Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Gastric Cancer ; 27(4): 747-759, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38796558

ABSTRACT

BACKGROUND: CDH1 and CTNNA1 remain as the main genes for hereditary gastric cancer. However, they only explain a small fraction of gastric cancer cases with suspected inherited basis. In this study, we aimed to identify new hereditary genes for early-onset gastric cancer patients (EOGC; < 50 years old). METHODS: After germline exome sequencing in 20 EOGC patients and replication of relevant findings by gene-panel sequencing in an independent cohort of 152 patients, CTNND1 stood out as an interesting candidate gene, since its protein product (p120ctn) directly interacts with E-cadherin. We proceeded with functional characterization by generating two knockout CTNND1 cellular models by gene editing and introducing the detected genetic variants using a lentiviral delivery system. We assessed ß-catenin and E-cadherin levels, cell detachment, as well as E-cadherin localization and cell-to-cell interaction by spheroid modeling. RESULTS: Three CTNND1 germline variants [c.28_29delinsCT, p.(Ala10Leu); c.1105C > T, p.(Pro369Ser); c.1537A > G, p.(Asn513Asp)] were identified in our EOGC cohorts. Cells encoding CTNND1 variants displayed altered E-cadherin levels and intercellular interactions. In addition, the p.(Pro369Ser) variant, located in a key region in the E-cadherin/p120ctn binding domain, showed E-cadherin mislocalization. CONCLUSIONS: Defects in CTNND1 could be involved in germline predisposition to gastric cancer by altering E-cadherin and, consequently, cell-to-cell interactions. In the present study, CTNND1 germline variants explained 2% (3/172) of the cases, although further studies in larger external cohorts are needed.


Subject(s)
Cadherins , Catenins , Delta Catenin , Genetic Predisposition to Disease , Germ-Line Mutation , Stomach Neoplasms , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Humans , Male , Catenins/genetics , Catenins/metabolism , Female , Middle Aged , Adult , Cadherins/genetics , Cell Communication , Age of Onset , Antigens, CD
2.
Dig Dis Sci ; 67(8): 4049-4058, 2022 08.
Article in English | MEDLINE | ID: mdl-34387810

ABSTRACT

INTRODUCTION: Unlike colorectal cancer (CRC), few studies have explored the predictive value of genetic risk scores (GRS) in the development of colorectal adenomas (CRA), either alone or in combination with other demographic and clinical factors. METHODS: In this study, genomic DNA from 613 Spanish Caucasian patients with CRA and 829 polyp-free individuals was genotyped for 88 single-nucleotide polymorphisms (SNPs) associated with CRC risk using the MassArray™ (Sequenom) platform. After applying a multivariate logistic regression model, five SNPs were selected to calculate the GRS. Regression models adjusted by sex, age, family history of CRC, chronic use of NSAIDs, low-dose ASA, and consumption of tobacco were built in order to study the association between GRS and CRA risk. We evaluated the discriminatory capacity using the area under the receiver operating characteristic curve (AUC). The interactions between demographic information and GRS were also analyzed. RESULTS: Significant associations between high GRS values and risk of CRA for analyzed models were observed. In particular, patients with higher GRS values had 2.3-2.6-fold increase in risk of CRA compared to patients with middle values. Combining sex and age with the GRS significantly increased the discriminatory accuracy of the univariate model with GRS alone. The best model achieved an AUC value of 0.665 (95% CI: 0.63-0.69). The GRS showed a different behavior depending on sex and age. CONCLUSION: Our findings showed that, besides sex and age, GRS is an important risk factor for development of CRA and may be useful for CRC risk stratification and adaptation of screening programs.


Subject(s)
Adenoma , Colorectal Neoplasms , Adenoma/diagnosis , Adenoma/epidemiology , Adenoma/genetics , Case-Control Studies , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/genetics , Genetic Predisposition to Disease , Humans , Polymorphism, Single Nucleotide , Risk Factors
3.
Int J Cancer ; 144(3): 489-502, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30194776

ABSTRACT

Our study aimed to evaluate the relevance of genetic susceptibility in the development of colorectal adenomas (CRA) and its relationship with the presence of family history of colorectal cancer (CRC). Genomic DNA from 750 cases (first degree relatives of patients with CRC) and 750 controls (subjects with no family history of CRC) was genotyped for 99 single nucleotide polymorphisms (SNPs) previously associated with CRC/CRA risk by GWAS and candidate gene studies by using the MassArray™ (Sequenom) platform. Cases and controls were matched by gender, age and histological lesion. Eight hundred and fifty-eight patients showed no neoplastic lesions, whereas 288 patients showed low-risk adenomas, and 354 patients presented high-risk adenomas. Two SNPs (rs10505477, rs6983267) in the CASC8 gene were associated with a reduced risk of CRA in controls (log-additive models, OR: 0.67, 95%CI:0.54-0.83, and OR:0.66, 95%CI:0.54-0.84, respectively). Stratified analysis by histological lesion revealed the association of rs10505477 and rs6983267 variants with reduced risk of low- and high-risk adenomas in controls, being this effect stronger in low-risk adenomas (log-additive models, OR:0.63, 95%CI:0.47-0.84 and OR:0.64, 95%CI:0.47-0.86, respectively). Moreover, 2 SNPs (rs10795668, rs11255841) in the noncoding LINC00709 gene were significantly associated with a reduced risk of low-risk adenomas in cases (recessive models, OR:0.22, 95%CI:0.06-0.72, and OR:0.08, 95%CI:0.03-0.61) and controls (dominant models, OR:0.50, 95%CI:0.34-0.75, and OR:0.52, 95%CI:0.35-0.78, respectively). In conclusion, some variants associated with CRC risk (rs10505477, rs6983267, rs10795668 and rs11255841) are also involved in the susceptibility to CRA and specific subtypes. These associations are influenced by the presence of family history of CRC.


Subject(s)
Adenoma/genetics , Colorectal Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Case-Control Studies , Family Health , Female , Genetic Predisposition to Disease , Genetic Profile , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide
4.
Dig Dis Sci ; 61(10): 2896-2907, 2016 10.
Article in English | MEDLINE | ID: mdl-27343037

ABSTRACT

BACKGROUND AND AIM: Recent observational studies have shown therapeutic benefits of acetylsalicylic acid (ASA) in several types of cancer. We examined whether ASA exerts antitumor activity in esophageal adenocarcinoma (EAC). METHODS: Human EAC cells (OE33) were treated with ASA (0-5 mM) to evaluate proliferation, apoptosis, and migration. In vivo model: OE33-derived tumors were subcutaneously implanted into athymic mice which were allocated to ASA (5 or 50 mg/kg/day)/vehicle (5-6 animals/group). Tumor growth was assessed every 2-3 days, and after 40 days, mice were euthanized. Plasma drug levels were determined by high-performance liquid chromatography. Histological and immunohistochemical (Ki67, activated caspase-3) analysis of tumors were performed. The effect of ASA on tumor prostaglandin E2 (PGE2) levels was also evaluated. RESULTS: In vitro cell proliferation and migration were significantly inhibited while apoptosis increased (p < 0.05) by ASA. Although ASA did not induce tumor remission, tumor progression was significantly lower in ASA-treated mice when compared to non-treated animals (478 % in mice treated with 5 mg/kg/day ASA vs. 2696 % control; 748 % in mice treated with 50 mg/kg/day ASA vs. 2670 % control). Maximum tumor inhibition was 92 and 85 %, respectively. This effect was associated with a significant decrease of proliferation index in tumors. ASA 5 mg/kg/day did not modify tumor PGE2 levels. Whereas tumor PGE2 content in mice treated with ASA 50 mg/kg was lower than in control mice, the difference was not significant. CONCLUSION: Although these results need to be confirmed in other EAC cells, our data suggest a role for ASA in the treatment of this tumor.


Subject(s)
Adenocarcinoma/drug therapy , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Aspirin/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Esophageal Neoplasms/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Caspase 3/drug effects , Caspase 3/metabolism , Cell Line, Tumor , Chromatography, High Pressure Liquid , Dinoprostone/metabolism , Disease Progression , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Female , Humans , In Vitro Techniques , Ki-67 Antigen/drug effects , Ki-67 Antigen/metabolism , Mice , Mice, Nude , Xenograft Model Antitumor Assays
5.
Int J Cancer ; 137(6): 1362-73, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-25721731

ABSTRACT

Two recent genome-wide association studies in Asians have reported the association between the PSCA (prostate stem cell antigen) rs2294008C>T gene polymorphism and two Helicobacter pylori infection-related diseases such as gastric cancer (GC) and duodenal ulcer (DU). Since rs2294008 allele frequencies differ notably among ethnicities, we aimed to assess the role of rs2294008 on the susceptibility to GC and DU in a Caucasian population in Spain. Moreover, the relevance of rs2294008 on GC prognosis was evaluated. Genomic DNA from 603 Spanish patients with primary GC, 139 with DU and 675 healthy controls was typed for the PSCA rs2294008C>T polymorphism by PCR-TaqMan assays. H. pylori infection [odds ratio (OR): 8.27; 95% confidence interval (CI): 3.45-15.33] and nonsteroidal anti-inflammatory drugs (OR: 6.54; 95% CI: 3.19-12.43) were identified as independent risk factors for DU whereas the rs2294008T allele was associated with reduced risk of developing the disease (OR: 0.52; 95% CI: 0.33-0.82). Infection with CagA strains (OR: 2.10; 95% CI: 1.63-2.34), smoking (OR: 1.93; 95% CI: 1.54-2.61), family history of GC (OR: 2.83; 95% CI: 2.01-3.83), and the rs2294008T allele (OR: 1.46; 95% CI: 1.07-1.99) were associated with increased risk of GC. Interestingly, the association with the rs2294008T allele was restricted to noncardia GC (OR: 1.43; 95% CI: 1.12-1.82), particularly of the diffuse histotype (OR: 1.59; 95% CI: 1.16-1.92). Finally, Cox regression analysis identified the rs2294008T variant as a prognosis factor associated with worse overall survival in patients with diffuse-type GC (hazard ratio: 1.85; 95% CI: 1.12-3.06). From these results we conclude that the PSCA rs2294008 polymorphism is involved in the susceptibility to GC and DU, as well as in the prognosis of the diffuse-type of GC in Caucasians.


Subject(s)
Antigens, Neoplasm/genetics , Duodenal Ulcer/genetics , Genetic Predisposition to Disease/genetics , Neoplasm Proteins/genetics , Polymorphism, Genetic/genetics , Stomach Neoplasms/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Alleles , Case-Control Studies , Duodenal Ulcer/microbiology , Duodenal Ulcer/pathology , Female , GPI-Linked Proteins/genetics , Genome-Wide Association Study/methods , Helicobacter Infections/genetics , Helicobacter Infections/pathology , Helicobacter pylori , Humans , Male , Middle Aged , Odds Ratio , Prognosis , Risk , Risk Factors , Spain , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology , White People/genetics , Young Adult
6.
Gastroenterol Hepatol ; 37(7): 421-6, 2014.
Article in English | MEDLINE | ID: mdl-24661935

ABSTRACT

Gastric cancer (GC) is a complex disease and a worldwide health burden due to its high prevalence and poor prognosis. A deeper knowledge of the factors involved in the development and progression of GC could help to identify subpopulations at risk that therefore require surveillance or early treatment strategies. Current research is based on the study of genetic variants that confer a higher risk of GC and their interactions with environmental exposure. Recently, meta-analysis has emerged as an important statistical method involving pooling of data from individual association studies to increase statistical power and obtain more conclusive results. Given the importance of chronic inflammation in the process of gastric carcinogenesis, the present article reviews the most recent meta-analyses of the contribution of cytokine gene polymorphisms to GC risk.


Subject(s)
Genetic Predisposition to Disease , Meta-Analysis as Topic , Stomach Neoplasms/genetics , Humans , Risk , Stomach Neoplasms/epidemiology
7.
J Physiol Biochem ; 79(1): 147-161, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36342616

ABSTRACT

As a consequence of altered glucose metabolism, cancer cell intake is increased, producing large amounts of lactate which is pumped out the cytosol by monocarboxylate transporters (MCTs). MCT 1 and MCT4 are frequently overexpressed in tumors, and recently, MCT inhibition has been reported to exert antineoplastic effects. In the present study, MCT1 and MCT4 levels were assessed in esophageal adenocarcinoma (EAC) cells and the effects of the MCT-1 selective inhibitor AZD3965, hypoxia, and a glucose overload were evaluated in vitro. Two EAC cell lines (OE33 and OACM5.1C) were treated with AZD3965 (10-100 nM) under different conditions (normoxia/hypoxia) and also different glucose concentrations, and parameters of cytotoxicity, oxidative stress, intracellular pH (pHi), and lactate levels were evaluated. MCT1 was present in both cell lines whereas MCT4 was expressed in OE33 cells and only in a small proportion of OACM5.1C cells. Glucose addition did not have any effect on apoptosis nor cell proliferation. AZD3965 increased apoptosis and reduced proliferation of OACM5.1C cells, effects which were abrogated when cells were growing in hypoxia. MCT1 inhibition increased intracellular lactate levels in all the cells evaluated, but this increase was higher in cells expressing only MCT1 and did not affect oxidative stress. AZD3965 induced a decrease in pHi of cells displaying low levels of MCT4 and also increased the sodium/hydrogen exchanger 1 (NHE-1) expression on these cells. These data provide in vitro evidence supporting the potential of MCT inhibitors as novel antineoplastic drugs for EAC and highlight the importance of achieving a complete MCT inhibition.


Subject(s)
Adenocarcinoma , Antineoplastic Agents , Symporters , Humans , Symporters/metabolism , Hypoxia , Lactates , Monocarboxylic Acid Transporters/metabolism
8.
Clin Transl Gastroenterol ; 12(2): e00301, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33534415

ABSTRACT

INTRODUCTION: Epidemiological studies estimate that having a first-degree relative (FDR) with colorectal cancer (CRC) increases 2-fold to 3-fold the risk of developing the disease. Because FDRs of CRC patients are more likely to co-inherit CRC risk variants, we aimed to evaluate potential differences in genotype distribution of single nucleotide polymorphisms (SNPs) related to CRC risk between FDRs of patients with nonsyndromic CRC (cases) and individuals with no family history of CRC (controls). METHODS: We designed a case-control study comprising 750 cases and 750 Spanish Caucasian controls matched by sex, age, and histological findings after colonoscopy. Genomic DNA from all participants was genotyped for 88 SNPs associated with CRC risk using the MassArray (Sequenom) platform. RESULTS: Ten of the 88 SNPs analyzed revealed significant associations (P < 0.05) with a family history of CRC in our population. The most robust associations were found for the rs17094983G>A SNP in the long noncoding RNA LINC01500 (odds ratio = 0.72; 95% confidence interval: 0.58-0.88, log-additive model), and the rs11255841T>A SNP in the long noncoding RNA LINC00709 (odds ratio = 2.04; 95% confidence interval: 1.19-3.51, dominant model). Of interest, the observed associations were in the same direction than those reported for CRC risk. DISCUSSION: FDRs of CRC patients show significant differences in genotype distribution of SNPs related to CRC risk as compared to individuals with no family history of CRC. Genotyping of CRC risk variants in FDRs of CRC patients may help to identify subjects at risk that would benefit from stricter surveillance and CRC screening programs.


Subject(s)
Adenoma/genetics , Colorectal Neoplasms/genetics , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Adult , Aged , Aged, 80 and over , Case-Control Studies , Female , Humans , Male , Middle Aged , Risk Factors
9.
Scand J Gastroenterol ; 44(10): 1173-85, 2009.
Article in English | MEDLINE | ID: mdl-19711225

ABSTRACT

OBJECTIVE: Functional gastrointestinal disorders (FGID) may appear after acute gastroenteritis. The aim of this study was to evaluate the possible mechanisms (inflammation, visceral hypersensitivity, psychological and immunogenetic factors) related to the development of postinfectious (PI) FGID 3 years after a Salmonella outbreak. MATERIAL AND METHODS: Biopsies of the antrum, and right- and left colon from 16 PI-FGID patients, 8 PI control patients, and 18 healthy controls (H-controls) were processed for immunohistochemistry, cytokines, and mast-cell electron microscopy. DNA was typed for cytokine gene polymorphisms. Visceral sensitivity (satiety test and rectal barostat) and psychological factors (SCL-90 and vital events) were assessed. RESULTS: The number of mast cells and T lymphocytes was similar among the groups in all locations. Mast cells within 5 microm of nerve fibers of both PI groups were increased compared to H-controls: (stomach: 5.6+/-1.2 versus 6.6+/-1.5 versus 2.5+/-1.1; right colon: 9.7+/-1.3 versus 8.0+/-1.3 versus 4.1+/-1.7; left colon: 8.9+/-0.9 versus 8.5+/-1.8 versus 2.2+/-2.0 per field) (p<0.05). No differences in the production of IL-1beta, IL-1ra, IL-6, and IL-10 or in their genotypes were found. PI-FGID patients showed a lower pain threshold to rectal distention (29+/-2 versus 37+/- 2 mmHg; p<0.05). Scores for anxiety (0.63+/-0.11 versus 0.28+/-0.14) and somatization (1.01+/-0.15 versus 0.45+/-0.15) were higher in PI-FGID patients than in PI controls (p<0.05). The number of stressful life events was not significantly different between both PI groups. CONCLUSIONS: Three years after salmonellosis, PI-FGID patients showed no evidence of inflammation in the gastric or colonic mucosa, but visceral sensitivity and anxiety/somatization levels were increased. The close anatomical mast cell-nerve fibers relation does not seem to be related to the FGID but to the infection itself.


Subject(s)
Gastroenteritis/complications , Gastrointestinal Diseases/etiology , Gastrointestinal Diseases/pathology , Intestinal Mucosa/pathology , Salmonella Infections/complications , Adult , Aged , Case-Control Studies , Female , Humans , Male , Middle Aged
10.
Oncotarget ; 8(22): 35848-35862, 2017 May 30.
Article in English | MEDLINE | ID: mdl-28415781

ABSTRACT

Variations in DNA repair genes have been reported as key factors in gastric cancer (GC) susceptibility but results among studies are inconsistent. We aimed to assess the relevance of DNA repair gene polymorphisms and environmental factors to GC risk and phenotype in a Caucasian population in Spain. Genomic DNA from 603 patients with primary GC and 603 healthy controls was typed for 123 single nucleotide polymorphisms in DNA repair genes using the Illumina platform. Helicobacter pylori infection with CagA strains (odds ratio (OR): 1.99; 95% confidence interval (CI): 1.55-2.54), tobacco smoking (OR: 1.77; 95% CI: 1.22-2.57), and family history of GC (OR: 2.87; 95% CI: 1.85-4.45) were identified as independent risk factors for GC. By contrast, the TP53 rs9894946A (OR: 0.73; 95% CI: 0.56-0.96), TP53 rs1042522C (OR: 0.76; 95% CI: 0.56-0.96), and BRIP1 rs4986764T (OR: 0.55; 95% CI: 0.38-0.78) variants were associated with lower GC risk. Significant associations with specific anatomopathological GC subtypes were also observed, most notably in the ERCC4 gene with the rs1799801C, rs2238463G, and rs3136038T variants being inversely associated with cardia GC risk. Moreover, the XRCC3 rs861528 allele A was significantly increased in the patient subgroup with diffuse GC (OR: 1.75; 95% CI: 1.30-2.37). Our data show that specific TP53, BRIP1, ERCC4, and XRCC3 polymorphisms are relevant in susceptibility to GC risk and specific subtypes in Caucasians.


Subject(s)
DNA Repair/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Polymorphism, Genetic , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Alleles , Case-Control Studies , Combined Modality Therapy , Female , Gene Frequency , Haplotypes , Humans , Linkage Disequilibrium , Male , Middle Aged , Neoplasm Staging , Odds Ratio , Phenotype , Risk , Stomach Neoplasms/mortality , Stomach Neoplasms/therapy
11.
BMC Gastroenterol ; 6: 7, 2006 Feb 16.
Article in English | MEDLINE | ID: mdl-16483364

ABSTRACT

BACKGROUND & AIM: The role that H. pylori infection plays in the development of and Barrett's esophagus (BE) is uncertain. We tested the hypothesis that infection with cagA+ Helicobacter pylori strains protects against the development of BE. METHODS: We studied 104 consecutive patients, residents in an area with a high prevalence of H. pylori infection, with BE and 213 sex- and age-matched controls. H. pylori infection and CagA antibody status were determined by western blot serology. RESULTS: H. pylori prevalence was higher in patients with BE than in controls (87.5% vs. 74.6%; OR. 2.3; 95% CI: 1.23-4.59). Increasing age was associated with a higher prevalence of H. pylori (p < 0.05). The prevalence of CagA+ H. pylori serology was similar in patients with BE and controls (64.4% vs. 54.5%; NS). Type I H. pylori infection (CagA+ and VacA+) was similar in patients with BE and controls (44.2% vs. 41.3%; NS). Logistic regression analysis identified alcohol (O.R. 7.09; 95% CI 2.23-22.51), and H. pylori infection (OR: 2.41; 95%CI: 1.20-4.84) but not CagA+ serology as independent factors. CONCLUSION: Neither H. pylori infection nor H. pylori infection by CagA+ strains reduce the risk of BE in a population with high prevalence of H. pylori infection.


Subject(s)
Antigens, Bacterial/blood , Bacterial Proteins/blood , Barrett Esophagus/epidemiology , Helicobacter Infections/epidemiology , Helicobacter pylori , Adult , Age Factors , Aged , Alcohol Drinking , Barrett Esophagus/complications , Barrett Esophagus/diagnosis , Blotting, Western , Endoscopy , Female , Helicobacter Infections/blood , Helicobacter Infections/complications , Helicobacter Infections/diagnosis , Humans , Logistic Models , Male , Middle Aged , Prevalence , Risk Factors , Sex Factors , Smoking , Spain/epidemiology
12.
Sci Rep ; 5: 7988, 2015 Jan 23.
Article in English | MEDLINE | ID: mdl-25614381

ABSTRACT

Recently, differential scanning calorimetry (DSC) has been acknowledged as a novel tool for diagnosing and monitoring several diseases. This highly sensitive technique has been traditionally used to study thermally induced protein folding/unfolding transitions. In previous research papers, DSC profiles from blood samples of patients were analyzed and they exhibited marked differences in the thermal denaturation profile. Thus, we investigated the use of this novel technology in blood serum samples from 25 healthy subjects and 30 patients with gastric adenocarcinoma (GAC) at different stages of tumor development with a new multiparametric approach. The analysis of the calorimetric profiles of blood serum from GAC patients allowed us to discriminate three stages of cancer development (I to III) from those of healthy individuals. After a multiparametric analysis, a classification of blood serum DSC parameters from patients with GAC is proposed. Certain parameters exhibited significant differences (P < 0.05) and allowed the discrimination of healthy subjects/patients from patients at different tumor stages. The results of this work validate DSC as a novel technique for GAC patient classification and staging, and offer new graphical tools and value ranges for the acquired parameters in order to discriminate healthy from diseased subjects with increased disease burden.


Subject(s)
Adenocarcinoma/blood , Adenocarcinoma/diagnosis , Blood Proteins/chemistry , Calorimetry, Differential Scanning , Stomach Neoplasms/blood , Stomach Neoplasms/diagnosis , Biomarkers , Case-Control Studies , Humans , Neoplasm Staging , Protein Denaturation , Protein Folding , Protein Unfolding , Spain , Thermography
13.
Oncol Rep ; 31(6): 2785-91, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24737143

ABSTRACT

Aspirin has been proposed in recent years as a candidate for chemoprevention of adenocarcinoma in patients with Barrett's esophagus. The aim of the present study was to evaluate the effect of acetylsalicylic acid (ASA) in an experimental model of esophageal adenocarcinoma. An animal model of gastroenteroesophageal reflux was established using Wistar rats undergoing esophagojejunostomy with gastric preservation. Following surgery, rats were divided into three groups: i) control (vehicle); ii) ASA 50 mg/kg/day; and iii) ASA 5 mg/kg/day. Four months after surgery, the surviving animals were sacrificed and the rat esophagi were assessed for histological and biochemical [prostaglandin E2 (PGE2) and lipoxin A4 (LXA4 ) levels] analysis. As in the control rats, those receiving aspirin treatment showed no decrease in inflammation grade, extent of ulcerated esophageal mucosa, length of intestinal metaplasia in continuity with anastomosis, presence of intestinal metaplasia beyond anastomosis, severity of dysplasia or incidence of adenocarcinoma. In contrast, aspirin-treated rats showed decreased esophageal tissue levels of PGE2 and increased LXA4, significantly in the high-dose aspirin group (p=0.008 and p=0.01, respectively). In this rat model of gastroesophageal reflux, the administration of aspirin modified esophageal tissue levels of PGE2 and LXA4, but was not effective in preventing the development of esophageal adenocarcinoma.


Subject(s)
Adenocarcinoma/drug therapy , Aspirin/administration & dosage , Barrett Esophagus/drug therapy , Esophageal Neoplasms/drug therapy , Gastroesophageal Reflux/drug therapy , Neoplasms, Experimental/drug therapy , Adenocarcinoma/pathology , Animals , Barrett Esophagus/pathology , Dinoprostone/biosynthesis , Disease Models, Animal , Esophageal Neoplasms/pathology , Gastroesophageal Reflux/pathology , Gastroesophageal Reflux/surgery , Gene Expression Regulation, Neoplastic , Humans , Lipoxins/biosynthesis , Neoplasms, Experimental/pathology , Neoplasms, Experimental/surgery , Rats
14.
World J Gastroenterol ; 18(35): 4866-74, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-23002358

ABSTRACT

AIM: To evaluate the effects of indomethacin [dual cyclooxygenase (COX)-1/COX-2 inhibitor] and 3-(3,4-difluorophenyl)-4-(4-(methylsulfonyl) phenyl)-2-(5H)-furanone (MF-tricyclic) (COX-2 selective inhibitor) in a rat experimental model of Barrett's esophagus and esophageal adenocarcinoma. METHODS: A total of 112 surviving post-surgery rats were randomly divided into three groups: the control group (n = 48), which did not receive any treatment; the indomethacin group (n = 32), which were given 2 mg/kg per day of the COX-1/COX-2 inhibitor; and the MF-tricyclic group (n = 32), which received 10 mg/kg per day of the selective COX-2 inhibitor. Randomly selected rats were killed either 8 wk or 16 wk after surgery. The timing of the deaths was in accordance with a previous study performed in our group. Only rats that were killed at the times designated by the protocol were included in the study. We then assessed the histology and prostaglandin E2 (PGE2) expression levels in the rat esophagi. An additional group of eight animals that did not undergo esophagojejunostomy were included in order to obtain normal esophageal tissue as a control. RESULTS: Compared to a control group with no treatment (vehicle-treated rats), indomethacin treatment was associated with decreases in ulcerated esophageal mucosa (16% vs 35% and 14% vs 17%, 2 mo and 4 mo after surgery, respectively; P = 0.021), length of intestinal metaplasia in continuity with anastomosis (2 ± 1.17 mm vs 2.29 ± 0.75 mm and 1.25 ± 0.42 mm vs 3.5 ± 1.54 mm, 2 mo and 4 mo after surgery, respectively; P = 0.007), presence of intestinal metaplasia beyond anastomosis (20% vs 71.4% and 0% vs 60%, 2 mo and 4 mo after surgery, respectively; P = 0.009), severity of dysplasia (0% vs 71.4% and 20% vs 85.7% high-grade dysplasia, 2 mo and 4 mo after surgery, respectively; P = 0.002), and adenocarcinoma incidence (0% vs 57.1% and 0% vs 60%, 2 mo and 4 mo after surgery, respectively; P < 0.0001). Treatment with the selective COX-2 inhibitor, MF-tricyclic, did not prevent development of intestinal metaplasia or adenocarcinoma. In parallel, we observed a significant decrease in PGE2 levels in indomethacin-treated rats, but not in those treated with MF-tricyclic, at both 2 mo and 4 mo. Compared to control rats that did not undergo surgery (68 ± 8 ng/g, P = 0.0022 Kruskal-Wallis test) there was a significant increase in PGE2 levels in the esophageal tissue of the rats that underwent surgery either 2 mo (1332 ± 656 ng/g) or 4 mo (1121 ± 1015 ng/g) after esophagojejunostomy. However, no differences were found when esophageal PGE2 levels were compared 2 mo vs 4 mo post-esophagojejunostomy. At both the 2- and 4-mo timepoints, we observed a significant decrease in PGE2 levels in indomethacin-treated rat esophagi compared to those in either the control or MF-tricyclic groups (P = 0.049 and P = 0.017, respectively). No differences in PGE2 levels were found when we compared levels in rats treated with MF-tricyclic to not-treated rats. CONCLUSION: In this rat model of gastrointestinal reflux, indomethacin was associated with a decrease in the severity of esophagitis and reduced development of esophageal intestinal metaplasia and adenocarcinoma.


Subject(s)
Adenocarcinoma/prevention & control , Anticarcinogenic Agents/pharmacology , Barrett Esophagus/prevention & control , Cell Transformation, Neoplastic/drug effects , Cyclooxygenase Inhibitors/pharmacology , Esophageal Neoplasms/prevention & control , Esophagus/drug effects , Gastroesophageal Reflux/drug therapy , Indomethacin/pharmacology , Membrane Proteins/antagonists & inhibitors , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Animals , Anticarcinogenic Agents/blood , Barrett Esophagus/enzymology , Barrett Esophagus/pathology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase Inhibitors/blood , Dinoprostone/metabolism , Disease Models, Animal , Disease Progression , Esophageal Neoplasms/enzymology , Esophageal Neoplasms/pathology , Esophagitis/enzymology , Esophagitis/pathology , Esophagitis/prevention & control , Esophagus/enzymology , Esophagus/pathology , Esophagus/surgery , Female , Furans/pharmacology , Gastroesophageal Reflux/enzymology , Gastroesophageal Reflux/pathology , Indomethacin/blood , Membrane Proteins/metabolism , Metaplasia , Mucous Membrane/drug effects , Mucous Membrane/enzymology , Mucous Membrane/pathology , Rats , Rats, Wistar , Time Factors
15.
PLoS One ; 7(9): e46179, 2012.
Article in English | MEDLINE | ID: mdl-23029430

ABSTRACT

BACKGROUND: Genetic factors influencing the prognosis of gastric adenocarcinoma (GAC) are not well known. Given the relevance of cytokines and other pro-inflammatory mediators in cancer progression and invasiveness, we aimed to assess the prognostic role of several functional cytokine and cyclooxygenase gene polymorphisms in patients with GAC. METHODOLOGY: Genomic DNA from 380 Spanish Caucasian patients with primary GAC was genotyped for 23 polymorphisms in pro-inflammatory (IL1B, TNFA, LTA, IL6, IL12p40), anti-inflammatory (IL4, IL1RN, IL10, TGFB1) cytokine, and cyclooxygenase (PTGS1 and PTGS2) genes by PCR, RFLP and TaqMan assays. Clinical and histological information was collected prospectively. Survival curves were estimated by the Kaplan-Meier method and compared using the log rank test. Outcome was determined by analysis of Cox proportional hazards, adjusting for confounding factors. RESULTS: The median follow-up period and median overall survival (OS) time were 9.9 months (range 0.4-120.3) and 10.9 months (95% CI: 8.9-14.1), respectively. Multivariate analysis identified tumor stages III (HR, 3.23; 95% CI:2-5.22) and IV (HR, 5.5; 95% CI: 3.51-8.63) as independent factors associated with a significantly reduced OS, whereas surgical treatment (HR: 0.44; 95%CI: 0.3-0.6) was related to a better prognosis of the disease. Concerning genetic factors, none of the 23 polymorphisms evaluated in the current study did influence survival. Moreover, no gene-environment interactions on GAC prognosis were observed. CONCLUSIONS: Our results show that, in our population, the panel of selected pro- and anti-inflammatory cytokine, and cyclooxygenase gene polymorphisms are not relevant in determining the prognosis of gastric adenocarcinoma.


Subject(s)
Adenocarcinoma/genetics , Cyclooxygenase 1/genetics , Cyclooxygenase 2/genetics , Cytokines/biosynthesis , Stomach Neoplasms/genetics , White People , Adenocarcinoma/diagnosis , Adenocarcinoma/mortality , Adenocarcinoma/surgery , Aged , Aged, 80 and over , Cohort Studies , Cytokines/genetics , Female , Gene-Environment Interaction , Genotype , Humans , Male , Middle Aged , Neoplasm Staging , Polymorphism, Restriction Fragment Length , Prognosis , Risk Factors , Stomach Neoplasms/diagnosis , Stomach Neoplasms/mortality , Stomach Neoplasms/surgery , Survival Rate
16.
Med Clin (Barc) ; 137(12): 555-60, 2011 Nov 12.
Article in Spanish | MEDLINE | ID: mdl-21208631

ABSTRACT

Gastric cancer is a complex pathology which represents a worldwide health burden due to its high prevalence and poor prognosis. Therefore, prevention and early detection are considered the best options for controlling the disease. Current research is based on the study of genetic variants that confer a higher risk and their interactions with environmental exposure. A deeper knowledge of factors involved on gastric cancer development may allow identification of those individuals at elevated risk and will provide useful predictive information for subgroups of patients who need surveillance or early treatment strategies. In the present paper we review in a comprehensive manner, the most recent published studies on the contribution of gene polymorphisms and Helicobacter pylori infection to gastric cancer risk.


Subject(s)
Gastritis/immunology , Helicobacter Infections/immunology , Helicobacter pylori/immunology , Polymorphism, Single Nucleotide , Stomach Neoplasms/epidemiology , Cocarcinogenesis , Cyclooxygenase 2/genetics , Cytokines/genetics , Cytokines/metabolism , Gastritis/epidemiology , Gastritis/microbiology , Genetic Predisposition to Disease , Helicobacter Infections/epidemiology , Humans , Inflammation Mediators/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Nitric Oxide/metabolism , Penetrance , Risk , Stomach Neoplasms/etiology , Stomach Neoplasms/microbiology
17.
Hum Immunol ; 72(9): 749-52, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21641950

ABSTRACT

Idiopathic achalasia is an esophageal motor disorder of unknown etiology. A wealth of evidence supports the concept that achalasia is an immune-mediated disease. According to this evidence, achalasia has been significantly associated with specific alleles of the human leukocyte antigen class II, PTPN22 and IL23R. Several studies have demonstrated association of the IL10 gene with different inflammatory disorders. Our aim was to evaluate the role of functional IL10 promoter polymorphisms in susceptibility to idiopathic achalasia. A case-control study was performed with the -1082, -819, and -592 IL10 promoter polymorphisms in 282 patients and 529 controls and in an independent replication set of 75 patients and 575 controls. The GCC haplotype of the IL10 promoter was reported to be associated with a lower risk of achalasia in the discovery sample (odds ratio [OR] = 0.79, 95% confidence interval [CI] = 0.64-0.98, p = 0.029). This association was validated in a replication set (OR = 0.69, 95% CI = 0.48-1.00, p = 0.04). In the combined analysis no heterogeneity was observed between the 2 sample sets and the GCC haplotype was significantly associated with the disease (OR(MH) = 0.76, 95% CI = 0.63-0.91, p = 0.003). Our results provide the first evidence for an association between IL10 promoter polymorphisms and idiopathic achalasia, suggesting that the interleukin-10 cytokine may contribute to the pathogenesis of this disease.


Subject(s)
Esophageal Achalasia/genetics , Interleukin-10/genetics , Adolescent , Adult , Aged , Aged, 80 and over , DNA Mutational Analysis , Female , Genetic Association Studies , Genetic Predisposition to Disease , Haplotypes , Humans , Immunity/genetics , Male , Middle Aged , Polymorphism, Genetic , Promoter Regions, Genetic , Spain
18.
Clin Ther ; 30(1): 121-30, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18343248

ABSTRACT

BACKGROUND: Previous studies have reported a potential genetic predisposition to NSAID-related upper gastrointestinal (GI) bleeding. OBJECTIVE: This study evaluated whether there was an association between 2 polymorphisms--the platelet glycoprotein (GP) IIIa PlA1/A2 polymorphism and the 27-bp VNTR (variable number of tandem repeats) polymorphism in intron 4 of the endothelial nitric oxide synthase (eNOS) gene--and a risk for nonvariceal upper GI bleeding in Spanish patients taking low-dose aspirin for secondary prophylaxis of vascular occlusive diseases. METHODS: Genotyping for the 2 polymorphisms was performed in patients hospitalized for upper GI bleeding associated with the use of low-dose aspirin between September 1998 and October 2000, and race-, age-, and sex-matched controls who were taking low-dose aspirin but had no history of upper GI bleeding. To ascertain allele frequencies in a healthy population, genotyping was also performed in an unmatched group of blood donors. RESULTS: The study included 88 white patients (65 men, 23 women; mean age, 67.5 years) with an episode of upper GI bleeding, 108 matched controls with no history of upper GI bleeding (79 men, 29 women; mean age, 65.9 years), and 158 blood-donor controls (109 men, 49 women; mean age, 53.4 years). No significant differences were found between cases and controls in terms of genotype, carriage, or allele frequency of the GPIIIa PlA1/A2 polymorphism. However, after adjustment for confounding variables, logistic regression analysis indicated an association between carriage of the eNOS "a" allele and a reduced risk of upper GI bleeding (odds ratio [OR] 0.39; 95 CI, 0.18-0.85; P 0.018). In this model, treatment with nitrovasodilators (OR 0.28; 95 CI, 0.12-0.66; P 0.004) and use of antisecretory drugs (OR 0.15; 95 CI, 0.05-0.47; P 0.001) were also identified as protective factors. Helicobacter pylori infection (OR 3.07; 95 CI, 1.23-7.70; P 0.017), alcohol consumption (OR 5.04; 95 CI, 1.86-13.70; P 0.001), and a history of peptic ulcer (OR 13.41; 95 CI, 3.78-47.64; P 0.001) were identified as risk factors for upper GI bleeding. CONCLUSION: In this small, selected population of individuals taking low-dose aspirin for secondary prevention, carriage of the "a" allele of the eNOS gene was associated with a decreased risk for upper GI bleeding.


Subject(s)
Aspirin/adverse effects , Gastrointestinal Hemorrhage/genetics , Integrin beta3/genetics , Nitric Oxide Synthase Type III/genetics , Platelet Aggregation Inhibitors/adverse effects , Polymorphism, Genetic , Aged , Alcohol Drinking/adverse effects , Aspirin/administration & dosage , Case-Control Studies , Female , Gastrointestinal Hemorrhage/chemically induced , Gene Frequency , Genetic Predisposition to Disease , Genotype , Helicobacter Infections/complications , Humans , Introns , Male , Middle Aged , Peptic Ulcer/complications , Platelet Aggregation Inhibitors/administration & dosage , Risk Factors , Vasodilator Agents/therapeutic use
19.
Am J Gastroenterol ; 102(9): 1878-92, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17640324

ABSTRACT

BACKGROUND AND AIMS: Recent studies have reported an association between cytokine gene polymorphisms and GC risk. However, results are inconsistent among studies from different geographic regions and ethnic groups. Our goal was to evaluate the influence of Helicobacter pylori (H. pylori) infection and host genetic factors on GC susceptibility in a population of Spanish white GC patients. METHODS: DNA from 404 unrelated patients with GC and 404 sex- and age-matched healthy controls was typed for several functional polymorphisms in pro- (IL-1B, TNFA, LTA, IL-12p40) and anti-inflammatory (IL-4, IL-1RN, IL-10, TGFB1) genes by PCR, RFLP, and TaqMan assays. H. pylori infection and CagA/VacA antibody status were also determined by western blot serology. RESULTS: Logistic regression analysis identified H. pylori infection with cagA strains (OR 2.54, 95% CI 1.77-3.66), smoking habit (OR 1.91, 95% CI 1.25-2.93), and positive family history of GC (OR 3.67, 95% CI 2.01-6.71) as independent risk factors for GC. None of the cytokine gene polymorphisms analyzed in this study were associated with susceptibility to GC development, whether GC patients were analyzed as a group or categorized according to anatomic location or histological subtype. Some simultaneous combinations of proinflammatory genotypes reportedly associated with greater GC risk yielded no significant differences between patients and controls. CONCLUSIONS: Our results show that, at least in some white populations, the contribution of the cytokine gene polymorphisms evaluated in this study (IL-1B, IL-1RN, IL-12p40, LTA, IL-10, IL-4, and TGF-B1) to GC susceptibility may be less relevant than previously reported.


Subject(s)
Cytokines/genetics , Genetic Predisposition to Disease , Polymorphism, Genetic , Stomach Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Antigens, Bacterial/analysis , Bacterial Proteins/analysis , Female , Genotype , Haplotypes , Helicobacter Infections/complications , Helicobacter Infections/diagnosis , Helicobacter pylori/genetics , Humans , Male , Middle Aged , Spain , Stomach Neoplasms/metabolism , Stomach Neoplasms/microbiology , White People
20.
Am J Gastroenterol ; 101(9): 1979-84, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16848803

ABSTRACT

BACKGROUND: Our group previously reported the absence of nitric oxide synthase (NOS) in the gastroesophageal junction of patients with achalasia. NOS exists in three distinct isoforms: neuronal NOS (nNOS), endothelial NOS (eNOS), and inducible isoform (iNOS). Some studies have shown that NO production is regulated by NOS polymorphisms. AIM: To assess whether some functional polymorphisms in the nNOS, iNOS, or eNOS genes are involved in susceptibility to suffer from achalasia. METHODS: Genomic DNA from 80 unrelated Spanish Caucasian patients with sporadic achalasia and 144 healthy subjects matched for age (+/-5 yr) and gender was typed by PCR and RFLP methods for the 27-bp variable number of tandem repeat (VNTR) polymorphism in intron 4 of the eNOS gene, a CA microsatellite repeat and a Nla III (C-->T) restriction fragment length polymorphism (RFLP) in exon 29 of the nNOS gene, and two nucleotide substitutions located in exon 16 (C-->T) and exon 22 (G-->A) of the iNOS gene. RESULTS: No significant differences in carriage, genotype, and allele frequencies of the nNOS, iNOS, or eNOS gene polymorphisms were found between patients with achalasia and controls. Individuals homozygous for genotype iNOS22*A/A tended to be more frequent in achalasia (20%vs 11%, odds ratio [OR] 1.79, 95% confidence interval [CI] 0.89-3.59, p= 0.09) as were those homozygous for the rare eNOS*4a allele (6.2%vs 1.4%, OR 4.5, 95% CI 0.89-22.67, p= 0.1) although the difference did not reach statistical significance. No differences in genotype and allele distribution were found with respect to epidemiological and clinical characteristics of patients with achalasia. CONCLUSION: Our data suggest that NOS gene polymorphisms are not involved in the susceptibility to and nature of the clinical course of sporadic achalasia. However, studies in a greater number of patients are required to analyze the tendency toward a higher prevalence of genotypes iNOS22*A/A and eNOS*4a4a.


Subject(s)
DNA/genetics , Esophageal Achalasia/enzymology , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type I/genetics , Polymorphism, Genetic , Alleles , Esophageal Achalasia/genetics , Esophageal Achalasia/physiopathology , Female , Gene Frequency , Genotype , Humans , Male , Manometry , Middle Aged , Minisatellite Repeats , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length , Pressure , Severity of Illness Index
SELECTION OF CITATIONS
SEARCH DETAIL