lncRNAs) are recognized as important regulators of gene expression and are often found dysregulated in diseases, including cancers. However, identifying lncRNAs with cell cycle functions is challenging due to their often low and cell-type specific expression. We present a highly effective method that analyses changes in promoter activity, transcription, and RNA levels for identifying genes enriched for cell cycle functions. Specifically, by combining RNA sequencing with ChIP sequencing through the cell cycle of synchronized human keratinocytes, we identified 1009 genes with cell cycle-dependent expression and correlated changes in RNA polymerase II occupancy or promoter activity as measured by histone 3 lysine 4 trimethylation (H3K4me3). These genes were highly enriched for genes with known cell cycle functions and included 57 lncRNAs. We selected four of these lncRNAs-SNHG26, EMSLR, ZFAS1, and EPB41L4A-AS1-for further experimental validation and found that knockdown of each of the four lncRNAs affected cell cycle phase distributions and reduced proliferation in multiple cell lines. These results show that many genes with cell cycle functions have concomitant cell-cycle dependent changes in promoter activity, transcription, and RNA levels and support that our multi-omics method is well suited for identifying lncRNAs involved in the cell cycle.


Subject(s)
Cell Cycle/genetics , Cell Proliferation/genetics , RNA Polymerase II/metabolism , RNA, Long Noncoding/metabolism , Chromatin Immunoprecipitation Sequencing , Gene Knockdown Techniques , HaCaT Cells , Humans , Promoter Regions, Genetic , RNA, Long Noncoding/genetics , RNA-Seq
2.
BMC Cancer ; 17(1): 68, 2017 01 21.
Article in English | MEDLINE | ID: mdl-28109268

ABSTRACT

BACKGROUND: The peptide hormone gastrin exerts a growth-promoting effect in both normal and malignant gastrointestinal tissue. Gastrin mediates its effect via the cholecystokinin 2 receptor (CCKBR/CCK2R). Although a substantial part of the gastric adenocarcinomas express gastrin and CCKBR, the role of gastrin in tumor development is not completely understood. Autophagy has been implicated in mechanisms governing cytoprotection, tumor growth, and contributes to chemoresistance. This study explores the role of autophagy in response to gastrin in gastric adenocarcinoma cell lines. METHODS: Immunoblotting, survival assays and the xCELLigence system were used to study gastrin induced autophagy. Chemical inhibitors of autophagy were utilized to assess the role of this process in the regulation of cellular responses induced by gastrin. Further, knockdown studies using siRNA and immunoblotting were performed to explore the signaling pathways that activate autophagy in response to gastrin treatment. RESULTS: We demonstrate that gastrin increases the expression of the autophagy markers MAP1LC3B-II and SQSTM1 in gastric adenocarcinoma cells. Gastrin induces autophagy via activation of the STK11-PRKAA2-ULK1 and that this signaling pathway is involved in increased migration and cell survival. Furthermore, gastrin mediated increase in survival of cells treated with cisplatin is partially dependent on induced autophagy. CONCLUSION: This study reveals a novel role of gastrin in the regulation of autophagy. It also opens up new avenues in the treatment of gastric cancer by targeting CCKBR mediated signaling and/or autophagy in combination with conventional cytostatic drugs.


Subject(s)
Adenocarcinoma/genetics , Gastrins/metabolism , Microtubule-Associated Proteins/genetics , Sequestosome-1 Protein/genetics , Stomach Neoplasms/genetics , Adenocarcinoma/metabolism , Autophagy , Cell Line, Tumor , Cell Movement , Cell Survival , Gene Expression Regulation, Neoplastic , Humans , Receptor, Cholecystokinin B/metabolism , Signal Transduction , Stomach Neoplasms/metabolism
3.
Eur J Med Chem ; 124: 583-607, 2016 Nov 29.
Article in English | MEDLINE | ID: mdl-27614407

ABSTRACT

The present study describes our continuous effort to develop epidermal growth factor receptor (EGFR) inhibitors based on the 6-aryl-pyrrolo[2,3-d]pyrimidin-4-amine scaffold. The activity-ADME space has been evaluated by synthesizing 43 new structures, including four variations of the 4-amino group and 34 different substitution patterns in the 6-aryl moiety. Most of the new pyrrolopyrimidines were highly active, with twelve analogues possessing lower IC50 values than the commercial drug Erlotinib in enzymatic assays. Ten EGFR inhibitors were also profiled in cell studies using the Ba/F3-EGFRL858R reporter cells, and all revealed nanomolar activity. However, some of the privileged structures in terms of potency had ADME short-comings: compounds containing amides, sulfonamides, amine and hydroxymethyl substituents in the 6-aryl group had low permeability and high efflux, derivatives having (R)-3-amino-3-phenylpropan-1-ol at C-4 induced hERG inhibition properties, and metabolic lability was seen for compounds having (S)-2-methoxy-1-phenylethan-1-amine at C-4. Based on a trade-off between enzymatic activity, cellular potency and ADME properties, (S)-2-phenyl-2-((6-phenyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)ethan-1-ol appeared as the most promising drug candidate. Cellular studies indicate this compound to have therapeutic use in EGFR driven diseases.


Subject(s)
Drug Design , ErbB Receptors/antagonists & inhibitors , Models, Molecular , Pyrimidines/chemistry , Pyrroles/chemistry , Cell Line , Cell Proliferation/drug effects , Drug Stability , Humans , Inhibitory Concentration 50 , Pyrimidines/chemical synthesis , Pyrimidines/metabolism , Pyrimidines/pharmacology , Pyrroles/chemical synthesis , Pyrroles/metabolism , Pyrroles/pharmacology , Structure-Activity Relationship
4.
Biochem Biophys Res Commun ; 475(1): 119-24, 2016 06 17.
Article in English | MEDLINE | ID: mdl-27179776

ABSTRACT

Connective tissue growth factor (CTGF) has been reported in gastric adenocarcinoma and in carcinoid tumors. The aim of this study was to explore a possible link between CTGF and gastrin in gastric epithelial cells and to study the role of CTGF in gastrin induced migration and invasion of AGS-GR cells. The effects of gastrin were studied using RT-qPCR, Western blot and assays for migration and invasion. We report an association between serum gastrin concentrations and CTGF abundancy in the gastric corpus mucosa of hypergastrinemic subjects and mice. We found a higher expression of CTGF in gastric mucosa tissue adjacent to tumor compared to normal control tissue. We showed that gastrin induced expression of CTGF in gastric epithelial AGS-GR cells via MEK, PKC and PKB/AKT pathways. CTGF inhibited gastrin induced migration and invasion of AGS-GR cells. We conclude that CTGF expression is stimulated by gastrin and involved in remodeling of the gastric epithelium.


Subject(s)
Adenocarcinoma/metabolism , Connective Tissue Growth Factor/metabolism , Gastrins/metabolism , Stomach Neoplasms/metabolism , Stomach/pathology , Adenocarcinoma/pathology , Animals , Cell Movement , Gastric Mucosa/metabolism , Humans , Mice , Neoplasm Invasiveness/pathology , Signal Transduction , Stomach Neoplasms/pathology
5.
Eur J Med Chem ; 119: 278-99, 2016 Aug 25.
Article in English | MEDLINE | ID: mdl-27235841

ABSTRACT

Epidermal growth factor receptor inhibitors are of importance in cancer therapy and possibly in the management of pain. Herein, we report a structure-activity relationship study with 29 new 6-aryl-furo[2,3-d]pyrimidin-4-amines, involving modification of the 4-amino group and 6-aryl function. The EGFR activity was especially dependent on having a chiral 4-benzylamino group with correct stereochemistry. Molecular dynamics indicate this to be due to favourable cation-π interactions. The most active inhibitor identified, equipotent to Erlotinib, was substituted with (R)-1-phenylethylamine at C-4 and a N(1), N(1)-dimethyl-1,2-diamine group in para position of the 6-aryl moiety. These new furopyrimidines had a different off-target kinase profile when compared to Erlotinib, and also possessed high activity towards Ba/F3 EGFR(L858R) reporter cells. Further, comparing the EGFR data of the furo[2,3-d]pyrimidin-4-amines with that of the corresponding thieno- and pyrrolopyrimidines concludes the furopyrimidine scaffold to be highly useful for development of new epidermal growth factor receptor antagonists.


Subject(s)
Amines/chemistry , ErbB Receptors/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , ErbB Receptors/chemistry , ErbB Receptors/metabolism , Inhibitory Concentration 50 , Kinetics , Molecular Docking Simulation , Protein Conformation , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/metabolism , Pyrimidines/chemical synthesis , Pyrimidines/metabolism , Stereoisomerism , Structure-Activity Relationship
6.
Peptides ; 79: 83-94, 2016 05.
Article in English | MEDLINE | ID: mdl-27038741

ABSTRACT

The gastric hormone gastrin plays a role in organizing the gastric mucosa. Gastrin also regulates the expression of genes that have important actions in extracellular matrix modelling, including plasminogen activator inhibitor (PAI)-1 which is part of the urokinase plasminogen activator (uPA) system. The uPA system (including PAI-1) is associated with cancer progression, fibrosis and thrombosis. Its biological role in the stomach and molecular mechanisms of action are not well understood. The aim of this study was to examine the effect of PAI-1 on the trophic changes observed in gastric corpus mucosa in hypergastrinemia using PAI-1 and/or HK-ATPase beta subunit knockout (KO) mice. HK-ATPase beta subunit KO mice were used as a model of hypergastrinemia. In 12 month old female mice, intragastric acidity and plasma gastrin were measured. The stomachs were examined for macroscopic and histological changes. In mice null for both PAI-1 and HK-ATPase beta (double KO), there was exaggerated hypergastrinemia, increased stomach weight and corpus mucosal thickness, and more pronounced trophic and architectural changes in the corpus compared with HK-ATPase beta KO mice. Genome-wide microarray expression data for the gastric corpus mucosa showed a distinct gene expression profile for the HK-ATPase beta KO mice; moreover, enrichment analysis revealed changes in expression of genes regulating intracellular processes including cytoskeleton remodelling, cell adhesion, signal transduction and epithelial-to-mesenchymal transition (EMT). Genes differentially expressed in the double KO compared with HK-ATPase beta KO mice included the transcription factor Barx2 and the chromatin remodeler gene Tet2, which may be involved in both normal gastric physiology and development of gastric cancer. Based on the present data, we suggest that PAI-1 plays a role in maintaining gastric mucosal organization in hypergastrinemia.


Subject(s)
Gastric Mucosa/enzymology , Gastrins/blood , Serpin E2/genetics , Animals , Enterochromaffin-like Cells/enzymology , Enterochromaffin-like Cells/pathology , Female , Gastric Mucosa/pathology , H(+)-K(+)-Exchanging ATPase/genetics , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Pyloric Antrum/enzymology , Pyloric Antrum/pathology , Serpin E2/metabolism
7.
Eur J Pharm Sci ; 59: 69-82, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24769040

ABSTRACT

The epidermal growth factor receptor is an important target in molecular cancer therapy. Herein, the enzymatic inhibition potential of a series of chiral and non chiral pyrrolopyrimidine based derivatives have been investigated and optimised. Overall, seven new compounds were identified having enzymatic IC50 values comparable to or better than the commercial drug Erlotinib. High activity was also confirmed towards the epidermal growth factor receptor L858R and L861Q mutants. Based on calculated druglike properties, eight compounds were further evaluated towards a panel of 52 other kinases revealing interesting Src-family kinase and colony stimulating factor 1 receptor kinase inhibitory activity. Cell proliferation studies with the cell lines A431, C-33A, AU-565, K-562 and genetically engineered Ba/F3-EGFR(L858R) cells also showed several molecules to be more active than Erlotinib, and thus confirming these pyrrolopyrimidines as attractive drug candidates or lead structures.


Subject(s)
Amines/pharmacology , ErbB Receptors/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , src-Family Kinases/metabolism , Amines/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , ErbB Receptors/metabolism , Humans , Models, Molecular , Pyrimidines/chemistry , Pyrroles/chemistry , Structure-Activity Relationship
8.
Am J Physiol Gastrointest Liver Physiol ; 301(3): G446-53, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21193525

ABSTRACT

Plasminogen activator inhibitor (PAI)-1 is associated with cancer progression, fibrosis and thrombosis. It is expressed in the stomach but the mechanisms controlling its expression there, and its biological role, are uncertain. We sought to define the role of gastrin in regulating PAI-1 expression and to determine the relevance for gastrin-stimulated cell migration and invasion. In gastric biopsies from subjects with elevated plasma gastrin, the abundances of PAI-1, urokinase plasminogen activator (uPA), and uPA receptor (uPAR) mRNAs measured by quantitative PCR were increased compared with subjects with plasma concentrations in the reference range. In patients with hypergastrinemia due to autoimmune chronic atrophic gastritis, there was increased abundance of PAI-1, uPA, and uPAR mRNAs that was reduced by octreotide or antrectomy. Immunohistochemistry revealed localization of PAI-1 to parietal cells and enterochromaffin-like cells in micronodular neuroendocrine tumors in hypergastrinemic subjects. Transcriptional mechanisms were studied by using a PAI-1-luciferase promoter-reporter construct transfected into AGS-G(R) cells. There was time- and concentration-dependent increase of PAI-1-luciferase expression in response to gastrin that was reversed by inhibitors of the PKC and MAPK pathways. In Boyden chamber assays, recombinant PAI-1 inhibited gastrin-stimulated AGS-G(R) cell migration and invasion, and small interfering RNA treatment increased responses to gastrin. We conclude that elevated plasma gastrin concentrations are associated with increased expression of gastric PAI-1, which may act to restrain gastrin-stimulated cell migration and invasion.


Subject(s)
Epithelial Cells/metabolism , Gastrins/pharmacology , Plasminogen Activator Inhibitor 1/biosynthesis , Enterochromaffin-like Cells/metabolism , Gastrins/blood , Humans , Octreotide , RNA, Messenger/metabolism , Receptors, Urokinase Plasminogen Activator/biosynthesis , Stomach/cytology , Urokinase-Type Plasminogen Activator/biosynthesis
9.
Eur J Gastroenterol Hepatol ; 20(7): 613-23, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18679062

ABSTRACT

BACKGROUND: Long-term therapy with potent acid inhibitors is a common treatment for gastro-esophageal reflux disease. Administration of proton pump inhibitors (PPIs) causes profound and continuous hypochlorhydria by inhibition of the proton pump in gastric parietal cells. Long-term hypergastrinaemia increases mucosal thickness and enterochromaffin-like cell density in oxyntic mucosa. OBJECTIVE: The aim of this study was to see whether this very common clinical intervention induces significant changes in the gastric mucosal gene expression pattern. METHODS: Seven patients suffering from gastro-esophageal reflux disease were included in this study. Endoscopic biopsies were taken from the corpus mucosa before and toward the end of a 3-month treatment with the PPI esomeprazole. RESULTS: Microarray analysis identified 186 differentially expressed genes. A high proportion of genes with changed gene expression levels during PPI treatment are involved in proliferation, apoptosis, and stress response. CONCLUSION: This study identified many genes that were not previously known to be affected by inhibition of gastric acid secretion. Further characterization of the functional roles of genes whose expression is modulated by potent acid inhibition may give new insight into the biological responses to potent acid inhibition, including the mucosal response to the moderately increased gastrin levels encountered in clinical practice.


Subject(s)
Gastric Mucosa/drug effects , Gastroesophageal Reflux/drug therapy , Gene Expression Regulation/drug effects , Proton Pump Inhibitors/pharmacology , Adult , Aged , Anti-Ulcer Agents/pharmacology , Anti-Ulcer Agents/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Biopsy , Cell Proliferation/drug effects , Esomeprazole/pharmacology , Esomeprazole/therapeutic use , Esophagoscopy , Female , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Gastrins/blood , Gastroesophageal Reflux/metabolism , Gastroesophageal Reflux/pathology , Gene Expression Profiling/methods , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis/methods , Proton Pump Inhibitors/therapeutic use
10.
Physiol Genomics ; 22(1): 24-32, 2005 Jun 16.
Article in English | MEDLINE | ID: mdl-15827235

ABSTRACT

Potent acid inhibition with proton pump inhibitors (PPIs) is widely used in clinical medicine, especially for gastroesophageal reflux disease. PPIs cause profound changes in the intragastric environment with near-neutral pH and increase serum concentration of the gastric secretagogue hormone gastrin. Long-term hypergastrinemia increases mucosal thickness and enterochromaffin-like cell density in gastric corpus mucosa and results in development of gastric carcinoids in experimental animals. Our aim was to study responses to potent acid inhibition by characterizing genome-wide gene expression changes in gastric corpus mucosa in rats dosed with the PPI omeprazole. Nine rats received 400 micromol/kg omeprazole daily for 10 wk. Seven rats received vehicle only. Analysis of gastric corpus with microarrays representing 11,848 genes identified 134 genes with changed gene expression levels in omeprazole-dosed rats. Several of the identified genes were previously known to be affected by potent acid inhibition. Of the 62 genes with known functions that changed gene expression levels after PPI dosing, 27 are known to be involved in proliferation and apoptosis and immune, inflammatory, and stress responses. Our study indicates that microarray analysis can detect relevant gene expression changes in the complex gastric tissue, and that cellular processes involved in cell growth and defense responses are strongly affected by PPI dosing. Many genes are identified that were not previously known to be affected by inhibition of gastric acid secretion or that have unknown biological functions. Characterization of the roles of these genes may give new insight into molecular responses to treatment with PPIs.


Subject(s)
Gastric Acid/metabolism , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Gene Expression Regulation/drug effects , Omeprazole/pharmacology , Animals , Blotting, Northern , Gastric Mucosa/cytology , Gene Expression Regulation/genetics , Humans , Immunohistochemistry , Male , Microarray Analysis , Parietal Cells, Gastric/cytology , Quality Control , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, CXCR4/metabolism , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA
11.
Cancer Lett ; 210(2): 227-37, 2004 Jul 16.
Article in English | MEDLINE | ID: mdl-15183539

ABSTRACT

The aim of the present work is to identify molecular markers that allow classification of gastric carcinoma with respect to important clinicopathological parameters. Gastric adenocarcinomas were subjected to cDNA microarray analysis with a 2.504 gene probe set. Using the Rosetta rough-set based learning system, good classifiers were generated for gene-expression based prediction of intestinal or diffuse growth pattern according to Laurén's classification and presence of lymph node metastases. To our knowledge, this is the first study on gastric carcinoma in which molecular classification has been achieved for more than one clinicopathological parameter based on microarray gene expression profiles.


Subject(s)
Adenocarcinoma/genetics , Gene Expression Profiling , Genetic Markers , Lymphatic Metastasis , Oligonucleotide Array Sequence Analysis , Stomach Neoplasms/genetics , Adenocarcinoma/pathology , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Prognosis , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...