Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Cancers (Basel) ; 13(9)2021 Apr 25.
Article in English | MEDLINE | ID: mdl-33923068

ABSTRACT

Colorectal cancer (CRC) develops through a multi-step process characterized by the acquisition of multiple somatic mutations in oncogenes and tumor-suppressor genes, epigenetic alterations and genomic instability. These events lead to the progression from precancerous lesions to advanced carcinomas. This process requires several years in a sporadic setting, while occurring at an early age and or faster in patients affected by hereditary CRC-predisposing syndromes. Since advanced CRC is largely untreatable or unresponsive to standard or targeted therapies, the endoscopic treatment of colonic lesions remains the most efficient CRC-preventive strategy. In this review, we discuss recent studies that have assessed the genetic alterations in early colorectal lesions in both hereditary and sporadic settings. Establishing the genetic profile of early colorectal lesions is a critical goal in the development of risk-based preventive strategies.

2.
Cell Mol Gastroenterol Hepatol ; 10(3): 491-506, 2020.
Article in English | MEDLINE | ID: mdl-32334125

ABSTRACT

Wnt/ß-catenin and phosphatidylinositol-3-kinase/protein kinase B/mammalian target of rapamycin complex 1 (PI3K/AKT/mTORC1) pathways both are critically involved in colorectal cancer (CRC) development, although they are implicated in the modulation of distinct oncogenic mechanisms. In homeostatic and pathologic conditions, these pathways show a fine regulation based mainly on feedback mechanisms, and are connected at multiple levels involving both upstream and downstream common effectors. The ability of the Wnt/ß-catenin and PI3K/AKT/mTORC1 pathways to reciprocally control themselves represents one of the main resistance mechanisms to selective inhibitors in CRC, leading to the hypothesis that in specific settings, particularly in cancer driven by genetic alterations in Wnt/ß-catenin signaling, the relationship between Wnt/ß-catenin and PI3K/AKT/mTORC1 pathways could be so close that they should be considered as a unique therapeutic target. This review provides an update on the Wnt/ß-catenin and PI3K/AKT/mTORC1 pathway interconnections in CRC, describing the main molecular players and the potential implications of combined inhibitors as an approach for CRC chemoprevention and treatment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/pathology , Signal Transduction/genetics , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/genetics , Wnt Proteins/metabolism , beta Catenin/antagonists & inhibitors , beta Catenin/genetics , beta Catenin/metabolism
3.
Front Oncol ; 9: 140, 2019.
Article in English | MEDLINE | ID: mdl-30915275

ABSTRACT

Objectives: Unhealthy dietary patterns have been associated with colorectal cancer (CRC) onset while Mediterranean Diet (MD) has been proposed for CRC prevention. This study evaluated the effect of a Mediterranean Diet Mix (MD-MIX) on colonic tumors development in A/J mice fed a low-fat (LFD) or a high-fat western diet (HFWD), and injected with the procarcinogen azoxymethane (AOM). Materials and Methods: Forty A/J male mice were randomly assigned into four feeding arms (10 mice/arm; LFD, LFD-MD-MIX, HFWD, HFWD-MD-MIX) to be treated with AOM. Ten mice were exposed to the diets alone (Healthy LFD and Healthy HFWD) to be used as control. Tumor incidence and multiplicity were evaluated at sacrifice. Mucosal fatty acid content and urinary phenolic compounds were assayed by mass spectrometry. Apoptosis was evaluated by TUNEL assay and gene expression markers. Cell proliferation was evaluated by Ki67 immunohistochemistry. Microbiota composition was assessed at different time points by 16S RNA sequencing. Results: A tumor incidence of 100% was obtained in AOM-treated mice. The MD-MIX supplementation was able to reduce the number of colonic lesions in both LFD and HFWD-fed mice and to induce apoptosis, in particular in the LFD-MD-MIX arm. Moreover, a preventive effect on low-grade dysplasia and macroscopical lesions (>1 mm) development was found in HFWD-fed mice together with a regulation of the AOM-driven intestinal dysbiosis. Conclusions: MD-MIX was able to counteract CRC development in mice under different dietary backgrounds through the regulation of apoptosis and gut microbiota.

4.
Mol Cancer Res ; 16(12): 1965-1976, 2018 12.
Article in English | MEDLINE | ID: mdl-30072583

ABSTRACT

Adenomatous Polyposis Coli (APC) gene mutations are responsible for the onset of familial adenomatous polyposis (FAP) and sporadic colorectal cancer and have been associated with miRNAs dysregulation. The capacity of miR-155, a cancer-related miRNA, to target components of the WNT/ß-CATENIN pathway suggests that APC gene mutations, controlling miRNAs expression, may critically regulate WNT/ß-CATENIN signaling. To this end, APC gene target sequencing was performed on colonic adenomatous polyps and paired normal mucosa clinical specimens from FAP patients (n = 9) to elucidate the role of miR-155-5p in APC-mutant setting. The expression of selected miRNAs and WNT/ß-CATENIN signaling components was characterized in FAP patients and non-FAP control subjects (n = 5). miR-155-5p expression and functional effects on WNT cascade, cell survival, growth, and apoptosis were investigated in different colorectal cancer cell lines. A somatic second hit in the APC gene was found in adenomatous polyps from 6 of 9 FAP patients. Heterozygous APC gene mutations in FAP patients were associated with altered expression of candidate miRNAs and increased levels of AXIN1 and AXIN2 mRNAs. miR-155-5p was downregulated in FAP patients and in the APC and ß-CATENIN-mutant colorectal cancer cell lines, and critically regulates WNT/ß-CATENIN cascade by targeting both AXIN1 and TCF4. Importantly, miR-155-5p may sustain long-term WNT/ß-CATENIN activation in colorectal cancer cells upon WNT3A stimulation. IMPLICATIONS: This study supports a key role of miR-155-5p in modulating WNT/ß-CATENIN signaling in colorectal cancer and unravels a new mechanism for AXIN1 regulation which represents a potential therapeutic target in specific tumor subtypes.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli/genetics , Axin Protein/genetics , Down-Regulation , MicroRNAs/genetics , Transcription Factor 7-Like 2 Protein/genetics , Caco-2 Cells , Cell Line, Tumor , Cell Proliferation , Cell Survival , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Male , Mutation , Sequence Analysis, RNA/methods , Wnt Signaling Pathway
5.
Eur J Hum Genet ; 26(3): 387-395, 2018 03.
Article in English | MEDLINE | ID: mdl-29367705

ABSTRACT

Germline variants in the APC gene cause familial adenomatous polyposis. Inherited variants in MutYH, POLE, POLD1, NTHL1, and MSH3 genes and somatic APC mosaicism have been reported as alternative causes of polyposis. However, ~30-50% of cases of polyposis remain genetically unsolved. Thus, the aim of this study was to investigate the genetic causes of unexplained adenomatous polyposis. Eight sporadic cases with >20 adenomatous polyps by 35 years of age or >50 adenomatous polyps by 55 years of age, and no causative germline variants in APC and/or MutYH, were enrolled from a cohort of 56 subjects with adenomatous colorectal polyposis. APC gene mosaicism was investigated on DNA from colonic adenomas by Sanger sequencing or Whole Exome Sequencing (WES). Mosaicism extension to other tissues (peripheral blood, saliva, hair follicles) was evaluated using Sanger sequencing and/or digital PCR. APC second hit was investigated in adenomas from mosaic patients. WES was performed on DNA from peripheral blood to identify additional polyposis candidate variants. We identified APC mosaicism in 50% of patients. In three cases mosaicism was restricted to the colon, while in one it also extended to the duodenum and saliva. One patient without APC mosaicism, carrying an APC in-frame deletion of uncertain significance, was found to harbor rare germline variants in OGG1, POLQ, and EXO1 genes. In conclusion, our restrictive selection criteria improved the detection of mosaic APC patients. In addition, we showed for the first time that an oligogenic inheritance of rare variants might have a cooperative role in sporadic colorectal polyposis onset.


Subject(s)
Adenomatous Polyposis Coli/genetics , Genes, APC , Mosaicism , Multifactorial Inheritance , Adenomatous Polyposis Coli/pathology , Adult , DNA Glycosylases/genetics , DNA Repair Enzymes/genetics , DNA-Directed DNA Polymerase/genetics , Exodeoxyribonucleases/genetics , Female , Humans , Male , Middle Aged , DNA Polymerase theta
6.
Sci Rep ; 7(1): 7458, 2017 08 07.
Article in English | MEDLINE | ID: mdl-28785079

ABSTRACT

Patients with long-standing ulcerative colitis (UC) have an increased colorectal cancer (CRC) risk. In this pilot study we evaluated the effect of Eicosapentaenoic acid as free fatty acid (EPA-FFA) supplementation on mucosal disease activity, colonic differentiation markers and microbiota composition in UC patients. Twenty long-standing UC patients in stable clinical remission and with fecal calprotectin (FC) > 150 µg/g were enrolled (T0) and supplemented with EPA-FFA 2 g/daily for 90 days (T3). Endoscopic and histologic disease activities were measured by Mayo and Geboes scores, respectively. HES1, KLF4, STAT3, IL-10 and SOCS3 levels were determined using western blotting and qRT-PCR, while phospho-STAT3 levels were assessed by western blotting. Goblet cells were stained by Alcian blue. Microbiota analyses were performed on both fecal and colonic samples. Nineteen patients completed the study; seventeen (89.5%) were compliant. EPA-FFA treatment reduced FC levels at T3. Patients with FC > 150 µg/g at T3 (n = 2) were assumed as non-responders. EPA-FFA improved endoscopic and histological inflammation and induced IL-10, SOCS3, HES1 and KLF4 in compliant and responder patients. Importantly, long-term UC-driven microbiota composition was partially redressed by EPA-FFA. In conclusion, EPA-FFA supplementation reduced mucosal inflammation, promoted goblet cells differentiation and modulated intestinal microbiota composition in long-standing UC patients.


Subject(s)
Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/immunology , Eicosapentaenoic Acid/administration & dosage , Fatty Acids, Nonesterified/administration & dosage , Microbiota/drug effects , Adult , Aged , Aged, 80 and over , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/microbiology , Eicosapentaenoic Acid/pharmacology , Fatty Acids, Nonesterified/pharmacology , Female , Gene Expression Regulation/drug effects , Humans , Interleukin-10/genetics , Interleukin-10/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Leukocyte L1 Antigen Complex/metabolism , Male , Middle Aged , Phosphorylation/drug effects , Pilot Projects , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/metabolism , Transcription Factor HES-1/genetics , Transcription Factor HES-1/metabolism , Treatment Outcome , Young Adult
7.
Sci Rep ; 6: 20670, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26864323

ABSTRACT

Aberrant NOTCH1 signalling is critically involved in multiple models of colorectal cancer (CRC) and a prominent role of NOTCH1 activity during inflammation has emerged. Epithelial to Mesenchymal Transition (EMT), a crucial event promoting malignant transformation, is regulated by inflammation and Metalloproteinase-9 (MMP9) plays an important role in this process. Eicosapentaenoic Acid (EPA), an omega-3 polyunsaturated fatty acid, was shown to prevent colonic tumors in different settings. We recently found that an extra-pure formulation of EPA as Free Fatty Acid (EPA-FFA) protects from colon cancer development in a mouse model of Colitis-Associated Cancer (CAC) through modulation of NOTCH1 signalling. In this study, we exposed colon cancer cells to an inflammatory stimulus represented by a cytokine-enriched Conditioned Medium (CM), obtained from THP1-differentiated macrophages. We found, for the first time, that CM strongly up-regulated NOTCH1 signalling and EMT markers, leading to increased invasiveness. Importantly, NOTCH1 signalling was dependent on MMP9 activity, upon CM exposure. We show that a non-cytotoxic pre-treatment with EPA-FFA antagonizes the effect of inflammation on NOTCH1 signalling, with reduction of MMP9 activity and invasiveness. In conclusion, our data suggest that, in CRC cells, inflammation induces NOTCH1 activity through MMP9 up-regulation and that this mechanism can be counteracted by EPA-FFA.


Subject(s)
Culture Media, Conditioned/pharmacology , Cytokines/metabolism , Eicosapentaenoic Acid/pharmacology , Matrix Metalloproteinase 9/genetics , Monocytes/metabolism , Receptor, Notch1/genetics , Cell Differentiation , Cell Line , Cell Movement/drug effects , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation , HCT116 Cells , HT29 Cells , Humans , Inflammation , Lipopolysaccharides/pharmacology , Matrix Metalloproteinase 9/metabolism , Monocytes/cytology , Monocytes/drug effects , Receptor, Notch1/agonists , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/metabolism , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology
8.
Carcinogenesis ; 35(10): 2314-20, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25123131

ABSTRACT

Colorectal cancer (CRC) is one of the major causes of cancer death worldwide. The development of novel anti-CRC agents able to overcome drug resistance and/or off-target toxicity is of pivotal importance. The mammalian target of rapamycin (mTOR) plays a critical role in CRC, regulating protein translation and controlling cell growth, proliferation, metabolism and survival. The aim of this study was to explore the effect of a combination of three natural compounds, eicosapentaenoic acid-free fatty acid (EPA-FFA), epigallocatechin-3-gallate (EGCG) and proanthocyanidins (grape seed [GS] extract) at low cytotoxic concentrations on CRC cells and test their activity on mTOR and translational regulation. The CRC cell lines HCT116 and SW480 were treated for 24h with combinations of EPA-FFA (0-150 µM), EGCG (0-175 µM) and GS extract (0-15 µM) to evaluate the effect on cell viability. The low cytotoxic combination of EPA-FFA 150 µM, EGCG 175 µM and GS extract 15 µM completely inhibited the mTOR signaling in HCT116 and SW480 cells, reaching an effect stronger than or comparable to that of the mTOR inhibitor Rapamycin in HCT116 or SW480 cells, respectively. Moreover, the treatment led to changes of protein translation of ribosomal proteins, c-Myc and cyclin D1. In addition, we found a reduction of clonal capability in both cell lines, with block of cell cycle in G0G1 and induction of apoptosis. Our data suggest that the low cytotoxic combination of EPA-FFA, EGCG and GS extract, tested for the first time here, inhibits mTOR signaling and thus could be considered for CRC treatment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Catechin/analogs & derivatives , Colorectal Neoplasms/drug therapy , Eicosapentaenoic Acid/pharmacology , Proanthocyanidins/pharmacology , TOR Serine-Threonine Kinases/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Catechin/administration & dosage , Catechin/pharmacology , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/metabolism , Cyclin D1/genetics , Cyclin D1/metabolism , Eicosapentaenoic Acid/administration & dosage , Grape Seed Extract/pharmacology , Humans , Proanthocyanidins/administration & dosage , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction/drug effects
9.
Int J Cancer ; 135(9): 2004-13, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-24676631

ABSTRACT

Inflammatory bowel diseases are associated with increased risk of developing colitis-associated colorectal cancer (CAC). Epidemiological data show that the consumption of ω-3 polyunsaturated fatty acids (ω-3 PUFAs) decreases the risk of sporadic colorectal cancer (CRC). Importantly, recent data have shown that eicosapentaenoic acid-free fatty acid (EPA-FFA) reduces polyp formation and growth in models of familial adenomatous polyposis. However, the effects of dietary EPA-FFA are unknown in CAC. We tested the effectiveness of substituting EPA-FFA, for other dietary fats, in preventing inflammation and cancer in the AOM-DSS model of CAC. The AOM-DSS protocols were designed to evaluate the effect of EPA-FFA on both initiation and promotion of carcinogenesis. We found that EPA-FFA diet strongly decreased tumor multiplicity, incidence and maximum tumor size in the promotion and initiation arms. Moreover EPA-FFA, in particular in the initiation arm, led to reduced cell proliferation and nuclear ß-catenin expression, whilst it increased apoptosis. In both arms, EPA-FFA treatment led to increased membrane switch from ω-6 to ω-3 PUFAs and a concomitant reduction in PGE2 production. We observed no significant changes in intestinal inflammation between EPA-FFA treated arms and AOM-DSS controls. Importantly, we found that EPA-FFA treatment restored the loss of Notch signaling found in the AOM-DSS control and resulted in the enrichment of Lactobacillus species in the gut microbiota. Taken together, our data suggest that EPA-FFA is an excellent candidate for CRC chemoprevention in CAC.


Subject(s)
Colitis/complications , Colon/pathology , Colorectal Neoplasms/prevention & control , Eicosapentaenoic Acid/administration & dosage , Fatty Acids, Nonesterified/administration & dosage , Gastrointestinal Tract/drug effects , Microbiota/physiology , Receptors, Notch/metabolism , Animals , Apoptosis , Cell Proliferation , Colitis/chemically induced , Colitis/pathology , Colon/microbiology , Colorectal Neoplasms/etiology , Colorectal Neoplasms/pathology , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Immunoenzyme Techniques , Inflammation/etiology , Inflammation/pathology , Inflammation/prevention & control , Male , Mice , Mice, Inbred C57BL , Microbiota/drug effects , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
10.
Cell Cycle ; 11(23): 4323-7, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23085543

ABSTRACT

The Notch signaling pathway drives proliferation, differentiation, apoptosis, cell fate choices and maintenance of stem cells during embryogenesis and in self-renewing tissues of the adult. In addition, aberrant Notch signaling has been implicated in several tumors, where Notch can function both as an oncogene or a tumor-suppressor gene, depending on the context.   This Extra View aims to review what is currently known about Notch signaling, in particular in gastrointestinal tumors, providing a summary of our data on Notch1 signaling in gastric cancer with results obtained in colorectal cancer (CRC). We have already reported that the epigenetic regulation of the Notch ligand DLL1 controls Notch1 signaling activation in gastric cancer, and that Notch1 inhibition is associated with the diffuse type of gastric cancer. Here, we describe additional data showing that in CRC cell lines, unlike gastric cancer, DLL1 expression is not regulated by promoter methylation. Moreover, in CRC, Notch1 receptor is not affected by any mutation. These data suggest a different regulation of Notch1 signaling between gastric cancer and CRC.


Subject(s)
Epigenesis, Genetic , Receptors, Notch/metabolism , Calcium-Binding Proteins , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Methylation , Promoter Regions, Genetic , Receptors, Notch/genetics , Signal Transduction
11.
Cancer Prev Res (Phila) ; 4(6): 907-15, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21383028

ABSTRACT

The Western diet (WD) is associated with a higher incidence of colorectal cancer (CRC) than the Mediterranean diet. Polyphenols extracted from Annurca apple showed chemopreventive properties in CRC cells. A multifactorial, four-arm study by using wild-type (wt) and Apc(Min/+) mice was carried out to evaluate the effect on polyp number and growth of APE treatment (60 µmol/L) ad libitum in drinking water combined with a WD or a balanced diet (BD) for 12 weeks. Compared with APE treatment, we found a significant drop in body weight (P < 0.0001), severe rectal bleeding (P = 0.0076), presence of extraintestinal tumors, and poorer activity status (P = 0.0034) in water-drinking Apc(Min/+) mice, more remarkably in the WD arm. In the BD and WD groups, APE reduced polyp number (35% and 42%, respectively, P < 0.001) and growth (60% and 52%, respectively, P < 0.0001) in both colon and small intestine. Increased antioxidant activity was found in wt animals fed both diets and in Apc(Min/+) mice fed WD and drinking APE. Reduced lipid peroxidation was found in Apc(Min/+) mice drinking APE fed both diets and in wt mice fed WD. In normal mucosa, mice drinking water had lower global levels of DNA methylation than mice drinking APE. APE treatment is highly effective in reducing polyps in Apc(Min/+) mice and supports the concept that a mixture of phytochemicals, as they are naturally present in foods, represent a plausible chemopreventive agent for CRC, particularly in populations at high risk for colorectal neoplasia.


Subject(s)
Adenomatous Polyposis Coli Protein/physiology , Chlorogenic Acid/therapeutic use , Colorectal Neoplasms/prevention & control , Diet , Flavonoids/therapeutic use , Intestinal Polyps/prevention & control , Phenols/therapeutic use , Tannins/therapeutic use , Animals , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , DNA Methylation , Disease Models, Animal , Drinking , Female , Flavonoids/blood , Intestinal Polyps/genetics , Intestinal Polyps/metabolism , Lipid Peroxidation/drug effects , Male , Mice , Mice, Inbred C57BL , Phenols/blood , Polyphenols
12.
Oncotarget ; 2(12): 1291-301, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22249198

ABSTRACT

The Notch signaling pathway drives proliferation, differentiation, apoptosis, cell fate, and maintenance of stem cells in several tissues. Aberrant activation of Notch signaling has been described in several tumours and in gastric cancer (GC), activated Notch1 has been associated with de-differentiation of lineage-committed stomach cells into stem progenitors and GC progression. However, the specific role of the Notch1 ligand DLL1 in GC has not yet been elucidated. To assess the role of DLL1 in GC cancer, the expression of Notch1 and its ligands DLL1 and Jagged1, was analyzed in 8 gastric cancer cell lines (KATOIII, SNU601, SNU719, AGS, SNU16, MKN1, MKN45, TMK1). DLL1 expression was absent in KATOIII, SNU601, SNU719 and AGS. The lack of DLL1 expression in these cells was associated with promoter hypermethylation and 5-aza-2'dC caused up-regulation of DLL1. The increase in DLL1 expression was associated with activation of Notch1 signalling, with an increase in cleaved Notch1 intracellular domain (NICD) and Hes1, and down-regulation in Hath1. Concordantly, Notch1 signalling was activated with the overexpression of DLL1. Moreover, Notch1 signalling together with DLL1 methylation were evaluated in samples from 52 GC patients and 21 healthy control as well as in INS-GAS mice infected with H. pylori and randomly treated with eradication therapy. In GC patients, we found a correlation between DLL1 and Hes1 expression, while DLL1 methylation and Hath1 expression were associated with the diffuse and mixed type of gastric cancer. Finally, none of the samples from INS-GAS mice infected with H. pylori, a model of intestinal-type gastric tumorigenesis, showed promoter methylation of DLL1. This study shows that Notch1 activity in gastric cancer is controlled by the epigenetic silencing of the ligand DLL1, and that Notch1 inhibition is associated with the diffuse type of gastric cancer.


Subject(s)
Epigenesis, Genetic , Intercellular Signaling Peptides and Proteins/genetics , Receptor, Notch1/metabolism , Stomach Neoplasms/genetics , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Calcium-Binding Proteins/genetics , Cell Differentiation/genetics , Cell Line, Tumor , DNA Methylation , Decitabine , Gene Expression Regulation, Neoplastic , Helicobacter Infections/pathology , Helicobacter pylori/pathogenicity , Homeodomain Proteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Membrane Proteins/genetics , Mice , Promoter Regions, Genetic , Receptor, Notch1/genetics , Serrate-Jagged Proteins , Signal Transduction/genetics , Stomach Neoplasms/metabolism , Transcription Factor HES-1
13.
Clin Cancer Res ; 16(23): 5703-11, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21030497

ABSTRACT

PURPOSE: Although cyclooxygenase (COX)-2 inhibitors could represent the most effective chemopreventive tool against colorectal cancer (CRC), their use in clinical practice is hampered by cardiovascular side effects. Consumption of ω-3-polyunsaturated fatty acids (ω-3-PUFAs) is associated with a reduced risk of CRC. Therefore, in this study, we assessed the efficacy of a novel 99% pure preparation of ω-3-PUFA eicosapentaenoic acid as free fatty acids (EPA-FFA) on polyps in Apc(Min/+) mice. EXPERIMENTAL DESIGN: Apc(Min/+) and corresponding wild-type mice were fed control diet (Ctrl) or diets containing either EPA-FFA 2.5% or 5%, for 12 weeks while monitoring food intake and body weight. RESULTS: We found that both EPA-FFA diets protected from the cachexia observed among Apc(Min/+) animals fed Ctrl diet (P < 0.0054), without toxic effect, in conjunction with a significant decrease in lipid peroxidation in the treated arms. Moreover, both EPA-FFA diets dramatically suppressed polyp number (by 71.5% and 78.6%, respectively; P < 0.0001) and load (by 82.5% and 93.4%, respectively; P < 0.0001) in both small intestine and colon. In addition, polyps less than 1 mm in size were predominantly found in the EPA-FFA 5% arm whereas those 1 to 3 mm in size were more frequent in the Ctrl arm (P < 0.0001). Interestingly, in the EPA-FFA groups, mucosal arachidonic acid was replaced by EPA (P < 0.0001), leading to a significant reduction in COX-2 expression and ß-catenin nuclear translocation. Moreover, in the EPA-FFA arms, we found a significant decrease in proliferation throughout the intestine together with an increase in apoptosis. CONCLUSIONS: Our data make 99% pure EPA-FFA an excellent candidate for CRC chemoprevention.


Subject(s)
Colonic Polyps/prevention & control , Eicosapentaenoic Acid/pharmacology , Eicosapentaenoic Acid/therapeutic use , Genes, APC , Animals , Carcinoma/genetics , Carcinoma/prevention & control , Colonic Polyps/diet therapy , Colonic Polyps/genetics , Colonic Polyps/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/prevention & control , Eicosapentaenoic Acid/chemistry , Eicosapentaenoic Acid/isolation & purification , Fatty Acids, Nonesterified/chemistry , Fatty Acids, Nonesterified/pharmacology , Fatty Acids, Nonesterified/therapeutic use , Genes, APC/physiology , Genetic Predisposition to Disease , Genotype , Lipid Peroxidation/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tumor Burden/drug effects
14.
Exp Cell Res ; 315(8): 1439-47, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19133256

ABSTRACT

Overexpressed cyclooxygenase-2 (COX-2) strongly contributes to the growth and invasiveness of tumoral cells in patients affected by colorectal cancer (CRC). It has been demonstrated that COX-2 overexpression depends on different cellular pathways involving both transcriptional and post-transcriptional regulations. We assumed that COX-2 expression could be regulated also by microRNAs (miRNAs) since these short RNA molecules participate to the fine regulation of several genes implicated in cell growth and differentiation. In this paper, we report the inverse correlation between COX-2 and miR-101 expression in colon cancer cell lines and we demonstrated in vitro the direct inhibition of COX-2 mRNA translation mediated by miR-101. Moreover, this correlation was supported by data collected ex vivo, in which colon cancer tissues and liver metastases derived from CRC patients were analyzed. These findings provide a novel molecular insight in the modulation of COX-2 at post-transcriptional level by miR-101 and strengthen the observation that miRNAs are highly implicated in the control of gene expression. An impairment of miR-101 levels could represent one of the leading causes of COX-2 overexpression in colon cancer cells.


Subject(s)
Colonic Neoplasms/physiopathology , Cyclooxygenase 2/metabolism , Down-Regulation , Gene Expression , MicroRNAs/metabolism , Base Sequence , Blotting, Western , Cell Line, Tumor , Humans , Molecular Sequence Data , Polymerase Chain Reaction , Protein Binding
15.
J Cell Mol Med ; 13(9B): 3876-87, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19017360

ABSTRACT

Inflammation promotes colorectal carcinogenesis. Tumour growth often generates a hypoxic environment in the inner tumour mass. We here report that, in colon cancer cells, the expression of the pro-inflammatory enzyme cyclooxygenase-2 (COX-2) associates with that of the hypoxia response gene carbonic anhydrase-IX (CA-IX). The COX-2 knockdown, achieved by the stable infection of a COX-2 specific short harpin RNA interference (shCOX-2), down-regulates CA-IX gene expression. In colorectal cancer (CRC) cells, PGE(2), the main COX-2 gene products, promotes CA-IX gene expression by ERK1/2 activation. In normoxic environment, shCOX-2 infected/CA-IX siRNA transfected CRC cells show a reduced level of active metalloproteinase-2 (MMP-2) that associates with a decreased extracellular matrix invasion capacity. In presence of hypoxia, COX-2 gene expression and PGE(2) production increase. The knockdown of COX-2/CA-IX blunts the survival capability of CRC cells in hypoxia. At a high cell density, a culture condition that creates a mild pericellular hypoxic environment, the expression of COX-2/CA-IX genes is increased and triggers the invasive potential of colon cancer cells. In human colon cancer tissues, COX-2/CA-IX protein expression levels, assessed by Western blot and immunohistochemistry, correlate each other and increase with tumour stage. In conclusion, these data indicate that COX-2/CA-IX interplay promotes the aggressive behaviour of CRC cells.


Subject(s)
Carbonic Anhydrases/biosynthesis , Colorectal Neoplasms/metabolism , Cyclooxygenase 2/biosynthesis , Up-Regulation , Adult , Aged , Aged, 80 and over , Caco-2 Cells , Cell Line, Tumor , Dinoprostone/metabolism , Female , Humans , Hypoxia , Male , Matrix Metalloproteinase 2/metabolism , Middle Aged , Neoplasm Invasiveness
SELECTION OF CITATIONS
SEARCH DETAIL
...