Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Genes (Basel) ; 13(11)2022 11 04.
Article in English | MEDLINE | ID: mdl-36360266

ABSTRACT

Gastric cancer is the subject of clinical and basic studies due to its high incidence and mortality rates worldwide. Due to the diagnosis occurring in advanced stages and the classic treatment methodologies such as gastrectomy and chemotherapy, they are extremely aggressive and limit the quality of life of these patients. CRISPR/Cas9 is a tool that allows gene editing and has been used to explore the functions of genes related to gastric cancer, in addition to being used in the treatment of this neoplasm, greatly increasing our understanding of cancer genomics. In this mini-review, we seek the current status of the CRISPR/Cas9 gene-editing technology in gastric cancer research and clinical research.


Subject(s)
Gene Editing , Stomach Neoplasms , Humans , Gene Editing/methods , CRISPR-Cas Systems/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/therapy , Quality of Life , Technology
2.
Anticancer Res ; 42(9): 4381-4394, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36039443

ABSTRACT

BACKGROUND/AIM: Previous studies from our research group have shown that trisomy 8 and the amplification of the 8q24.21 region is very frequent in gastric cancer (GC). Little is known about the role of most genes located in this region. Thus, the aim of this study was to understand the possible impact of transcriptional alterations and copy number variation (CNV) of four genes located in the 8q24.21 region - FAM49B, FAM84B, GSDMC and miR-5194 - in GC. MATERIALS AND METHODS: Fifty-one to 85 matched pairs of tumoral and adjacent non-tumoral gastric tissues, from patients with primary GC, were used to analyze gene expression and CNV of the selected genes. We also included 29 H. pylori negative and gastritis negative gastric mucosa tissues from individuals without cancer obtained by endoscopy, as control samples. RESULTS: The expression of FAM49B, GSDMC and miR-5194 was higher in both tumoral and adjacent non-tumoral samples compared to the negative control. The expression of FAM84B showed no significant difference between tumoral samples and negative controls. However, the expression of FAM84B in the adjacent non-tumoral samples was higher compared to negative control and tumoral samples. Moreover, the higher expression of GSDMC was associated with T3 and T4 tumors, with tumors on stage III and IV and with advanced tumors. Higher copy numbers of FAM49B and GSDMC were associated with intestinal tumor type and with moderately or well-differentiated tumors. Higher copy number of FAM84B was associated with moderately or well-differentiated tumors. Furthermore, the expression of all four genes was positively correlated. CONCLUSION: All four genes are upregulated in GC and may play an important role in these neoplasms. GSDMC expression was associated with more aggressive tumors.


Subject(s)
MicroRNAs , Stomach Neoplasms , Biomarkers, Tumor/genetics , Chromosomes, Human, Pair 8 , DNA Copy Number Variations/genetics , DNA-Binding Proteins/genetics , Gastric Mucosa/pathology , Humans , MicroRNAs/genetics , Pore Forming Cytotoxic Proteins , Stomach Neoplasms/pathology
3.
Epigenetics ; 17(1): 110-116, 2022 01.
Article in English | MEDLINE | ID: mdl-33491552

ABSTRACT

Gastric cancer (GC) is one of the leading types of fatal cancer worldwide. Epigenetic manipulation of cancer cells is a useful tool to better understand gene expression regulatory mechanisms and contributes to the discovery of novel biomarkers. Our research group recently reported a list of 83 genes that are potentially modulated by DNA methylation in GC cell lines. Herein, we further explored the regulation of one of these genes, LRRC37A2, in clinical samples. LRRC37A2 expression was evaluated by RT-qPCR, and DNA methylation was studied using next-generation bisulphite sequencing in 36 GC and paired adjacent nonneoplastic tissue samples. We showed that both reduced LRRC37A2 mRNA levels and increased LRRC37A2 exon methylation were associated with undifferentiated and poorly differentiated tumours. Moreover, LRRC37A2 gene expression and methylation levels were inversely correlated at the +45 exon CpG site. We suggest that DNA hypermethylation may contribute to reducing LRRC37A2 expression in undifferentiated and poorly differentiated GC. Therefore, our results show how some genes may be useful to stratify patients who are more likely to benefit from epigenetic therapy.Abbreviations: AR: androgen receptor; 5-AZAdC: 5-aza-2'-deoxycytidine; B2M: beta-2-microglobulin; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GC: gastric cancer; GLM: general linear model; LRRC37A2: leucine-rich repeat containing 37 member A2; SD: standard deviation; TFII-I: general transcription factor II-I; TSS: transcription start site; XBP1: X-box binding protein 1.


Subject(s)
DNA Methylation , Stomach Neoplasms , Cell Line, Tumor , CpG Islands , Decitabine , Gene Expression Regulation, Neoplastic , Humans , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism
4.
J Mol Med (Berl) ; 98(5): 707-717, 2020 05.
Article in English | MEDLINE | ID: mdl-32285140

ABSTRACT

Gastric cancer (GC) is the third leading cause of cancer-related death worldwide. Very few therapeutic options are currently available in this neoplasia. The use of 5-Aza-2'-deoxycytidine (5-AZAdC) was approved for the treatment of myelodysplastic syndromes, and this drug can treat solid tumours at low doses. Epigenetic manipulation of GC cell lines is a useful tool to better understand gene expression regulatory mechanisms for clinical applications. Therefore, we compared the gene expression profile of 5-AZAdC-treated and untreated GC cell lines by a microarray assay. Among the genes identified in this analysis, we selected NRN1 and TNFAIP3 to be evaluated for gene expression by RT-qPCR and DNA methylation by bisulfite DNA next-generation sequencing in 43 and 52 pairs of GC and adjacent non-neoplastic tissue samples, respectively. We identified 83 candidate genes modulated by DNA methylation in GC cell lines. Increased expression of NRN1 and TNFAIP3 was associated with advanced tumours (P < 0.05). We showed that increased NRN1 and TNFAIP3 expression seems to be regulated by DNA demethylation in GC samples: inverse correlations between the mRNA and DNA methylation levels in the promoter of NRN1 (P < 0.05) and the intron of TNFAIP3 (P < 0.05) were detected. Reduced NRN1 promoter methylation was associated with III/IV TNM stage tumours (P = 0.03) and the presence of Helicobacter pylori infection (P = 0.02). The identification of demethylated activated genes in GC may be useful in clinical practice, stratifying patients who are less likely to benefit from 5-AZAdC-based therapies. KEY MESSAGES: Higher expression of NRN1 and TNFAIP3 is associated with advanced gastric cancer (GC). NRN1 promoter hypomethylation contributes to gene upregulation in advanced GC. TNFAIP3 intronic-specific CpG site demethylation contributes to gene upregulation in GC. These findings may be useful to stratify GC patients who are less likely to benefit from DNA demethylating-based therapies.


Subject(s)
DNA Demethylation , Gene Expression Regulation, Neoplastic , Neuropeptides/genetics , Stomach Neoplasms/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Azacitidine/pharmacology , Biomarkers, Tumor , Cell Line, Tumor , Computational Biology/methods , CpG Islands , DNA Methylation , Decitabine/pharmacology , Epigenesis, Genetic , GPI-Linked Proteins/genetics , Gene Expression Profiling , Humans , Neoplasm Staging , Prognosis , Stomach Neoplasms/pathology , Transcriptome
5.
Int J Mol Sci ; 21(5)2020 Mar 05.
Article in English | MEDLINE | ID: mdl-32150871

ABSTRACT

Despite the advancements in cancer treatments, gastric cancer is still one of the leading causes of death worldwide. In this context, it is of great interest to discover new and more effective ways of treating this disease. Accumulated evidences have demonstrated the amplification of 8q24.21 region in gastric tumors. Furthermore, this is the region where the widely known MYC oncogene and different microRNAs are located. MYC deregulation is key in tumorigenesis in various types of tissues, once it is associated with cell proliferation, survival, and drug resistance. microRNAs are a class of noncoding RNAs that negatively regulate the protein translation, and which deregulation is related with gastric cancer development. However, little is understood about the interactions between microRNAs and MYC. Here, we overview the MYC role and its relationship with the microRNAs network in gastric cancer aiming to identify potential targets useful to be used in clinic, not only as biomarkers, but also as molecules for development of promising therapies.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Proto-Oncogene Proteins c-myc/metabolism , Stomach Neoplasms/pathology , Animals , Humans , Proto-Oncogene Proteins c-myc/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism
6.
Future Med Chem ; 11(9): 947-958, 2019 05.
Article in English | MEDLINE | ID: mdl-31141411

ABSTRACT

Aim: To analyze gene expression and copy number of five miRNAs (miR-1204, miR-1205, miR-1206, miR-1207 and miR-1208) localized in this chromosome region in gastric cancer (GC). Materials & methods: 65 paired neoplastic and non-neoplastic specimens collected from GC patients and 20 non-neoplastic gastric tissues from cancer-free individuals were included in this study. The expression levels of the five miRNAs were accessed by real time qPCR and were correlated. Results: MiR-1207-3p, miR-1205, miR-1207-5p and miR-1208 were upregulated in approximately 50% of GC tumors in relation to those of adjacent non-neoplastic tissues. MiR-1205 expression was associated with gain of gene copies and was upregulated in adjacent non-neoplastic samples relative to external controls. Conclusion: The coexpression of the 8q24 miRNAs indicated the role of miR-1205 in the initiation of gastric cancer development.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Stomach Neoplasms/genetics , Adult , Cell Line, Tumor , DNA Copy Number Variations , Female , Gene Expression Profiling , Humans , Male , Middle Aged , Up-Regulation
7.
Epigenomics ; 11(3): 349-362, 2019 02.
Article in English | MEDLINE | ID: mdl-30672330

ABSTRACT

Histone modifications regulate the structural status of chromatin and thereby influence the transcriptional status of genes. These processes are controlled by the recruitment of different enzymes to a specific genomic site. Furthermore, obtaining an understanding of these mechanisms could help delineate alternative treatment and preventive strategies for cancer. For example, in gastric cancer, cholecalciferol, curcumin, resveratrol, quercetin, garcinol and sodium butyrate are natural regulators of acetylation and deacetylation enzyme activity that exert chemopreventive and anticancer effects. Here, we review the recent findings on histone acetylation in gastric cancer and discuss the effects of nutrients and bioactive compounds on histone acetylation and their potential role in the prevention and treatment of this type of cancer.


Subject(s)
Disease Susceptibility , Histones/metabolism , Stomach Neoplasms/etiology , Stomach Neoplasms/metabolism , Acetylation/drug effects , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Dietetics , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Humans , Protein Processing, Post-Translational , Stomach Neoplasms/pathology , Stomach Neoplasms/therapy
8.
Exp Mol Pathol ; 103(1): 101-111, 2017 08.
Article in English | MEDLINE | ID: mdl-28736214

ABSTRACT

Cancer is a multifactorial disease that involves many molecular alterations. Gastric cancer (GC) is the third leading cause of cancer death worldwide. GC is a highly heterogeneous disease with different molecular and genetics features. Therefore, this review focuses on an overview of the genetic aspects of gastric cancer by highlighting the important impact and role of deletions and/or duplications of chromosomal segments, genomic variants, H. pylori infection and interleukin variants, as found in gene expression and newly proposed molecular classification studies. The challenge is to better understand the mechanisms and different pathways that lead to the development and progression of GC.


Subject(s)
Polymorphism, Genetic , Stomach Neoplasms/diagnosis , Stomach Neoplasms/genetics , DNA Copy Number Variations , Genome-Wide Association Study , Helicobacter Infections/complications , Helicobacter Infections/diagnosis , Helicobacter pylori , Humans , Interleukins/genetics , Interleukins/metabolism , Meta-Analysis as Topic , Signal Transduction , Stomach Neoplasms/etiology
10.
J Cell Biochem ; 118(4): 869-877, 2017 04.
Article in English | MEDLINE | ID: mdl-27748538

ABSTRACT

Different from genetic alterations, the reversible nature of epigenetic modifications provides an interesting opportunity for the development of clinically relevant therapeutics in different tumors. In this study, we aimed to screen and validate candidate genes regulated by the epigenetic marker associated with transcriptional activation, histone acetylation, in gastric cancer (GC). We first compared gene expression profile of trichostatin A-treated and control GC cell lines using microarray assay. Among the 55 differentially expressed genes identified in this analysis, we chose the up-regulated genes BMP8B and BAMBI for further analyses, that included mRNA and histone acetylation quantification in paired GC and nontumor tissue samples. BMP8B expression was reduced in GC compared to nontumor samples (P < 0.01). In addition, reduced BMP8B expression was associated with poorly differentiated GC (P = 0.02). No differences or histopathological associations were identified concerning BAMBI expression. Furthermore, acetylated H3K9 and H4K16 levels at BMP8B were increased in GC compared to nontumors (P < 0.05). However, reduced levels of acetylated H3K9 and H4K16 were associated with poorly differentiated GC (P < 0.05). Reduced levels of acetylated H3K9 was also associated with diffuse-type histological GC (P < 0.05). Notably, reduced BMP8B mRNA and acetylated H4K16 levels were positively correlated in poorly differentiated GC (P < 0.05). Our study demonstrated that BMP8B seems to be a tumor suppressor gene regulated by H4K16 acetylation in poorly differentiated GC. Therefore, BMP8B may be a potential target for TSA-based therapies in this GC sample subset. J. Cell. Biochem. 118: 869-877, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Bone Morphogenetic Proteins/genetics , Genes, Tumor Suppressor , Histones/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Acetylation , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adult , Cell Differentiation/genetics , Cell Line, Tumor , Epigenesis, Genetic , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydroxamic Acids/pharmacology , Male , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Stomach Neoplasms/drug therapy
11.
Clin Exp Med ; 17(1): 121-129, 2017 Feb.
Article in English | MEDLINE | ID: mdl-26567008

ABSTRACT

CDKN1A is a tumor suppressor gene involved in gastric carcinogenesis and is a potential target for histone deacetylase inhibitor-based therapies. Upregulation of CDKN1A is generally observed in several cell lines after histone deacetylase inhibitor treatment; however, little is known about the histone acetylation status associated with this gene in clinical samples, including gastric tumor tissue samples. Therefore, our goal was to quantify the H3K9 and H4K16 acetylation levels associated with three CDKN1A regions in 21 matched pairs of gastric adenocarcinoma and corresponding adjacent non-tumor samples by chromatin immunoprecipitation and to correlate these data with the gene expression. Our results demonstrated that the -402, -20, and +182 CDKN1A regions showed a significantly increased acetylation level in at least one of the histones evaluated (p < 0.05, for all comparisons), and these levels were positively correlated in gastric tumors. However, an inverse correlation was detected between both H3K9 and H4K16 acetylation at the -402 CDKN1A region and mRNA levels in gastric tumors (r = -0.51, p = 0.02; r = -0.60, p < 0.01, respectively). Furthermore, increased H4K16 acetylation at the -20 CDKN1A region was associated with gastric tumors of patients without lymph node metastasis (p = 0.04). These results highlight the complexity of these processes in gastric adenocarcinoma and contribute to a better understanding of CDKN1A regulation in carcinogenesis.


Subject(s)
Adenocarcinoma/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Gene Expression Regulation, Neoplastic , Histones/genetics , Stomach Neoplasms/genetics , Acetylation , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adult , Chromatin Immunoprecipitation , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Epigenesis, Genetic , Female , Histones/metabolism , Humans , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
12.
World J Gastroenterol ; 22(43): 9506-9514, 2016 Nov 21.
Article in English | MEDLINE | ID: mdl-27920471

ABSTRACT

AIM: To identify common copy number alterations on gastric cancer cell lines. METHODS: Four gastric cancer cell lines (ACP02, ACP03, AGP01 and PG100) underwent chromosomal comparative genome hybridization and array comparative genome hybridization. We also confirmed the results by fluorescence in situ hybridization analysis using the bacterial artificial chromosome clone and quantitative real time PCR analysis. RESULTS: The amplification of 9p13.3 was detected in all cell lines by both methodologies. An increase in the copy number of 9p13.3 was also confirmed by fluorescence in situ hybridization analysis. Moreover, the interleukin 11 receptor alpha (IL11RA) and maternal embryonic leucine zipper kinase (MELK) genes, which are present in the 9p13.3 amplicon, revealed gains of the MELK gene in all the cell lines studied. Additionally, a gain in the copy number of IL11RA and MELK was observed in 19.1% (13/68) and 55.9% (38/68) of primary gastric adenocarcinoma samples, respectively. CONCLUSION: The characterization of a small gain region at 9p13.3 in gastric cancer cell lines and primary gastric adenocarcinoma samples has revealed MELK as a candidate target gene that is possibly related to the development of gastric cancer.


Subject(s)
Adenocarcinoma/genetics , Gene Amplification , Gene Expression Profiling/methods , Interleukin-11 Receptor alpha Subunit/genetics , Protein Serine-Threonine Kinases/genetics , Stomach Neoplasms/genetics , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Adult , Aged , Cell Line, Tumor , Comparative Genomic Hybridization , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology
13.
World J Gastroenterol ; 22(35): 7951-62, 2016 Sep 21.
Article in English | MEDLINE | ID: mdl-27672290

ABSTRACT

Alterations in epigenetic control of gene expression play an important role in many diseases, including gastric cancer. Many studies have identified a large number of upregulated oncogenic miRNAs and downregulated tumour-suppressor miRNAs in this type of cancer. In this review, we provide an overview of the role of miRNAs, pointing to their potential to be useful as diagnostic and/or prognostic biomarkers in gastric cancer. Moreover, we discuss the influence of polymorphisms and epigenetic modifications on miRNA activity.


Subject(s)
Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , MicroRNAs/therapeutic use , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Epigenesis, Genetic , Humans , Polymorphism, Genetic , Polymorphism, Single Nucleotide , Prognosis , RNA, Messenger/metabolism
14.
Tumour Biol ; 37(8): 9991-10010, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27126070

ABSTRACT

Gastric cancer is a complex, heterogeneous, and multistep disease. Over the past decades, several studies have aimed to determine the molecular factors that lead to gastric cancer development and progression. After completing the human genome sequencing, proteomic technologies have presented rapid progress. Differently from the relative static state of genome, the cell proteome is dynamic and changes in pathologic conditions. Proteomic approaches have been used to determine proteome profiles and identify differentially expressed proteins between groups of samples, such as neoplastic and nonneoplastic samples or between samples of different cancer subtypes or stages. Therefore, proteomic technologies are a useful tool toward improving the knowledge of gastric cancer molecular pathogenesis and the understanding of tumor heterogeneity. This review aimed to summarize the proteins or protein families that are frequently identified by using high-throughput screening methods and which thus may have a key role in gastric carcinogenesis. The increased knowledge of gastric carcinogenesis will clearly help in the development of new anticancer treatments. Although the studies are still in their infancy, the reviewed proteins may be useful for gastric cancer diagnosis, prognosis, and patient management.


Subject(s)
Cell Transformation, Neoplastic , Neoplasm Proteins/analysis , Proteomics/methods , Stomach Neoplasms/etiology , High-Throughput Screening Assays , Humans , Neoplasm Proteins/classification , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Protein Processing, Post-Translational , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Two-Dimensional Difference Gel Electrophoresis
15.
J Gastrointest Cancer ; 47(1): 20-6, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26585943

ABSTRACT

PURPOSE: Gastritis caused by infection with Helicobacter pylori is characterized by chronic inflammation and damage in gastric tissue, which is a main risk factor for gastric cancer. Associated with H. pylori, the TP53 gene tumor suppressor and the cell adhesion glycoprotein epithelial cadherin develop a relevant role in the integrity and carcinogenesis of the epithelium. We aimed to detection of H. pylori and its main virulence markers and measured the messenger RNA (mRNA) expression levels of E-cadherin and TP53 genes. METHODS: The detection of H. pylori and its virulence markers, as well as the mRNA expression levels of E-cadherin and TP53 genes, were obtained for 161 samples of gastric biopsies including 37 with normal gastric tissue, 70 with gastritis, 24 from neoplastic tissue, and 27 from adjacent non-neoplastic by means of a quantitative real-time polymerase chain reaction. RESULTS: The mRNA expression levels of E-cadherin and TP53 were found to be decreased in patients with gastritis, independently of H. pylori infection. In samples from gastric patients, the neoplastic tissue showed an accentuated decrease of expression; on the other hand, the expression of E-cadherin was normal in adjacent non-neoplastic. CONCLUSIONS: No evidence was found of the involvement of the cagA and vacA genes in the decreased expression of E-cadherin and TP53. The process of carcinogenesis is complex, and the decrease of the E-cadherin gene expression and TP53 gene expression appears to contribute significantly.


Subject(s)
Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Biomarkers/analysis , Cadherins/genetics , Helicobacter Infections/genetics , Stomach Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Virulence/genetics , Adult , Case-Control Studies , DNA, Viral/genetics , Down-Regulation , Female , Follow-Up Studies , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Gastritis/genetics , Gastritis/pathology , Gastritis/virology , Helicobacter Infections/metabolism , Helicobacter Infections/pathology , Helicobacter Infections/virology , Helicobacter pylori , Humans , Male , Neoplasm Staging , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/pathology , Stomach Neoplasms/virology , Survival Rate
16.
Article in English | MEDLINE | ID: mdl-26719751

ABSTRACT

BACKGROUND: Tumor necrosis factor plays a critical role in the pathogenesis of gastric diseases such as gastric cancer, and an abnormal inflammatory response has frequently been observed in dyspeptic patients. Helicobacter pylori infection can induce a gastric mucosal inflammatory response that may be influenced by -308 (G > A) polymorphisms and gene expression of the TNF-α gene. METHODS: One hundred and thirty-four gastric biopsy samples were collected from patients of both genders (61♂ and 73♀, mean age 40.3 ± 24.2 years) with gastric symptoms. The -308 (G > A) polymorphism of TNF-α was characterized using polymerase chain reaction (PCR) and restriction fragment length polymorphism (RFLP). The expression level was measured using real-time PCR, and relative quantification (RQ) was calculated using the comparative CT method (2(-ΔΔCT)). RESULTS: The analysis revealed an increase in TNF-α gene expression in patients with gastritis; on the other hand, no statistical differences were observed in patients with gastric cancer. In addition, no association was found among -308 polymorphism genotypes, virulence markers, or TNF-α gene expression. CONCLUSIONS: Helicobacter pylori induces a large increase in TNF-α expression in patients with gastritis, regardless of tissue inflammation, but after the tissue becomes neoplastic, the presence of bacteria did not influence expression. These results suggest that the TNF-α pathway may play an important role in the progression from gastritis to gastric cancer.

17.
J. venom. anim. toxins incl. trop. dis ; 21: 1-7, 31/03/2015. tab
Article in English | LILACS, VETINDEX | ID: biblio-1484647

ABSTRACT

Background Tumor necrosis factor plays a critical role in the pathogenesis of gastric diseases such as gastric cancer, and an abnormal inflammatory response has frequently been observed in dyspeptic patients. Helicobacter pylori infection can induce a gastric mucosal inflammatory response that may be influenced by -308 (G > A) polymorphisms and gene expression of theTNF- gene. Methods One hundred and thirty-four gastric biopsy samples were collected from patients of both genders (61 and 73, mean age 40.3 ± 24.2 years) with gastric symptoms. The -308 (G > A) polymorphism of TNF- was characterized using polymerase chain reaction (PCR) and restriction fragment length polymorphism (RFLP). The expression level was measured using real-time PCR, and relative quantification (RQ) was calculated using the comparative CT method (2-CT). Results The analysis revealed an increase in TNF- gene expression in patients with gastritis; on the other hand, no statistical differences were observed in patients with gastric cancer. In addition, no association was found among -308 polymorphism genotypes, virulence markers, or TNF- gene expression. Conclusions Helicobacter pylori induces a large increase in TNF- expression in patients with gastritis, regardless of tissue inflammation, but after the tissue becomes neoplastic, the presence of bacteria did not influence expression. These results suggest that the TNF- pathway may play an important role in the progression from gastritis to gastric cancer.


Subject(s)
Animals , Tumor Necrosis Factor-alpha , Helicobacter pylori/genetics , Stomach Neoplasms , Polymorphism, Genetic
18.
Article in English | LILACS, VETINDEX | ID: biblio-954749

ABSTRACT

Background Tumor necrosis factor plays a critical role in the pathogenesis of gastric diseases such as gastric cancer, and an abnormal inflammatory response has frequently been observed in dyspeptic patients. Helicobacter pylori infection can induce a gastric mucosal inflammatory response that may be influenced by -308 (G > A) polymorphisms and gene expression of theTNF-α gene. Methods One hundred and thirty-four gastric biopsy samples were collected from patients of both genders (61♂ and 73♀, mean age 40.3 ± 24.2 years) with gastric symptoms. The -308 (G > A) polymorphism of TNF-α; was characterized using polymerase chain reaction (PCR) and restriction fragment length polymorphism (RFLP). The expression level was measured using real-time PCR, and relative quantification (RQ) was calculated using the comparative CT method (2-ΔΔCT). Results The analysis revealed an increase in TNF-α gene expression in patients with gastritis; on the other hand, no statistical differences were observed in patients with gastric cancer. In addition, no association was found among -308 polymorphism genotypes, virulence markers, or TNF-α gene expression. Conclusions Helicobacter pylori induces a large increase in TNF-α expression in patients with gastritis, regardless of tissue inflammation, but after the tissue becomes neoplastic, the presence of bacteria did not influence expression. These results suggest that the TNF-α; pathway may play an important role in the progression from gastritis to gastric cancer(AU)


Subject(s)
Stomach Neoplasms , Biomarkers , Gene Expression , Helicobacter pylori , Biopsy
19.
PLoS One ; 9(5): e98583, 2014.
Article in English | MEDLINE | ID: mdl-24879411

ABSTRACT

PHB is a reported oncogene and tumor suppressor in gastric cancer. Here, we evaluated whether the PHB copy number and the rs6917 polymorphism affect its expression in gastric cancer. Down-regulation and up-regulation of PHB were observed in the evaluated tumors. Reduced expression was associated with tumor dedifferentiation and cancer initiation. The T allele of the rs6917 polymorphism was associated with reduced PHB mRNA levels. Moreover, the up-regulation of PHB appeared to be regulated by the gain of additional gene copies. Thus, PHB copy number variation and differential expression of the rs6917 polymorphism may play a role in PHB transcriptional regulation.


Subject(s)
3' Untranslated Regions/genetics , DNA Copy Number Variations/genetics , Gene Expression Regulation, Neoplastic/genetics , Polymorphism, Single Nucleotide/genetics , Repressor Proteins/genetics , Stomach Neoplasms/genetics , Alleles , Cell Differentiation/genetics , Down-Regulation/genetics , Female , Genetic Predisposition to Disease/genetics , Humans , Male , Middle Aged , Prohibitins , RNA, Messenger , Up-Regulation/genetics
20.
Tumour Biol ; 35(7): 6373-81, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24668547

ABSTRACT

Gastric cancer is still the second leading cause of cancer-related death worldwide, even though its incidence and mortality have declined over the recent few decades. Epigenetic control using histone deacetylase inhibitors, such as trichostatin A (TSA), is a promising cancer therapy. This study aimed to assess the messenger RNA (mRNA) levels of three histone deacetylases (HDAC1, HDAC2, and HDAC3), two histone acetyltransferases (GCN5 and PCAF), and two possible targets of these histone modifiers (MYC and CDKN1A) in 50 matched pairs of gastric tumors and corresponding adjacent nontumors samples from patients with gastric adenocarcinoma, as well as their correlations and their possible associations with clinicopathological features. Additionally, we evaluated whether these genes are sensitive to TSA in gastric cancer cell lines. Our results demonstrated downregulation of HDAC1, PCAF, and CDKN1A in gastric tumors compared with adjacent nontumors (P < 0.05). On the other hand, upregulation of HDAC2, GCN5, and MYC was observed in gastric tumors compared with adjacent nontumors (P < 0.05). The mRNA level of MYC was correlated to HDAC3 and GCN5 (P < 0.05), whereas CDKN1A was correlated to HDAC1 and GCN5 (P < 0.05 and P < 0.01, respectively). In addition, the reduced expression of PCAF was associated with intestinal-type gastric cancer (P = 0.03) and TNM stages I/II (P = 0.01). The increased expression of GCN5 was associated with advanced stage gastric cancer (P = 0.02) and tumor invasion (P = 0.03). The gastric cell lines treated with TSA showed different patterns of histone deacetylase and acetyltransferase mRNA expression, downregulation of MYC, and upregulation of CDKN1A. Our findings suggest that alteration of histone modifier genes play an important role in gastric carcinogenesis, contributing to MYC and CDKN1A deregulation. In addition, all genes studied here are modulated by TSA, although this modulation appears to be dependent of the genetic background of the cell line.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors/administration & dosage , Hydroxamic Acids/administration & dosage , RNA, Messenger/biosynthesis , Stomach Neoplasms/enzymology , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/genetics , Gene Expression Regulation, Neoplastic/genetics , Genes, myc/genetics , Histone Deacetylase 1/biosynthesis , Histone Deacetylase 1/genetics , Histone Deacetylase 2/biosynthesis , Histone Deacetylase 2/genetics , Histone Deacetylases/biosynthesis , Histone Deacetylases/genetics , Humans , RNA, Messenger/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Transcriptional Activation/drug effects , p300-CBP Transcription Factors/biosynthesis , p300-CBP Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...