Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
2.
World J Gastroenterol ; 26(3): 307-323, 2020 Jan 21.
Article in English | MEDLINE | ID: mdl-31988591

ABSTRACT

BACKGROUND: Single nucleotide polymorphisms (SNPs) are universally present in nucleotide excision repair (NER) pathway genes, which could make impacts on colorectal carcinogenesis and prognosis. AIM: To explore the association of all tagSNPs in NER pathway genes with colorectal cancer (CRC) risk and prognosis in a northern Chinese population by a two-stage case-control design composed of a discovery and validation stage. METHODS: Genotyping for NER SNPs was performed using kompetitive allele specific PCR. In the discovery stage, 39 tagSNPs in eight genes were genotyped in 368 subjects, including 184 CRC cases and 184 individual-matched controls. In the validation stage, 13 SNPs in six genes were analyzed in a total of 1712 subjects, including 854 CRC cases and 858 CRC-free controls. RESULTS: Two SNPs (XPA rs10817938 and XPC rs2607775) were associated with an increased CRC risk in overall and stratification analyses. Significant cumulative and interaction effects were also demonstrated in the studied SNPs on CRC risk. Another two SNPs (ERCC2 rs1052555 and ERCC5 rs2228959) were newly found to be associated with a poor overall survival of CRC patients. CONCLUSION: Our findings suggest novel SNPs in NER pathway genes that can be predictive for CRC risk and prognosis in a large-scale Chinese population. The present study has referential values for the identification of all-round NER-based genetic biomarkers in predicting the susceptibility and clinical outcome of CRC.


Subject(s)
Colorectal Neoplasms/genetics , DNA Repair/genetics , Genetic Predisposition to Disease/genetics , Signal Transduction/genetics , Asian People/genetics , Carcinogenesis/genetics , Case-Control Studies , Colorectal Neoplasms/mortality , DNA-Binding Proteins/genetics , Endonucleases/genetics , Female , Genetic Markers/genetics , Genotype , Humans , Male , Middle Aged , Nuclear Proteins/genetics , Polymorphism, Single Nucleotide , Prognosis , Risk Factors , Transcription Factors/genetics , Xeroderma Pigmentosum Group D Protein/genetics
3.
J Zhejiang Univ Sci B ; 21(1): 87-92, 2020.
Article in English | MEDLINE | ID: mdl-31898445

ABSTRACT

Non-obstructive azoospermia (NOA), which is defined as the absence of spermatozoa in the ejaculate secondary to impaired spermatogenesis within the testis, may be caused by a variety of etiologies, including varicocele-induced testicular damage, cryptorchidism, prior testicular torsion, post-pubertal mumps orchitis, gonadotoxic effects from medications, genetic abnormalities, chemotherapy/radiation, and other unknown causes currently classified as idiopathic (Cocuzza et al., 2013). The microdissection testicular sperm extraction (micro-TESE) technique involves a meticulous microsurgical exploration of the testicular parenchyma to identify and selectively extract larger seminiferous tubules that carry a higher probability of complete spermatogenesis (Schlegel, 1999). The Cornell group evaluated the efficacy of micro-TESE in 152 NOA patients with an associated history of cryptorchidism. In their series, spermatozoa were successfully retrieved in 116/181 attempts (64%), and the resulting pregnancy rate was 50% with a delivery rate of 38% (Dabaja and Schlegel, 2013). Franco et al. (2016) described a stepwise micro-TESE approach in NOA patients, which was considered to reduce the cost, time, and effort associated with the surgery. Alrabeeah et al. (2016) further reported that a mini-incision micro-TESE, carried through a 1-cm equatorial testicular incision, can be useful for micro-TESE candidates, particularly in patients with cryptozoospermia. We conducted a retrospective study of 20 consecutive NOA patients with a history of orchidopexy from May 2015 to March 2017.


Subject(s)
Azoospermia/surgery , Microdissection/methods , Orchiopexy , Sperm Retrieval , Adult , Humans , Male , Middle Aged , Retrospective Studies
4.
Front Microbiol ; 10: 1261, 2019.
Article in English | MEDLINE | ID: mdl-31231345

ABSTRACT

The link between microbiota and gastric cancer (GC) has attracted widespread attention. However, the phylogenetic profiles of niche-specific microbiota in the tumor microenvironment is still unclear. Here, mucosa-associated microorganisms from 62 pairs of matched GC tissues and adjacent non-cancerous tissues were characterized by 16S rRNA gene sequencing. Functional profiles of the microbiota were predicted using PICRUSt, and a co-occurrence network was constructed to analyze interactions among gastric microbiota. Results demonstrated that mucosa-associated microbiota from cancerous and non-cancerous tissues established micro-ecological systems that differed in composition, structure, interaction networks, and functions. Microbial richness and diversity were increased in cancerous tissues, with the co-occurrence network exhibiting greater complexity compared with that in non-cancerous tissue. The bacterial taxa enriched in the cancer samples were predominantly represented by oral bacteria (such as Peptostreptococcus, Streptococcus, and Fusobacterium), while lactic acid-producing bacteria (such as Lactococcus lactis and Lactobacillus brevis) were more abundant in adjacent non-tumor tissues. Colonization by Helicobacter pylori, which is a GC risk factor, also impacted the structure of the microbiota. Enhanced bacterial purine metabolism, carbohydrate metabolism and denitrification functions were predicted in the cancer associated microbial communities, which was consistent with the increased energy metabolism and concentration of nitrogen-containing compounds in the tumor microenvironment. Furthermore, the microbial co-occurrence networks in cancerous and non-cancerous tissues of GC patients were described for the first time. And differential taxa and functions between the two groups were identified. Changes in the abundance of certain bacterial taxa, especially oral microbiota, may play a role in the maintenance of the local microenvironment, which is associated with the development or progression of GC.

5.
BMC Cardiovasc Disord ; 18(1): 219, 2018 11 29.
Article in English | MEDLINE | ID: mdl-30497388

ABSTRACT

BACKGROUND: Matrix metalloproteinase 9 (MMP9) and Toll-like receptor 4 (TLR4) play important roles in aortic pathophysiology. However, there is lacking research on serum TLR4 levels in acute aortic dissection (AAD) patients, and the performance of serum MMP9 and TLR4 for the diagnosis of AAD is still unknown. This study aimed to evaluate the serum levels of MMP9 and TLR4 in AAD patients, identify their associations with circulating C-reactive protein (CRP) and D-dimer, which are well-known classical biomarkers of AAD, and further explore the potential diagnostic role of MMP9 and TLR4 in AAD. METHODS: Serum levels of MMP9 and TLR4 were measured by enzyme-linked immunosorbent assay (ELISA) in 88 AAD patients and 88 controls. The clinical test related information was collected from patients' electronic medical records. RESULTS: Serum MMP9 and TLR4 levels were significantly higher in AAD patients than those in healthy controls in the general and stratified comparisons. Either serum MMP9 or TLR4 was independently associated with the risk of AAD (all p < 0.001). There was a positive significant association between serum MMP9 and TLR4 (r = 0.518, p < 0.001). Both MMP9 and TLR4 levels were statistically correlated with circulating CRP, but not D-dimer. Based on receiver-operating characteristic (ROC) analysis, the area under the curves (AUCs) of MMP9 and TLR4 alone for the diagnosis of AAD were 0.810 and 0.799 with optimal cut-off points of 379.47 ng/ml and 7.83 ng/ml, respectively. Moreover, a combination of serum MMP9 and TLR4 increased the AUC to 0.89 with a sensitivity of 60.2% and specificity of 94.3%. CONCLUSIONS: Serum MMP9 and TLR4 could be potential biomarkers for identifying AAD, while the combined diagnostic value was higher in safely ruling out AAD.


Subject(s)
Aortic Aneurysm/blood , Aortic Dissection/blood , Matrix Metalloproteinase 9/blood , Toll-Like Receptor 4/blood , Acute Disease , Adult , Aged , Aortic Dissection/diagnosis , Aortic Dissection/enzymology , Aortic Aneurysm/diagnosis , Aortic Aneurysm/enzymology , Biomarkers/blood , C-Reactive Protein/analysis , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Female , Fibrin Fibrinogen Degradation Products/analysis , Humans , Male , Middle Aged , Predictive Value of Tests , Up-Regulation
6.
J Int Med Res ; 46(12): 5155-5166, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30205739

ABSTRACT

OBJECTIVE: To investigate the association between polymorphisms of the interleukin 10 ( IL10) gene and risk of gastric cancer (GC) and atrophic gastritis (AG). METHODS: This study enrolled patients with GC, patients with AG and healthy control subjects. Demographic data were collected and the IL10 gene -1082A/G, -819C/T and -592A/C polymorphisms were genotyped. An enzyme-linked immunosorbent assay was performed to detect Helicobacter pylori infection. RESULTS: The study enrolled 556 participants including 208 in the GC group, 116 in the AG group and 232 controls (CON group). In a recessive model of the IL10-819C/T polymorphism, a significantly decreased risk of GC was found compared with AG and non-cancer subjects, respectively (AG→GC: odds ratio OR 0.41; non-cancer→GC: OR 0.57). The CC genotype demonstrated a significantly increased risk of AG compared with CON. Similar significant results were detected in males and H. pylori-negative subgroups. The ACC haplotype was associated with a decreased risk of GC compared with AG. The ATC haplotype was associated with a decreased risk of AG compared with the CON group, but it was associated with an increased risk of GC compared with AG. CONCLUSION: The IL10 gene promoter -819C/T (rs1800871) polymorphism was associated with the risk of GC and AG in a Chinese population.


Subject(s)
Asian People/genetics , Gastritis, Atrophic/genetics , Interleukin-10/genetics , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Stomach Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Case-Control Studies , Female , Follow-Up Studies , Gastritis, Atrophic/epidemiology , Gastritis, Atrophic/pathology , Genetic Predisposition to Disease , Genotype , Humans , Male , Middle Aged , Prognosis , Risk Factors , Stomach Neoplasms/epidemiology , Stomach Neoplasms/pathology , Young Adult
7.
Clin Chim Acta ; 483: 69-75, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29649455

ABSTRACT

BACKGROUND: Immune inflammation appears to play a role in aortic aneurysm (AA) pathology. Toll-like receptor 4 (TLR4) has been proved to involve in immune inflammatory diseases. However, the relationship between serum TLR4 and AA is still unclear. METHODS: The study included 282 AA patients and 287 controls. The clinical test related information was collected in medical records. The levels of serum TLR4 were measured by enzyme-linked immunosorbent assay. RESULTS: Serum TLR4 levels were significantly higher in case groups and could be influenced by age, smoking, hypertension, diabetes and hyperlipidemia. Serum TLR4 was positively correlated with circulating CRP, Hcy, D-dimer, Fg and Cys-c in AA patients, even after adjusting the possible influencing factors. The optimal cut-off value of TLR4 was 13.64 ng/ml for discriminating AA, and the screening accuracy was higher for those who were males (sensitivity of 63.5% and specificity of 68.6%), smokers (sensitivity of 63.5% and specificity of 82.7%) and hyperlipidemia (sensitivity of 59.1% and specificity of 81.2%). Multiple logistic analyses showed that serum TLR4 was significantly correlated with AA risk (OR = 1.119, 95% CI = 1.077-1.162, p < 0.001) and subjects with high TLR4 levels (>13.64 ng/ml) were more likely to have AA (OR = 4.225, 95% CI = 2.477-7.206, p < 0.001). CONCLUSIONS: Serum TLR4 was closely related to AA and associated with some AA-related circulating markers. Serum TLR4 could be a novel and promising biomarker with important diagnostic and predictive value in the identification of aortic aneurysmal diseases.


Subject(s)
Aortic Aneurysm/blood , Toll-Like Receptor 4/blood , Biomarkers/blood , Female , Humans , Male , Middle Aged , Predictive Value of Tests
9.
Oncotarget ; 8(26): 43140-43152, 2017 Jun 27.
Article in English | MEDLINE | ID: mdl-28562347

ABSTRACT

Excision repair cross-complementing group 6 and 8 (ERCC6 and ERCC8) are two indispensable genes for the initiation of transcription-coupled nucleotide excision repair pathway. This study aimed to evaluate the interactions between single nucleotide polymorphisms of ERCC6 (rs1917799) and ERCC8 (rs158572 and rs158916) in gastric cancer and its precancerous diseases. Besides, protein level analysis were performed to compare ERCC6 and ERCC8 expression in different stages of gastric diseases, and to correlate SNPs jointly with gene expression. Sequenom MassARRAY platform method was used to detect polymorphisms of ERCC6 and ERCC8 in 1916 subjects. In situ ERCC6 and ERCC8 protein expression were detected by immunohistochemistry in 109 chronic superficial gastritis, 109 chronic atrophic gastritis and 109 gastric cancer cases. Our results demonstrated pairwise epistatic interactions between ERCC6 and ERCC8 SNPs that ERCC6 rs1917799-ERCC8 rs158572 combination was associated with decreased risk of chronic atrophic gastritis and increased risk of gastric cancer. ERCC6 rs1917799 also showed a significant interaction with ERCC8 rs158916 to reduce gastric cancer risk. The expressions of ERCC6, ERCC8 and ERCC6-ERCC8 combination have similarities that higher positivity was observed in chronic superficial gastritis compared with chronic atrophic gastritis and gastric cancer. As for the effects of ERCC6 and ERCC8 SNPs on the protein expression, single SNP had no correlation with corresponding gene expression, whereas the ERCC6 rs1917799-ERCC8 rs158572 pair had significant influence on ERCC6 and ERCC6-ERCC8 expression. In conclusion, ERCC6 rs1917799, ERCC8 rs158572 and rs158916 demonstrated pairwise epistatic interactions to associate with chronic atrophic gastritis and gastric cancer risk. The ERCC6 rs1917799-ERCC8 rs158572 pair significantly influence ERCC6 and ERCC6-ERCC8 expression.


Subject(s)
DNA Helicases/genetics , DNA Repair Enzymes/genetics , Gastritis, Atrophic/genetics , Poly-ADP-Ribose Binding Proteins/genetics , Stomach Neoplasms/genetics , Transcription Factors/genetics , Case-Control Studies , DNA Helicases/biosynthesis , DNA Repair Enzymes/biosynthesis , Female , Gastritis, Atrophic/enzymology , Gastritis, Atrophic/pathology , Genetic Predisposition to Disease , Humans , Immunohistochemistry , Male , Middle Aged , Poly-ADP-Ribose Binding Proteins/biosynthesis , Polymorphism, Single Nucleotide , Risk Factors , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology , Transcription Factors/biosynthesis
10.
PLoS One ; 9(2): e89203, 2014.
Article in English | MEDLINE | ID: mdl-24586594

ABSTRACT

BACKGROUND: The aim of this study was to investigate the interaction effects of pri-let-7a-1 rs10739971 with pepsinogen C (PGC) and excision repair cross complementing group 6 (ERCC6) gene polymorphisms and its association with the risks of gastric cancer and atrophic gastritis. We hoped to identify miRNA polymorphism or a combination of several polymorphisms that could serve as biomarkers for predicting the risk of gastric cancer and its precancerous diseases. METHODS: Sequenom MassARRAY platform method was used to detect polymorphisms of pri-let-7a-1 rs10739971 G → A, PGC rs4711690 C → G, PGC rs6458238 G → A, PGC rs9471643 G → C, and ERCC6 rs1917799 in 471 gastric cancer patients, 645 atrophic gastritis patients and 717 controls. RESULTS: An interaction effect of pri-let-7a-1 rs10739971 polymorphism with ERCC6 rs1917799 polymorphism was observed for the risk of gastric cancer (P interaction = 0.026); and interaction effects of pri-let-7a-1 rs10739971 polymorphism with PGC rs6458238 polymorphism (P interaction = 0.012) and PGC rs9471643 polymorphism (P interaction = 0.039) were observed for the risk of atrophic gastritis. CONCLUSION: The combination of pri-let-7a-1 rs10739971 polymorphism and ERCC6 and PGC polymorphisms could provide a greater prediction potential than a single polymorphism on its own. Large-scale studies and molecular mechanism research are needed to confirm our findings.


Subject(s)
DNA Helicases/genetics , DNA Repair Enzymes/genetics , Gastritis, Atrophic/genetics , Genetic Predisposition to Disease/genetics , MicroRNAs/genetics , Pepsinogen C/genetics , Polymorphism, Genetic/genetics , Stomach Neoplasms/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Female , Genotype , Humans , Male , Middle Aged , Poly-ADP-Ribose Binding Proteins , Precancerous Conditions/genetics , Risk , Young Adult
11.
Asian Pac J Cancer Prev ; 15(1): 215-20, 2014.
Article in English | MEDLINE | ID: mdl-24528029

ABSTRACT

OBJECTIVE: To determine the expression of E-cadherin, ß-catenin, and transcription factor 4 (TCF4) proteins in gastric diseases with relation to Helicobacter pylori infection. METHODS: A total of 309 patients including 60 with superficial gastritis (SG), 57 with atrophic gastritis (AG) and 192 with gastric cancer (GC), were enrolled. The expression of E-cadherin, ß-catenin, TCF4 proteins in the gastric mucosa was detected by immunohistochemistry and H. pylori infection by immunohistochemistry and PCR. RESULTS: The expression rates of E-cadherin were significantly higher in SG and AG than in GC (P<0.01), while those of ß-catenin in the nucleus were significantly lower in SG and AG than in GC (P<0.05). In GC cases, the expression rates of E-cadherin, ß-catenin and TCF4 were significantly higher in the intestinal type than in the diffuse type (P<0.05). In GC patients, the expression rate of E-cadherin was significantly higher in the presence of H. pylori than in the absence of infection (P=0.011). Moreover, the expression level of TCF4 and ß-catenin protein was significantly higher in the nucleus and cytoplasm in H. pylori positive than in H. pylori negative GC patients, especially in those with the intestinal type (all P < 0.05). CONCLUSION: The expression of E-cadherin and ß-catenin progressively decreases during the process of GC tumorigenesis, while overexpression of TCF4 occurs. H. pylori infection is associated with a significant increase in the expression of E-cadherin and ß-catenin in the cytoplasm and nucleus in GC patients, especially those with the intestinal type.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cadherins/metabolism , Helicobacter Infections/metabolism , Helicobacter pylori , Stomach Neoplasms/metabolism , Transcription Factors/metabolism , beta Catenin/metabolism , Adult , Aged , Carcinogenesis/metabolism , Cell Nucleus/metabolism , Cytoplasm/metabolism , Female , Gastric Mucosa/metabolism , Gastritis, Atrophic/complications , Gastritis, Atrophic/metabolism , Gastritis, Atrophic/pathology , Helicobacter Infections/complications , Humans , Male , Middle Aged , Stomach Neoplasms/complications , Stomach Neoplasms/pathology , Transcription Factor 4 , Wnt Signaling Pathway
12.
Pol J Microbiol ; 61(2): 147-50, 2012.
Article in English | MEDLINE | ID: mdl-23163215

ABSTRACT

There have been no reports on the relationship between virulence genes and gastric diseases based on the same bacterial colonization density. Our results indicated that Helicobacter pylori virulence genes were more relevant than colonization density as a pathogenic mechanism of gastric diseases, which helps elucidate the pathogenic mechanisms of bacteria and aids in the development of improved strategies for the treatment of gastric disease.


Subject(s)
Bacterial Proteins/genetics , Helicobacter Infections/microbiology , Helicobacter pylori/genetics , Stomach Diseases/microbiology , Virulence Factors/genetics , Adult , Aged , Aged, 80 and over , Female , Genotype , Helicobacter pylori/isolation & purification , Helicobacter pylori/pathogenicity , Humans , Male , Middle Aged , Virulence
13.
PLoS One ; 7(10): e47178, 2012.
Article in English | MEDLINE | ID: mdl-23077566

ABSTRACT

Glutathione S-transferase P1 (GSTP1) is a critical enzyme in the phase II detoxification pathway. One of the common functional polymorphisms of GSTP1 is A→G at nucleotide 313, which results in an amino acid substitution (Ile105Val) at the substrate binding site and reduced catalytic activity. We evaluated the interaction between GSTP1 Val allele and Helicobacter pylori infection, smoking and alcohol consumption, increasing the risk of gastric cancer among the Chinese population. Information on potential gastric cancer risk factors and blood specimens were collected from 618 incident gastric cancer cases and 1,830 non-cancer controls between March 2002 and December 2011 in Liaoning Province, China. GSTP1 Ile105Val was genotyped by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry and polymerase chain reaction-restriction fragment length polymorphism. Serum levels of anti-H. pylori IgG were measured by ELISA. Odds ratio (OR) and 95% confidence interval (CI) were calculated using multivariate logistic regression, adjusted by sex and age. The risk of gastric cancer was significantly elevated in patients with the GSTP1 Val/Val genotype (adjusted OR = 3.324; 95% CI = 1.790-6.172). An elevated risk of gastric cancer was observed in patients with H. pylori infection, smoking, or alcohol consumption, and together with the GSTP1 Ile/Val +Val/Val genotype (OR = 3.696; 95% CI = 2.475-5.521; OR = 1.638; 95% CI = 1.044-2.571; OR = 1.641; 95% CI = 0.983-2.739, respectively) (p<0.05). The GSTP1 Val allele shows an interaction with smoking, alcohol consumption, and especially H. pylori infection for increasing the risk of gastric cancer. These findings could demonstrate new pathophysiological pathways for the development of gastric cancer.


Subject(s)
Alcohol Drinking/adverse effects , Asian People/genetics , Glutathione S-Transferase pi/genetics , Helicobacter Infections/complications , Smoking/adverse effects , Stomach Neoplasms/genetics , Stomach Neoplasms/microbiology , China/epidemiology , Gastric Mucosa/metabolism , Helicobacter Infections/diagnosis , Helicobacter Infections/genetics , Helicobacter pylori/isolation & purification , Humans , Middle Aged , Polymorphism, Single Nucleotide , Risk Factors , Stomach/microbiology , Stomach Neoplasms/epidemiology , Valine/genetics
14.
FEMS Microbiol Lett ; 320(2): 103-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21535099

ABSTRACT

Helicobacter pylori, a microaerophilic Gram-negative bacterium, is known to cause chronic gastritis, peptic ulcer and gastric cancer. Genes that are present in certain isolates may determine strain-specific traits such as disease association and drug resistance. In order to understand the pathogenic mechanisms of gastric diseases, identify molecular markers of the diseases associated with H. pylori strains and provide clues for target treatment of H. pylori-related diseases, a subtracted DNA library was constructed from a gastric cancer-associated H. pylori strain and a superficial gastritis-associated H. pylori strain by suppression subtractive hybridization. The presence of gastric cancer-specific genes was identified by dot blot hybridization, DNA sequencing and PCR-based screening. Twelve gastric cancer-specific high-copy genes and nine low-copy genes were found in gastric cancer compared with the superficial gastritis strain. These genes were confirmed by PCR analysis of H. pylori isolates. Notably, peptidyl-prolyl cis-trans isomerase (PPIase) was detected positively in 11 out of 22 (50%) gastric cancer-associated H. pylori strains. In contrast, <24% of the H. pylori strains from superficial gastritis showed positive results. Given the potential role of PPIases in cell growth, apoptosis and oncogenic transformation, our results suggest that PPIase may represent a novel marker and potential therapeutic target for gastric cancer.


Subject(s)
Helicobacter pylori/enzymology , Helicobacter pylori/genetics , Peptidylprolyl Isomerase/genetics , Stomach Neoplasms/microbiology , Bacterial Proteins/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Chi-Square Distribution , DNA, Bacterial/genetics , Gastritis/microbiology , Gene Library , Genetic Markers/genetics , Humans , Immunoblotting , Nucleic Acid Hybridization/methods , Polymerase Chain Reaction , Sequence Analysis, DNA , Sequence Homology, Nucleic Acid
15.
J Gastroenterol Hepatol ; 26(6): 1039-46, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21303408

ABSTRACT

BACKGROUND AND AIM: Serum pepsinogen II (sPGII) is underutilized and considered an inconspicuous biomarker in clinical practice. We refocused on this neglected but novel biomarker and conducted the present study, aiming to elucidate the normal level of sPGII in healthy Chinese patients and to investigate the clinical utility of sPGII for gastric disease screening. METHODS: In 2008-2009, a total of 2022 participants from northern China were selected and enrolled in the study. sPGII and Helicobacter pylori (H. pylori)-immunoglobulin G were measured with ELISA. RESULTS: sPGII showed a normal value of 6.6 microg/L in a total of 466 patients with endoscopically- and histologically-normal stomachs. A small sex difference was observed: the average value of sPGII was 7 microg/L and 6 microg/L in males and females, respectively (P < 0.001). In the differentiation between healthy and diseased (endoscopically-diseased stomach or gastritis/atrophic gastritis in endoscopic biopsies) stomach mucosae, the best sPGII cut-off value was 8.25 microg/L (sensitivity 70.6%, specificity 70.8%). In screening the H. pylori seropositivity, the optimum cut-off sPGII value was 10.25 microg/L (sensitivity 71.6%, specificity 70.1%). CONCLUSIONS: We demonstrated that the mean values of sPGII in a healthy Chinese population are 7 microg/L and 6 microg/L for males and females, respectively. sPGII significantly increases in diseased and H. pylori-infected stomach, and the best sPGII cut-off value is 8.25 microg/L in the differentiation between patients with healthy and diseased stomach mucosae. Furthermore, Chinese patients with sPGII greater than 10.25 microg/L are at greater risk of various H. pylori-related gastropathies, and are therefore prior candidates for gastro-protection therapy.


Subject(s)
Clinical Enzyme Tests , Gastric Mucosa/enzymology , Gastritis/diagnosis , Pepsinogen C/blood , Stomach Neoplasms/diagnosis , Adult , Aged , Aged, 80 and over , Antibodies, Bacterial/blood , Biomarkers/blood , Case-Control Studies , China , Enzyme-Linked Immunosorbent Assay , Female , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Gastritis/blood , Gastritis/microbiology , Gastritis/pathology , Gastroscopy , Helicobacter pylori/immunology , Humans , Immunoglobulin G/blood , Male , Metaplasia , Middle Aged , Predictive Value of Tests , Prognosis , ROC Curve , Stomach Neoplasms/blood , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology
16.
APMIS ; 119(3): 187-97, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21284736

ABSTRACT

Epidemiologic studies have demonstrated that Helicobacter pylori infection is associated with increased risk for the development of gastric cancer. Animal studies have also shown that H. pylori infection leads to gastric carcinogenesis, especially intestinal phenotypes. However, no in vitro study has been carried out for cell transformation induced by H. pylori. The present study aimed to investigate whether 'chronic'H. pylori infection induces gastric epithelial cell transformation, and elucidate the underlying mechanisms of transformation induced by H. pylori. The immortalized 'normal' gastric epithelial cell line, GES-1, was co-cultured for 45 days with H. pylori strains B975 and L301. The cell proliferation was measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, Ki-67 antigen, and colony formation assay. The cell transformation was determined by observing cell morphology and measuring the expression of E-cadherin, ß-catenin, and transcription factor-4 (TCF-4) at both protein and mRNA levels. H. pylori induced morphologic changes in GES-1 cells and significantly increased the proliferation of GES-1 cells. Moreover, H. pylori up-regulated the expression of ß-catenin and TCF-4, and also induced the nuclear accumulation of ß-catenin. In addition, the diffusive gastric cancer-related gene, E-cadherin, was up-regulated at the protein level, but down-regulated at the mRNA level. H. pylori infection is capable of inducing GES-1 transformation to present with the characteristics of intestinal-type gastric cancers in vitro, likely through the ß-catenin/TCF-4 signaling pathway.


Subject(s)
Cell Transformation, Neoplastic , Helicobacter Infections/complications , Helicobacter pylori/pathogenicity , Stomach Neoplasms/etiology , Base Sequence , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Line, Transformed , Cell Proliferation , DNA Primers/genetics , Epithelial Cells/microbiology , Epithelial Cells/pathology , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Humans , In Vitro Techniques , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Transcription Factor 4 , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Stem Cell Assay , beta Catenin/genetics , beta Catenin/metabolism
17.
Asian Pac J Cancer Prev ; 12(10): 2637-41, 2011.
Article in English | MEDLINE | ID: mdl-22320966

ABSTRACT

The effect of high salt environments on biological characteristics of Helicobacter pylori is still unclear. In the present study, we therefore investigated biological characteristics of the bacterium exposed to high salt concentrations. H. pylori strain, L301, was cultured in media supplemented with different concentrations (3%, 15% and 30%) of sodium chloride (NaCl) under microaerophilic conditions for 48 h. Morphology was assessed by light microscopy, the ATP content was quantitated by single-tube fluorescent light-emission and the levels of CagA and UreB proteins were determined by Western blotting. After exposure to NaCl, H. pylori transformed from common spiral shape to U or even coccoid shapes. The ATP content was significantly higher in 30% NaCl group than in 15% and 3% NaCl group and the level of CagA protein increased with the salt concentration. The urease reaction was all strongly positive in H. pylori exposed to different salt concentrations. The level of 8-OHdG expression was significantly increased in GES-1 cells co-cultured with H. pylori exposed to high salt, compared with the level in uninfected cells. H. pylori survives under exposure to high salt concentrations up to 30%, exhibiting changes in mobility, morphology and CagA expression, associated with increased 8-OHdG in the gastric epithelial cells, indicative of DNA damage.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Deoxyguanosine/analogs & derivatives , Helicobacter pylori/drug effects , Helicobacter pylori/metabolism , Sodium Chloride/pharmacology , 8-Hydroxy-2'-Deoxyguanosine , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Cell Line , DNA Damage , Deoxyguanosine/analysis , Epithelial Cells/metabolism , Gastric Mucosa/metabolism , Helicobacter pylori/pathogenicity , Humans , Urease
18.
Int J Oncol ; 35(6): 1313-20, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19885554

ABSTRACT

Identifying the genetic variants that alter MUC1 protein expression may further our understanding of the risk for development of gastric cancer (GC). We used PCR-SSPs to identify the genotype of MUC1 A/G polymorphism at its 568 site of exon 2 and immunohistochemistry to detect MUC1 protein expression in GC patients and non-cancer subjects and analyzed the association between this polymorphism and MUC1 protein expression. We found that the frequency of AA genotype was significantly high in the GC patients and the risk for GC in AA genotype carriers increased 1.81-fold. Moreover, we found a significant underexpression of MUC1 protein in GC as compared to non-cancer subjects, which was negatively correlated to AA genotype of MUC1 (r=-0.1790, P=0.004). Furthermore, this study provides a possible mechanistic insight that the MUC1 A/G polymorphism at its 568 site disrupts the physiological functions of MUC1 which is important to the physiological protection of gastric mucosa. Thus we have provided evidence that may identify the MUC1 A/G polymorphism at 568 site, as a potential genetic factor which leads to an increase in susceptibility for GC through alteration of MUC1 gene and MUC1 expression in the population that carry the A allele.


Subject(s)
Carcinoma/genetics , Genetic Predisposition to Disease , Mucin-1/genetics , Stomach Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Base Sequence , Female , Genotype , Humans , Immunohistochemistry , Male , Middle Aged , Molecular Sequence Data , Polymerase Chain Reaction , Polymorphism, Single Nucleotide , Risk Factors , Tissue Array Analysis
19.
Ai Zheng ; 28(5): 487-92, 2009 May.
Article in Chinese | MEDLINE | ID: mdl-19624876

ABSTRACT

BACKGROUND AND OBJECTIVE: Human pepsinogen C (PGC) is an aspartic protease synthesized in gastric mucosa. PGC gene insertion/deletion polymorphism, which is located between exon 7 and 8, has been found to associate with gastric cancer (GC) susceptibility. This study was to investigate the relationship between PGC polymorphism with protein expression of PGC in gastric mucosa and serum. METHODS: PGC insertion/deletion polymorphism was evaluated by PCR, followed by direct DNA sequencing in 493 cases of GC, atrophic gastritis (AG), gastric erosion ulcer (GEU) and superficial gastritis (SG). PGC protein expression in gastric mucosa was measured by immunohistochemistry. The serum PGC level was determined by enzyme-linked immunosorbent assay (ELISA). RESULTS: In accordance with the following order SG-->GEU-->GA-->GC, the frequency of PGC homozygous allele 1 was gradually increased, which was higher in GC than in SG (P=0.018); while the protein expression of PGC in gastric mucosa was gradually decreased (P<0.01), along with a gradual decrease in the strong positive rate of PGC (P<0.05) except for SG vs. GEU. The serum level of PGC was significantly lower in SG than in GU(P=0.000) and GC(P=0.000). The frequency of PGC homozygous allele 1 was negatively correlated to PGC protein expression in gastric mucosa (r=-0.1085, P=0.023). From homozygous allele 1 to heterozygous allele 1, and to other genotypes, the PGC positive rate was gradually increased in gastric mucosa, with significant differences between homozygous allele 1 and other genotypes (P=0.009); while the strong-positive rate of PGC was gradually decreased only in SG group (P=0.047). CONCLUSION: PGC gene insertion/deletion polymorphism is negatively related to PGC protein expression in gastric mucosa, but is not related to the serum PGC level.


Subject(s)
Gastric Mucosa/metabolism , INDEL Mutation , Pepsinogen C/genetics , Polymorphism, Genetic , Stomach Neoplasms , Adult , Aged , Aged, 80 and over , Alleles , Female , Gastritis/blood , Gastritis/genetics , Gastritis/metabolism , Gastritis, Atrophic/blood , Gastritis, Atrophic/genetics , Gastritis, Atrophic/metabolism , Genetic Predisposition to Disease , Genotype , Humans , Male , Middle Aged , Pepsinogen C/blood , Pepsinogen C/metabolism , Sequence Analysis, DNA , Stomach Neoplasms/blood , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Ulcer/blood , Stomach Ulcer/genetics , Stomach Ulcer/metabolism
20.
Zhonghua Yi Xue Za Zhi ; 88(19): 1342-6, 2008 May 20.
Article in Chinese | MEDLINE | ID: mdl-18956706

ABSTRACT

OBJECTIVE: To investigate the feasibility of detecting Helicobacter pylori (Hp) directly from gastric mucosa and the relationship of HP genotypes to gastric diseases. METHODS: Specimens of gastric mucosa were collected by biopsy from 217 patients, 90 with superficial gastritis (GS), 70 with atrophic gastritis (GA), 28 with gastroesophageal erosions and ulcers (GEU), and 29 with gastric cancer (GC), to undergo pathological examination, culture of Hp, and DNA isolation from the gastric mucosa respectively. Routine phenol/chloroform method was used to isolate the DNA in the cultured Hp. PCR was conducted to detect the ureB, cagA, vacAs1, vacAm1, vacAm2, iceA1, iceA2, and baba2 genotypes in both the gastric mucosa-originated Hp-DNA and cultured Hp-DNA. RESULTS: The concordance rates of ureB, cagA, vacAs1, vacAm1, vacAm2, iceA1, iceA2, and baba2 genotypes between the gastric mucosa-originated Hp-DNA and cultured Hp-DNA were 74.23%, 73.39%, 93.69%, 62.16%, 78.16%, 89.13%, 88.37%, and 75% respectively (all P > 0.05). There were no significant differences in the distribution rates of ureB, cagA, vacAs1, vacAm1b, iceA1, iceA2, and baba2 genotypes among different gastric diseases (all P > 0.05). However, the distribution frequency of vacAs1m1 strain in the GA patients was 22.22%, significantly higher than that in the GA patients (5.71%, chi2 = 8.416, P = 0.004), and the distribution frequency of vacAs1m2 strains in the GA patients was 47.14%, significantly higher than that in GS patients (18.89%, chi2 = 13.336, P = 0.000). CONCLUSION: Rapid detection of Hp can be achieved by using DNA isolated directly from infected gastric mucosa. VacAs1 m1 is associated with GA and vacAs1m2 is associated with GA.


Subject(s)
Gastric Mucosa/microbiology , Gastritis/microbiology , Helicobacter Infections/microbiology , Helicobacter pylori/genetics , Adult , Aged , Aged, 80 and over , Bacterial Proteins/genetics , DNA, Bacterial/genetics , Female , Gastritis, Atrophic/microbiology , Gene Frequency , Genotype , Helicobacter pylori/isolation & purification , Humans , Male , Middle Aged , Polymerase Chain Reaction , Stomach Neoplasms/microbiology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...